Medical biochemistry [4th ed] 0867200308, 9780867200300, 0867100508, 9780867100501


268 93 90MB

English Pages 1012 Year 1992

Report DMCA / Copyright

DOWNLOAD PDF FILE

Table of contents :
Preface.pdf......Page 1
Acknowledgments.pdf......Page 2
Contributors.pdf......Page 4
1.pdf......Page 5
2.pdf......Page 21
3.pdf......Page 38
4.pdf......Page 54
5.pdf......Page 69
6.pdf......Page 87
7.pdf......Page 111
8.pdf......Page 122
9.pdf......Page 134
10.pdf......Page 153
11.pdf......Page 172
12.pdf......Page 195
13.pdf......Page 223
14.pdf......Page 245
15.pdf......Page 273
16.pdf......Page 304
17.pdf......Page 328
18.pdf......Page 361
19.pdf......Page 396
20.pdf......Page 423
21.pdf......Page 446
22.pdf......Page 478
23.pdf......Page 513
24.pdf......Page 536
25.pdf......Page 554
26.pdf......Page 583
27.pdf......Page 605
28.pdf......Page 635
29.pdf......Page 665
30.pdf......Page 688
31.pdf......Page 717
32.pdf......Page 736
33.pdf......Page 755
34.pdf......Page 766
35.pdf......Page 787
36.pdf......Page 833
37.pdf......Page 876
38.pdf......Page 904
39.pdf......Page 932
Appendix I.pdf......Page 942
Appendix II.pdf......Page 943
Appendix III.pdf......Page 944
Appendix IV.pdf......Page 945
Appendix IX.pdf......Page 947
Appendix V.pdf......Page 958
Appendix VI.pdf......Page 960
Appendix VII.pdf......Page 967
Appendix VIII.pdf......Page 972
Index.pdf......Page 975
Recommend Papers

Medical biochemistry [4th ed]
 0867200308, 9780867200300, 0867100508, 9780867100501

  • 0 0 0
  • Like this paper and download? You can publish your own PDF file online for free in a few minutes! Sign Up
File loading please wait...
Citation preview

PREFACE

In keeping with the previous editions, the primary purpose of Medical Biochemistry, Fourth Edition, is to present the fundamentals of biochemistry and related materials in a way that is useful to students pursuing medical and other health-related careers. The book was conceived and written with the hope that it would generate interest and enthusiasm among these students, particularly because biochemistry has a crucial role in human health and disease. Since it is assumed that most students in medicine and health-related fields eventually will apply biochemical principles to the art of healing, discussion of the factual information is integrated with frequent use of clinical examples and applications. A vast number of constituents of the human body m cells, enzymes, hormones, sugars, salts, vitamins, and so forthmvary in normal and abnormal states of human health. Understanding the metabolic and regulatory processes that underlie metabolism is essential to any practice of the healing arts and the relief of human suffering. I have tried to keep this idea foremost. Progress in biochemistry, molecular biology, cellular biology, endocrinology, and other disciplines has been so rapid and profound in the past 20 years that biochemistry texts obsolesce rapidly. To maximize the usefulness of this book, authors actively involved in research have written several chapters dealing with the most rapidly changing fields. The overall organization of topics is designed to lead the student logically through the biochemical organization of cells. Emphasis is placed on the structures and functions of the molecular components of cells and on metabolic controls. The text begins with a discussion of water, acids, bases, and buffers, amino acids, proteins, and thermodynamics (Chapters 1-5). This is fol-

lowed by a detailed discussion of important aspects of enzymology (Chapters 6-8). The broad subject of carbohydrate chemistry (Chapters 9-11, 13, 15, 16) is integrated with chapters that discuss gastrointestinal digestion (Chapter 12), oxidative phosphorylation (Chapter 14), and protein metabolism (Chapter 17). Three chapters on lipids (Chapters 18-20) are integrated with chapters coveting muscle systems (Chapter 21) and metabolic homeostasis (Chapter 22). The principles of molecular biology including nucleic acid chemistry and the regulation of gene expression and protein synthesis are presented in Chapters 23-26. These chapters are followed by ones on nucleotide, hemoglobin, and heme metabolism (Chapters 27-29). The endocrine system and its organs are discussed in Chapters 30-34. Molecular immunology is presented in Chapter 35, and the biochemistry of blood coagulation appears in Chapter 36. The last section discusses mineral and vitamin metabolism and electrolyte balance (Chapters 37-39). Nine appendices contain tables of nutritional values and clinical laboratory measurements that are important in diagnosis. The importance of human nutrition is emphasized th~ughout the text and has not been relegated to a single ~apter. Likewise, hereditary disorders are discussed throughout, along with other clinical examples that relate the relevant biochemistry to diagnosis and treatment. This book is not small, although conciseness consistent with clarity was a primary goal. Detailed discussions of experiments have, for the most part, been omitted, and discussion of more subtle points has been minimized. References provided at the end of each chapter will lead interested students deeper into particular topics and case studies. xxvii

ACKNOWLEDGMENTS

I am grateful to the authors for their contributions and for sharing their knowledge and insights. I also appreciate their cooperation during the lengthy writing and production process. I am indebted to Gordon Edlin, who participated in and supported all aspects of the publication of this book, to Alan E Goldstein, who reviewed many chapters and helped integrate basic science and clinical medicine, and Chung-eun Ha for spending tireless hours in the preparation of the manuscript during its many developmental stages. I am especially appreciative to the following individuals, who contributed to this book as reviewers of selected chapters, as writers of subsections, by offering constructive suggestions, or by providing encouragement and support during difficult times: R. A. Dubanoski, J. M. Hardman, K. H. Higa, S. A. A. Honda, K. Harohalli, D. K. Kikuta, W. Lee, A. A. Manoukian, H. E Mower, D. S. Park, C. E. Petersen, C. N. Rios, R. T. Sakaguchi, C. E. Sugiyama, and L. E. Takenaka. My thanks also to the following reviewers for their valuable chapter critiques: Dr. James W. Campbell, Department of Biochemistry and Cell Biology, Rice University; Dr. David Daleke, Department of Biochemistry, Indiana University; Dr. Beverly Delidow, School of Medicine, Marshall University; Dr. JoAnne Flynn, School of Medicine, University of Pittsburgh; Dr. Jennifer A. Pietenpol, School of Medicine, Vanderbilt University; Dr. Connie Prosser, Department of Laboratory Medicine, University of Alberta; Dr. Kevin D. Sarge, Department of Biochemistry, University of Kentucky;

Dr. Peter B. Smith, School of Medicine, Wake Forest University; Dr. Terry Stoming, Department of Biochemistry and Molecular Biology, Medical College of Georgia; Dr. Francis Vella, International Union of Biochemistry and Molecular Biology; and, Dr. D. Eric Walters, Department of Biochemistry and Molecular Biology, The Chicago Medical School. Dr. Craig M. Jackson acknowledges the images shown in Chapters 35 and 36 were created using RasMol 2.6, Molecular Graphics Visualization Tool by Roger Sayle, Bio-Molecular Structures Group, Glaxo Research & Development, Greenford, Middlesex, UK. RasMol can be obtained at http://www.umass.edu/microbio/rasmol/. The coordinates were obtained from the Protein Data Bank at Brookhaven National Laboratory, as described in F. C. Bernstein, T. E Koetzle, G. J. B. Williams, E. F. Meyer, Jr., M. D. Brice, J. R. Rodgers, O. Kennard, T. Shimanouchi, and M. Tasuxni, "The Protein Data Bank: A Computer-Based Archival File for Macromolecular Structures," J. Mol. Biol. 112, 535-542 (1977). Gene information has been obtained from GenBank, as described in C. Burks, M. Cassidy, M. J. Cinkosky, K. E. Cumella, E Gilna, J. E-D. Hayden, G. M. Keen, T. A. Kelley, M. Kelly, D. Kristofferson, and J. Ryals GenBank. Nucl. Acids Res. 19(Suppl), 2221-2225 (1991). For the coordinates of the molecules for which structures are shown, see http://www.rcsb.org/. The author thanks James G. White, M. D., Regents Professor, University of Minnesota, Minneapolis, Minnesota, for the photographs of platelets, and Meir Rigbi,

xxix

XXX

Professor Emeritus, Hebrew University of Jerusalem, for his many helpful comments on the manuscript. I also express my thanks to C. Agbayani, J. Gerber, and N. Mead for unfailing assistance in the preparation of

Acknowledgments

the manuscript. Finally, I greatly appreciate the assistance and advice provided to me by the editorial staff of Harcourt/Academic Press, in particular J. R. Hayhurst, N. M. Donaghy, and R. L. Orbegoso.

CONTRIBUTORS

Gordon Edlin, Ph.D. (Chapters 23, 24, 25, 26) Department of Biochemistry and Biophysics, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii

Richard J. Guillory, Ph.D. (Chapter 14) Department of Biochemistry and Biophysics, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii Craig M. Jackson, Ph.D. (Chapters 35, 36) Hemo SAGA Corporation, San Diego, California Conrad A. Hornick, Ph.D. (Chapters 18, 19, 20) Departments of Physiology and Pathology, Louisiana State University, New Orleans, Louisiana

David A. Lally, Ph.D. (Chapter 21) Department of Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii Walter K. Morishige, Ph.D. (Chapters 30, 31, 32, 33, 34) Department of Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii James R. Wilson, Ph.D. (Chapter 16) Department of Biology, La-Sierra University, Riverside, California

XXV

CHAPTER

1

Water, Acids, Bases, and Buffers

1.1 Properties of Water Acid and base concentrations in living systems are carefully regulated to maintain conditions compatible with normal life. Biochemical reactions involving acids and bases occur in the body water, whereas buffer systems protect the body from significant variations in the concentrations of acids and bases. This chapter introduces basic concepts of the properties of water, acids, bases, and buffers, and Chapter 39 presents a detailed discussion of both normal and pathological aspects of acid-base metabolism. Life cannot be sustained without water. Water constitutes 45-73% of total human body weight. It is distributed in intracellular (55%) and extracellular (45%)compartments and provides a continuous solvent phase between body compartments. As the biological solvent, water plays a major role in all aspects of metabolism: absorption, transport, digestion, and excretion of inorganic and organic substances as well as maintenance of body temperature. The unique properties of water are due to its structure.

Hydrogen Bonding Water (H20) is a hydride of oxygen in which the highly electronegative oxygen atom attracts the bonding electrons from two hydrogen atoms. This leads to polar H-O bonds in which the hydrogen atoms have a slightly positive

charge (~+) and the oxygen atom has a slightly negative charge (~)-, (Figure 1-1). "Water molecules have a relatively high dipole moment because of the angle (104.5 ~ of the H - O - H bond and the polarity of the bonds. Neighboring liquid water molecules interact with one another to form an extensive lattice-like structure similar to the structure of ice. The intermolecular bonding between water molecules arises from the attraction between the partial negative charge on the oxygen atom and the partial positive charge on the hydrogen atom of adjacent water molecules. This type of attraction involving a hydrogen atom is known as a hydrogen bond (Figure 1-2). Hydrogen bonds contain a hydrogen atom between two electronegative atoms (e.g., O and N). One is the formal hydrogen donor; the other is the hydrogen acceptor. The amount of energy required to break a hydrogen bond (bond energy) is estimated to be 2-5 kcal/mol (8.4-20.9 kJ/mol) in the gas phase. Covalent bonds have bond energies of 50-100 kcal/mol (209-418 kJ/mol). The cumulative effect of many hydrogen bonds is equivalent to the stabilizing effect of covalent bonds. In proteins, nucleic acids, and water, hydrogen bonds are essential to stabilize overall structure. In ice, each water molecule forms a hydrogen bond with four other water molecules, giving rise to a rigid tetrahedral arrangement (Figure 1-2). In the liquid state, water maintains a tetrahedrally coordinated structure over short ranges and for short time periods.

CHAPTER1 Water, Acids, Bases, and Buffers T A B L E 1-1

5 +

Physical Properties of Water*

m

25

m

F I G U R E 1-1 Structure of the water molecule.

Physical Properties Other properties of water uniquely suited to biological systems include melting point, boiling point, heat of vaporization (quantity of heat energy required to transform 1 g of liquid to vapor at the boiling point), heat of fusion (quantity of heat energy required to convert 1 g of solid to liquid at the melting point), specific heat (the amount of heat required to raise the temperature of 1 g of substance by I~ and surface tension (Table 1-1). All these values for water are much higher than those for other lowmolecular-weight substances because of the strong intermolecular hydrogen bonding of water. These properties contribute to maintenance of temperature and to dissipation of heat in living systems. Thus, water plays a major role in thermoregulation in living systems. The optimal body temperature is a balance between heat production and heat dissipation. Impaired thermoregulation causes either hypothermia or hyperthermia and has serious metabolic consequences; if uncorrected, impaired thermoregulation may lead to death (Chapter 39). Water freezes to form ice at 0~ but its maximum density is at 4~ Aquatic

H

J O"

H

= H

I ~O,~

H~

o/

I

H

__E~H

~

"',,,o,/

I

I

0

H ~H

F I G U R E 1-2 Tetrahedral hydrogen-bonded structure of water molecules in ice. The tetrahedral arrangement is due to the fact that each water molecule has four fractional charges: two negative charges due to the presence of a lone pair of electrons on the oxygen atom and two positive charges, one on each of the two hydrogen atoms. In the liquid phase this tetrahedral array occurs transiently.

Density (at 4~ Molecular weight Liquid range Melting point Boiling point Heat of fusion Heat of vaporization Dipole moment Dielectric constant (E) Solid/liquid density ratio

1.0 g/mL 18 0~176 0~ 100~ 80 cal/g 540 cal/g 1.86 Debye unit 78.4 0.92

*Some of these properties are measured at 1 atm pressure.

organisms survive cold winters because ice floats over and insulates liquid water from sub-zero-degree temperature. If ice were denser than liquid water--which is the case for most liquid-to-solid transformations~the solid form would sink and the entire amount of fluid would solidify rapidly in freezing weather. During hot weather, deep lakes and oceans remain cool because heat generated by sunlight can be dissipated by evaporation of surface water. Water is transported across cell membranes in one of two ways: 1. by simple diffusion through the phospholipid bilayer and 2. by the action of membrane-spanning transport proteins known as aquaporins. Thus, the concentration of water is in thermodynamic equilibrium across the cell membrane. In the renal collecting duct, water is reabsorbed through a specific aquaporin channel protein (aquaporin 2). This reabsorption of water is regulated by the antidiuretic hormone (also known as vasopressin). A defect or lack of functional aquaporin 2, vasopressin, or its receptor leads to enormous loss of water in the urine, causing the disease known as diabetes insipidus (Chapter 39). Water plays a significant role in enzyme functions, molecular assembly of macromolecules, and allosteric regulation of proteins. For example, the effect of protein solvation in allosteric regulation is implicated in the transition of deoxyhemoglobin to oxyhemoglobin. During this process about 60 extra water molecules bind to oxyhemoglobin (Chapter 28). Solutes, Micelles, a n d H y d r o p h o b i c Interactions Water is an excellent solvent for both ionic compounds (e.g., NaC1) and low-molecular-weight nonionic polar

SECTION1.1 Properties of Water

compounds (e.g., sugars and alcohols). Ionic compounds are soluble because water can overcome the electrostatic attraction between ions through solvation of the ions. Nonionic polar compounds are soluble because water molecules can form hydrogen bonds to polar groups (e.g., -OH). Amphipathic compounds, which contain both large nonpolar hydrocarbon chains (hydrophobic groups) and polar or ionic groups (hydrophilic groups) may associate with each other in submicroscopic aggregations called micelles. Micelles have hydrophilic (water-liking) groups on their exterior (bonding with solvent water), and hydrophobic (water-disliking) groups clustered in their interior (Figure 1-3). They occur in spherical, cylindrical, or ellipsoidal shapes. Micelle structures are stabilized by hydrogen bonding with water, by van der Waals attractive forces between hydrocarbon groups in the interior, and by energy of hydrophobic reactions. The last is the stabilization energy that would be lost if each hydrocarbon group were transferred from the hydrophobic medium to the polar aqueous solvent. As with hydrogen bonds, each hydrophobic interaction is very weak, but many such interactions result in formation of large, stable structures. A micelle may contain many hundreds of thousands of amphipathic molecules. The interior molecular organization of micelles has been likened to a "liquid hydrocarbon droplet." However, a recent model departs from this conventional concept and suggests that because of severe constraints in the space-filling requirements of hydrocarbon chains in the interior as well as because of micellar

"~~ ~ jHydrophobicchain Z J . ~ ./Hydrophilicgroup

3

geometry, the chain ends are not uniformly distributed throughout the micelle but tend to be clustered between the center of the micelle and the outer surface, implying that many of the hydrocarbon side chains are bent back upon themselves (Figure 1-3). In this model, there appears to be a progression from ordered (as in crystals) to disordered (as in liquids) structures proceeding from the center of the micelle to the periphery. Hydrophobic interaction plays a major role in maintaining the structure and function of cell membranes, the activity of proteins, the anesthetic action of nonpolar compounds such as chloroform and nitrous oxide, the absorption of digested fats, and the circulation of hydrophobic molecules in the interior of micelles in blood plasma.

Colligative Properties The colligative properties of a solvent depend upon the concentration of solute particles. These properties include freezing point depression, vapor pressure depression, osmotic pressure, and boiling point elevation. The freezing point of water is depressed by 1.86~ when 1 mol of nonvolatile solute, which neither dissociates nor associates in solution, is dissolved in 1 kg of water. The same concentration of solute elevates the boiling point by 0.543~ Osmotic pressure is a measure of the tendency of water molecules to migrate from a dilute to a concentrated solution through a semipermeable membrane. This migration of water molecules is termed osmosis. A solution containing 1 mol of solute particles in 1 kg of water is a 1-osmolal solution. When 1 mol of a solute (such as NaC1) that dissociates into two ions (Na + and C1-) is dissolved in 1 kg of water, the solution is 2-osmolal. Measurement of colligative properties is useful in estimating solute concentrations in biological fluids. For example, in blood plasma, the normal total concentration of solutes is remarkably constant (275-295 milliosmolal). Pathological conditions (e.g., dehydration, renal failure) involving abnormal plasma osmolality are discussed in Chapter 39.

Dissociation of Water and the pH Scale Water dissociates to yield a hydrogen ion (H +) and a hydroxyl ion (OH-). F I G U R E 1-3 A geometrical representation of a spherical micelle showing the hydrocarbon chains in the micelle. The hydrophilic groups are attracted to water, and the hydrophobic chains are within the micelle. The ends of the hydrocarbon chains are nonuniformly distributed, and many are located in the middle. The degree of disorder is much higher in the periphery than in the center of the micelle.

H20 ~ H + + OH-

(1.1)

The H + bonds to the oxygen atom of an undissociated H20 molecule to form a hydronium ion (H30+). H 2 0 if- H 2 0

~

H3 O + -+- O H -

CHAPTER1 Water, Acids, Bases, and Buffers

Thus, water functions both as an acid (donor of H + or proton) and as a base (acceptor of H + or proton). This description of an acid and a base follows from the Br6nstedLowry theory. According to the Lewis theory, acids are electron pair acceptors and bases are electron pair donors. The equilibrium constant, K, for the dissociation reaction in Equation (1.1) is K =

[H+][OH -]

(1.2)

[H20]

where the square brackets refer to the molar concentrations of the ions involved. K can be determined by measurement of the electrical conductivity of pure water, which has the value of 1.8 x 10 -16 M at 25~ indicative of a very small ion concentration, where M (molar) is the units of moles per liter. Therefore, the concentration of undissociated water is essentially unchanged by the dissociation reaction. Since 1 L of water weighs 1000 g and 1 mol of water weighs 18 g, the molar concentration of pure water is 55.5 M. Substitution for K and [H20] in Equation (1.2) yields

TABLE 1-2 The pH Scale [H+], M

pH

[OH-l, M

10.0 1.0 0.1 0.01(10 -2 ) 10-3 10-4 10-5 10 -6 10-7 < 10-8 10-9 10-10 10- l l 10-12 10-13 10-14 10-15

-1 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15

10-15 10-14 10-13 10-12 10-11 10-10 10-9 10-8 10-7 10-6 10-5 10-4 10-3 0.01 0.1 1 10

Acidic

Neutral

T 1

Basic

[H+][OH -] = (55.5 M) x (1.8 x 10 -16 M) [H+][OH -] = 1.0 • 10 -14 M 2 = Kw Kw is known as the ion product of water. In pure water, [H +] and [OH-] are equal, so that

discussions of acid-base problems in human biochemistry, it is often preferable to express H + concentration as nanomoles per liter (nmol/L).

[ O H - ] = [H + ] = 1.0• 10 - 7 M . pH is employed to express these ion concentrations in a convenient form, where the "p" of pH symbolizes "negative logarithm (to the base 10)" of the concentration in question. Thus, pH - - loglo[H +] -- log

[H + ]

Similarly, pOH - - loglo[OH-] -- log

[OH-I

Therefore, for water,

1.2 Buffers Buffers resist change in pH in solutions when acids or bases are added. They are either a mixture of a weak acid (HA) and its conjugate base (A-) or a mixture of a weak base (B) and its conjugate acid (HB+). EXAMPLE 1 Acetic acid (CH3COOH) and carbonic acid (H2CO3) are weak acids. Ammonia (NH3) is a weak base. CH3COOH/CH3COO-, H2CO3/HCO 3, and NH3/NH~- constitute buffer systems.

log[H +] -4- log[OH-] -- log 10 -14 or

pH + pOH -- 14. The pH value of 7 for pure water at 25~ is considered to be neutral, and values below 7 are considered acidic and above 7 basic. Table 1-2 illustrates the pH scale extending from - 1 to + 15. It is important to recognize that as the pH decreases, [H +] increases. A decrease in one pH unit reflects a 10-fold increase in H + concentration. In

A buffer solution functions in the following manner to resist changes in acidity or alkalinity. In an acetic acid/sodium acetate buffer system, the species present in solution are CH3COOH, CH3COO-, Na +, and H20. Amounts of H + and O H - are initially assumed to be small. When acid is added to the buffer, almost all of the H + ions react with acetate ions to produce weakly ionized acetic acid ( H + + CH3COO- Z CH3COOH). The H + ions are thereby prevented from appreciably changing the pH.

SECTION1.2 Buffers

5

weak base (B) and water, the ions O H - and HB + (the conjugate acid) are produced. B + H20 Z

HB + + O H -

The concentration of the conjugate base (or acid) generated from a weak acid (or base) is small, since, by definition, weak acids and bases are only slightly dissociated in aqueous solution. Examples of weak acids are organic acids (e.g., acetic) and of strong acids are mineral acids (e.g., hydrochloric and sulfuric).

Henderson-Hasselbalch Equation

F I G U R E 1-4 Titration profile of acetic acid (CH3COOH) with sodium hydroxide (NaOH). Maximum buffering capacity is at pH = pK ', at which point minimal change in pH occurs upon addition of acid or base.

When O H - is added, almost all of the hydroxyl radicals react with acetic acid molecules to produce more acetate ions and w a t e r ( O H - + CH3COOH ~ CH3COO- + H20). The additional O H - is thus consumed with little increase in pH. Adding H + or O H - to a buffer causes only slight pH changes provided there is excess salt (CH3COO-) or acid (CH3COOH). If all of the acid is converted to the salt form by the addition of a large amount of O H - , the solution can no longer behave as a buffer. Adding more O H - will cause the pH to rise rapidly, as if the solution contained no buffer or only salt. The maximum buffering capacity exists when the molarities of the salt and acid are equal, i.e., when pH = pK' (or - log K'). The pK t is at an inflection point on the titration curve and hence is the point of minimum slope or minimum change in pH for a given addition of acid or base (Figure 1-4). In a generalized weak acid buffer reaction, H20 +

HA ~

K =

[H+][A -] [HA]

(1.4)

where K is the equilibrium constant at a given temperature. For a defined set of experimental conditions, this equilibrium constant is designated as K' (K prime) and referred to as an apparent dissociation constant. The higher the value of K t, the greater the number of H + ions liberated per mole of acid in solution and hence the stronger the acid. K t is thus a measure of the strength of an acid. Rearrangement of Equation (1.4) yields [H +] =

K'[HA] [A-]

(1.5)

Taking logarithms of both sides of Equation (1.5) and multiplying throughout by - 1 gives - log[H +] = - log K' - log[HA] + log[A-]

(1.6)

Substituting pH for - log[H + ] and pK' for - log K' yields

H30 + + A-

a hydronium ion, H3 O+, is formed by the association of a hydrogen ion with a water molecule. In dilute solutions, the concentration of water changes very little when HA is added; therefore, by convention, the dissociation reaction equation is usually written as HA ~-- H + + A -

The Henderson-Hasselbalch equation was developed independently by the American biological chemist L. J. Henderson and the Swedish physiologist K. A. Hasselbalch, for relating the pH to the bicarbonate buffer system of the blood (see below). In its general form, the Henderson-Hasselbalch equation is a useful expression for buffer calculations. It can be derived from the equilibrium constant expression for a dissociation reaction of the general weak acid (HA) in Equation (1.3):

(1.3)

A weak acid, HA, does not readily dissociate, owing to the high affinity of the conjugate base, A - , for the hydrogen ion. Similarly in the hydrolysis reaction of a

[A-] pH = pK' + log ~ [HA]

(1.7)

or

pH = pK ~+ log

[conjugate base] [acid]

(1.8)

This relationship is represented by the HendersonHasselbalch equation. Since a buffer is intended to give only a small change in pH with added H + or O H - , the best buffer for a given pH is the one that gives the smallest change. As may be

6

CHAPTER1 Water, Acids, Bases, and Buffers

TABLE 1-3

TABLE 1-4

Percent Unprotonated Species and Ratio of Unprotonated Forms Relative to the Difference between thepH andpK'*

pH Values of Human Body Fluids and Secretions

%A-

[A-]/[HA]

Log [A-] / [HA] ( p H - pK')

"100" 99 98 96 94 92 91 90 80 70 60 50 40 30 20 10 8 6 4 2 1 "0"

999/1 99/1 98/2 96/4 94/6 92/8 91/9 90/10 80/20 70/30 60/40 50/50 40/60 30/70 20/80 10/90 8/92 6/94 4/96 2/98 1/99 1/999

3.00 2.00 1.69 1.38 1.20 1.06 1.00 0.95 0.60 0.37 0.18 0.00 - 0.18 -0.37 -0.60 -0.95 - 1.06 -1.20 -1.38 -1.69 -2.00 -3.00

*Reproduced, with permission, from J. N. Aronson: The HendersonHasselbalch equation revisited. Biochemical Education 11(2), 68 (1981).

seen from the Henderson-Hasselbalch equation, when the pH of the solution equals the pK' of the buffer, [conjugate base] = [acid], and the buffer can therefore respond equally to both added acid and added base. It also follows from Equation (1.7) that when the pH of the solution is one pH unit above or below the pK' value, the solution contains approximately 9% unprotonated or protonated species, respectively. Similarly, if the pH of the solution is two units above or below the pK' value, the solution contains almost entirely (99%) unprotonated or protonated species, respectively. Table 1-3 provides percent unprotonated species and the corresponding unprotonated/protonated ratios for selected (pH-pK') values. Buffer Systems of Blood and Exchange of 02 and CO2 If the H + concentration departs significantly from its normal value in blood, the health and survival of the human

Body Fluid or Secretion Blood Milk Hepatic bile Gall bladder bile Urine (normal) Gastric juice (parietal secretion) Pancreatic juice Intestinal juice Cerebrospinal fluid Saliva Aqueous humor of eye Tears Urine (range in various disease states) Feces Muscle cell, resting (at 37~ extracellular pH = 7.4)

pH 7.4 6.6-6.9 7.4-8.5 5.4-6.9 6.0 0.87 8.0 7.7 7.4 7.2 7.2 7.4 4.8-7.5 7.0-7.5 6.94-7.06 (intracellular)

body are in jeopardy. H + is the smallest ion, and it combines with many negatively charged and neutral functional groups. Changes of [H+], therefore, affect the charged regions of many molecular structures, such as enzymes, cell membranes, and nucleic acids, and dramatically alter physiological activity. If the plasma pH reaches either 6.8 or 7.8, death may be unavoidable. Despite the fact that large amounts of acidic and basic metabolites are produced and eliminated from the body, buffer systems maintain a fairly constant pH in body fluids (Table 1-4). The major metabolic product from oxidation of ingested carbon compounds is CO2. Hydration of CO2 dissolved in water yields the weak acid H2CO3 (carbonic acid). Depending on the type of food ingested and oxidized, 0.7-1.0 mol of CO2 is produced per mole of O2 consumed. This results in the metabolic production of about 13 mol of hydrated CO2 each day in a normal person. For efficient transport of relatively insoluble CO2 from the tissues where it is formed to the lungs where it must be exhaled, the buffers of the blood convert CO2 to the very soluble anionic form HCO 3 (bicarbonate ion). The principal buffers in blood are bicarbonate-carbonic acid in plasma, hemoglobin in red blood cells, and protein functional groups in both. The normal balance between rates of elimination and production of CO2 yields a steady-state concentration CO2 in the body fluids and a relatively constant pH.

SECTION1.2 Buffers

7

F I G U R E 1-5 Schematic representation of the transport of CO2 from the tissues to the blood. Note that the majority of CO2 is transported as HCO~- in the plasma and that the principal buffer in the red blood cell is hemoglobin. Solid lines refer to major pathways, and broken lines refer to minor pathways. Hb = hemoglobin.

Other acids that are products of metabolism are lactic acid, acetoacetic acid, fi-hydroxybutyric acid, phosphoric acid, sulfuric acid, and hydrochloric acid. The organic acids (e.g., lactate, acetoacetate, and fi-hydroxybutyrate) are normally oxidized further to CO2 and H20. The hydrogen ions and anions contributed by mineral acids and any unmetabolized organic acids are eliminated via the excretory system of the kidneys. Thus, although body metabolism produces a large amount of acid, a constant pH is maintained by transport of H + ions and other acid anions in buffer systems and by elimination of CO2 through alveolar ventilation in the lungs and through excretion of aqueous acids in the urine. Metabolic activities continuously release CO2 to the blood (Figure 1-5), and the lungs continuously eliminate CO2 (Figure 1-6). As oxygen is consumed in peripheral tissues, CO2 is formed and its pressure (Pco2) builds to about 50 mm Hg, whereas the blood entering the tissue capillaries has a Pco2 of about 40 mm Hg. Because of this difference in Pco2 values, CO2 diffuses through the cell membranes of the capillary endothelium and the blood Pco2 rises to 45-46 mm Hg. Despite this increase in Pco2, the blood pH value drops by only about 0.03 during the flow from the arterial capillary (pH 7.41) to the venous capillary (pH 7.38) as a consequence of buffering. About 95% of the CO2 entering the blood diffuses into the red blood cells. Within the red blood cells, the enzyme carbonic anhydrase catalyzes conversion of most of the

C 0 2 to H 2 C O 3 : C O 2 -Jr- H 2 0 ~

H2CO3

H2CO3 dissociates to H + and HCO 3. Although H2CO3 is a weak acid, its dissociation is essentially 100% because of removal of H + ions by the buffering action of hemoglobin. The presence of CO2 and the production of H + cause a reduction in the affinity of hemoglobin for oxygen. Oxyhemoglobin (HbO2)consequently dissociates into oxygen and deoxyhemoglobin (Hb). This effect of pH on the binding of O2 to hemoglobin is known as the Bohr effect (Chapter 28). Oxygen diffuses into the tissues because the Po2 in blood is greater than the Po2 in tissue cells and because protonated deoxyhemoglobin (HHb) is a weaker acid than HbO2 and thereby binds H + more strongly than HbO2. When purified HbO2 dissociates at pH 7.4 to yield oxygen and Hb, the Hb binds 0.7 mol of H + per mole of oxygen released. However, under physiological conditions in whole blood, the Hb combines 0.31 mol of H + per mole of oxygen released. This process is reversible. The remainder of the H + is buffered by phosphate and proteins other than hemoglobin. The major buffering group involved in the transport of H + is an imidazolium group of a histidine residue in hemoglobin. The imidazolium group has a pK' value of about 6.5 (Figure 1-7 depicts the reactions). The difference in acidbase properties between the two forms of hemoglobin molecules is explained by the conformational change that

CHAPTER1 Water, Acids, Bases, and Buffers

F I G U R E 1-6 Schematic representation of the transfer of CO2 from the alveolus (and its loss in the expired air in the lungs) and oxygenation of hemoglobin. Note that the sequence of events occurring in the pulmonary capillaries is the opposite of the process taking place in the tissue capillaries (Figure 1-5). Solid lines indicate major pathways and broken lines indicate minor pathways. Hb - hemoglobin.

accompanies conversion from HbO2 to HHb + (Chapters 7 and 28). As the concentration of HCO 3 (i.e., of metabolic CO2) in red blood cells increases, an imbalance occurs between the bicarbonate ion concentrations in the red blood cell and plasma. This osmotic imbalance causes a marked efflux of HCO 3 to plasma and consequent influx of C1- from plasma in order to maintain the balance of electrostatic charges. The latter osmotic influx, known as the chloride shift, is accompanied by migration of water to red blood cells. Thus, transport of metabolic CO2 in the blood occurs primarily in the form of plasma bicarbonate formed after CO2 diffuses into red blood cells.

F I G U R E 1-7 Buffer function of the imidazole/imidazolium functional groups of histidine residues in protein.

A small percentage of C 0 2 entering the red blood cells combines reversibly with an un-ionized amino group (-NH2) of hemoglobin: hemoglobin-NH2 + CO2 ~ hemoglobin-NH-COO- + H + Hemoglobin-NH-COO-, commonly known as carbaminohemoglobin, is more correctly named hemoglobin carbamate. Formation of this compound causes a lowering of the affinity of hemoglobin for oxygen. Thus, an elevated concentration of CO2 favors dissociation of oxyhemoglobin to oxygen and deoxyhemoglobin. Conversely, CO2 binds more tightly to deoxyhemoglobin than to oxyhemoglobin. All of these processes occurring in the red blood cells of peripheral capillaries are functionally reversed in the lungs (Figure 1-6). Since alveolar P02 is higher than that of the incoming deoxygenated blood, oxygenation of hemoglobin and release of H + occur. The H + release takes place because HbO2 is a stronger acid (i.e., has a lower pK') than deoxyhemoglobin. The released bicarbonate, which is transported to the red blood cells with the corresponding efflux of CI-, combines with the released H + to form H2CO3. Cellular carbonic anhydrase catalyzes dehydration of H2CO3 and release of CO2 from the red blood cells. Thus, red blood cell carbonic anhydrase which catalyzes the reversible hydration of CO2, plays a vital role in carbon dioxide transport and elimination. Carbonic anhydrase is a monomeric (M.W. 29,000) zinc metalloenzyme and is

9

SECTION 1.2 Buffers

present in several different isoenzyme forms. The most prevalent red blood cell isoenzyme of carbonic anhydrase is type I (CAI). Zinc ion, held by coordinate covalent linkage by three imidazole groups of three histidine residues, is involved in the catalytic mechanism of carbonic anhydrase. Water bound to zinc ion reacts with CO2 bound to the nearby catalytic site of carbonic anhydrase to produce H2CO3. The action of carbonic anhydrase is essential for a number of metabolic functions. Some include the formation of H + ion in stomach parietal cells (Chapter 12), in bone resorption by the osteoclasts (Chapter 37) and reclamation of HCO~ in renal tubule cells (Chapter 39). In osteoclasts and in the renal tubule cells, the isoenzyme CAII catalyzes the hydration reaction of CO2. A deficiency of CAII caused by an autosomal recessive disorder consists of osteopetrosis (marble bone disease), renal tubular acidosis, and cerebral calcification. The diffusion of CO2 from venous blood into the alveoli is facilitated by a pressure gradient of CO2 between the venous blood (45 mm Hg) and the alveoli (40 mm Hg) and by the high permeability of the pulmonary membrane to CO2. Blood leaving the lungs has a Pco2 of about 40 mm Hg; thus, essentially complete equilibration occurs between alveolar CO2 and blood CO2.

log

1.3

Taking antilogarithms, [HCOy]

20

proton acceptor

[H2CO3]

1

proton donor

This ratio is large because the pH is greater than the pK' (see Table 1-3). At pH 7.4, the bicarbonate system is a good buffer toward acid (i.e., it can neutralize large amounts of acid) but a poor buffer for alkali. However, blood H2CO3 is in rapid equilibrium with a relatively large (about 1000 times as much) reservoir of cellular CO2 and can function as an effective buffer against increases in alkalinity. The HCO3-/H2CO3 ratio in blood is coupled to the partial pressure of CO2, i.e., to the metabolic production of CO2 and to the loss of CO2 during respiration. In the equilibrium expression for the bicarbonate-carbonic acid buffer system at pH 7.4, the carbonic acid term can be replaced by a pressure term because the carbonic acid concentration is proportional to Pco2 in the blood. pH -- 6.1 + log

[Hco ] a Pco2

[H2CO3] -- aPco2

Carbonic acid has the following pK' values: H + 4- HCO~-

HCO3- ~ CO 2- + H +

[H2CO3]

(1.15)

where a, a proportionality constant, is defined by the equation

Blood Buffer Calculations

H2CO3 ~

[Hco3 ] =

pK'I -- 3.8

(1.9)

pK~ -- 10.2

(1.10)

It is apparent from the pK' values that neither equilibrium can serve as a buffer system at the physiological pH of 7.4. However, carbonic acid (the proton donor) is in equilibrium with dissolved CO2, which in turn is in equilibrium with gaseous CO2: H20 + CO2(aqueous) +~- H2CO3

(1.11)

The hydration reaction (1.11), coupled with the first dissociation of carbonic acid (1.9), produces an apparent pK' of 6.1 for bicarbonate formation. Thus, the summation of Equations (1.9) and (1.11) yields H20 + CO2 ~ H + + HCO3pK' (apparent) -- [HCO~-] [H +] = 6.1 [H2CO3]

(1.12) (1.13)

The ratio of HCO~- to H2CO3 at a physiological pH of 7.4 can be calculated by using the Henderson-Hasselbalch relationship: 7.4 -- 6.1 4- log

[Hco3 ] [H2CO3]

(1.14)

(1.16)

The numerical value of a depends on the solvent and the temperature. For normal plasma at 37~ a 0.0301 mmol of dissolved CO2 per liter of plasma per mm Hg of CO2 pressure. Assuming that 37~ (310 K) is approximately the same as standard temperature, 25~ (298 K), and that [H2CO3] in Equation (1.16) includes both carbonic acid and dissolved CO2, the value of a can be derived from two facts: 1. At 37~ and 760 mm Hg of CO2 pressure, 521 mL of CO2 will dissolve per liter of normal plasma; 2. At standard temperature and pressure, 1 mol of dry CO2 occupies a volume of 22.26 L (not 22.4 L since CO2 is not an ideal gas). a--

521 mL CO2/L of plasma 760 mm Hg x 22.26 mL/mmol of CO2 mmol CO2/L of plasma = 0.0301 mm Hg of CO2 pressure

The equation form of the Henderson-Hasselbalch expression [Equation (1.15)] can be further modified by substituting another expression for the bicarbonate term. When excess strong acid is added to plasma, CO2 is stoichiometrically released from dissolved CO2, carbonic

10

CHAPTER1 Water, Acids, Bases, and Buffers

acid, bicarbonate ions, and carbonate. Carbonate concentration is negligible; thus, Total [CO2] - [HCO3] + dissolved[CO2] + [H2CO3] But Dissolved[CO2] + [H2CO3] = 0.0301Pco2 so that [HCOy ] -- total[CO2] - 0.0301Pco2 Finally, pH -- 6.1 + log

(total [CO2] - 0.0301 Pco2 ) [0.0301Pco2 ] (1.17)

Equation (1.17) is useful for calculating the pH from the total [CO2] and Pco2. The HCO~-/H2CO3 buffer system effectively maintains a constant blood pH of 7.4 if bicarbonate and H2CO3 concentrations are maintained at a ratio of 20:1. The concentration of HCO~- is regulated by its selective excretion and reclamation by the membranes of the renal tubular epithelial cell. Pco2 and [H2CO3] in the blood can be altered by changes in the rate and depth of respiration. For examples, hypoventilation (slow, shallow breathing) leads to increased blood Pco2, whereas hyperventilation (rapid, deep breathing) has the opposite effect. Pco2 changes mediated by the lungs are more rapid than [HCO~-] changes affected through the kidneys (Chapter 39). Nonbicarbonate Buffers in Blood

Other important nonbicarbonate blood buffers are protein and phosphate. The predominant buffer system in the red blood cells is hemoglobin. Protein amino acid side chains (R-groups) that act as buffers are carboxylate groups of glutamate and aspartate and the weakly basic groups of lysine, arginine, and histidine. To be effective, the pK' value of a buffer should be close to the pH of the system to be buffered. Except for the R-group of histidine, which is an imidazolium group (Figure 1-7), the pK' values of the other amino acids mentioned above are not close enough to the physiological pH of blood to be effective buffers. The imidazolium group has a pK' value of 6.5 but it can vary from 5.3 to 8.3 depending on differences in electrostatic environment either within the same protein molecule or in different proteins. Another potential buffering group in protein is the a-amino group of the amino acid residues at the amino terminus of the protein. This group has a pK' value ranging from 7.8 to 10.6, with a typical value of about 8 (acid-base properties of amino acids and proteins are discussed in detail in Chapters 2 and 3). In plasma,

F I G U R E 1-8 Titration profile of phosphoric acid (H3PO4) with sodium hydroxide (NaOH). The three pK' values correspond to three buffer regions. The physiological buffering occurs at the pK' region with H2PO 4 (acid) and HPO 2- (conjugate base) ionic species.

the protein buffer system has a limited role; the principal plasma buffer is the bicarbonate-carbonic acid system. Compared with hemoglobin in the red blood cells and HCO3/HzCO3 in plasma, phosphates (both organic and inorganic) play minor roles in physiological buffering. Phosphoric acid (H3PO4) has three dissociable protons: H3PO4 Z H2PO 2 + H +

p K ' ~ - 1.9

(1.18)

H2PO 2 ~ n P O 2- + n +

pK~ -- 6.8

(1.19)

pK~ = 12.4

(1.20)

HPO42- = PO43- + n +

The titration profile of phosphoric acid with NaOH is shown in Figure 1-8. The principal dissociation expression functioning at a given pH depends on which pK' is closest to the pH. At a plasma pH of 7.4, the important conjugate pair is HPOZ-/HzPO4. The HendersonHasselbalch equation can be used to obtain the value of the ratio HPOZ-/HzPO4 at pH 7.4: [HPO 2- ] 7.4 -- 6.8 + log [H2PO2 ] [HPO2-] = 0.6 log [H2PO 4 ]

and

(1.21)

[HPO2-] =4 [H2PO 4 ]

As was the case for the bicarbonate-carbonic acid system, the conjugate base form (HPO 2-) of the phosphate buffer is present in large (fourfold) excess compared to the acid form (HzPO4) and provides acid buffering capacity. Since the body metabolism produces more acid than

SECTION1.3

Measurementof pH

11

H2N\ NH~

base, this ratio assists in neutralizing acid and in maintaining a constant pH. The HPOZ-/HzP04 buffering system plays a minor role in plasma because of the low concentrations of these ions, but it is important in raising the plasma pH through the excretion of HzPO 4 via the kidney (Chapter 39). In summary, hemoglobin absorbs a major portion of the hydrogen ions produced by the dissociation of H2C03 generated by the hydration of CO2--the most important buffer system in the blood. However, since hemoglobin and carbonic anhydrase are present only in red blood cells, the HCO3/HzCO 3 system in the plasma is an indispensable intermediary in transporting the acid. Thus, the principal method of C02 transport is in the form of HCOf in blood plasma.

_

(CH2)s

y

I

I

Nuclear Magnetic Resonance and Magnetic Resonance Imaging The noninvasive technique of nuclear magnetic resonance (NMR) spectrometry has been used to measure the concentration of H + and other selected ions in isolated cells and tissues. NMR analysis is based on the principle that some atomic nuclei behave like tiny bar magnets because the spinning of charged nuclei generates a magnetic moment along the axis of the spin. If a nucleus with a magnetic dipole (spin) is placed in an external magnetic field, it will acquire an orientation aligned either with the applied field (low-energy state) or against the applied field (high-energy state). The former state is analogous to the way in which a compass needle aligns itself with the earth's magnetic field. Thus, the nuclei, in the presence of an external magnetic field, can remain in either of two unequal energy states. If the aligned nuclei are excited with electromagnetic energy of the proper frequency, some of the nuclei in the low-energy state (ground state) will be excited to the high-energy state. Subsequent release of energy by excited nuclei leads to relaxation back to the ground state and completes the resonance cycle between the two energy states. The NMR spectrum is essentially a measure of the emission of electromagnetic radiation associated with the

I ?H 2

.,

/

ooo-

Glycine

HCNH~ I

COOArginine

J~ Arginine:glycine ~~midinotransferase

/

..;

~ H2N\ //NH~

(OH=)s

C I NH

I

HCNH~

I

I

CH2

COOOrnithine

I

COOGuanidinoacetate

S-adenosylmethionine --~ Guanidinoacetate S-adenosylhomocysteine,~ Methyltransferase(GMT)

1.3 Measurement of pH Blood and urine pH can be measured easily by means of a calibrated glass electrode, whereas pH measurement inside the metabolizing cells is not easily accomplished. Techniques for estimating intracellular pH include glass electrode measurements on homogenates, calorimetric or fluorometric analysis of intracellular distribution of indicator dyes, and microelectrode methods.

NH;

C '1 NH

§

H2 N+\\ / HN-pO2" C

I

Creatine Kinase

HNCH 3 cH2l I COOPhosphocreatine


44 nM), the condition is called acidemia. Conversely, if the pH rises above pH 7.44 ([H +] < 36 nM), the condition is called alkalemia. The suffix -emia refers to blood and usually to an abnormal concentration in blood. Over the pH range of 7.20-7.50, for every change of 0.01 pH unit, there is a change of approximately 1 nM [H +] in the opposite direction. Since the Henderson-Hasselbalch expression uses pH terms, its utility in clinical situations is less than optimal. Kassirer and B leich have derived a modified HendersonHasselbalch expression that relates [H+], instead of pH, to P c o 2 and HCO;, as follows: H2CO3

Clinical applications of Equation (1.23) are discussed in Chapter 39.

Supplemental Readings and References Properties of Water M. E Colombo, D. C. Rau, and V. A. Parsegian: Protein solvation in allosteric regulation: A water effect on hemoglobin. Science 256, 655 (1992). M. A. Knepper: Molecular physiology of urinary concentrating mechanism: Regulation of aquaporin water channels by vasopressin. American Journal of Physiology 272 (Renal Physiology 41), F3 (1997). M. A. Knepper, J. G. Verbalis, and S. Nielsen: Role of aquaporins in water balance disorders. Current Opinion in Nephrology and Hypertension 6, 367 (1997). M. D. Lee, L. S. King, and E Agre: The aquaporin family of water channel proteins in clinical medicine. Medicine 76, 141 (1997). R. E Rand: Raising water to new heights. Science 256, 618 (1992). P. M. Wiggins: Role of water in some biological processes. Microbiological Reviews 54, 432 (1990).

~ H + + HCO 3

Acid-Base Chemistry and Respiratory Function of Hemoglobin

for which

[HC03 ] [H + ] K! =

[H2CO3]

Substituting a x Pco2 for [H2CO3], as in Equation (1.16), yields

H. J. Androgue and N. E. Madias: Management of life-threatening acid-base disorders. New England Journal of Medicine; First of two parts 338, 26 (1998); Second of two parts 338, 107 (1998). C. C. W. Hsia: Respiratory function of hemoglobin. New England Journal of Medicine 338, 239 (1998).

K' = [HCO3] [H+]

Nuclear Magnetic Resonance and Magnetic Resonance Imaging

a x Pc02

and H+ = K'a

x Pco 2

[HCO;]

(1.22)

In Equation (1.22), K' and a are constants, and the numerical value of K'a is 24 when Pco2 is expressed in mm Hg, [HCO3] in mM, and [H+] in nM. Therefore, H+ = 24 • Pco2

[HCO;]

(1.23)

The above formulation expresses the interdependence of three factors; if two of them are known, the third can be readily calculated. For example, at a blood [HCO3] of 24 nM and a Pco2 of 40 mmHg, [H +] is 40 mM.

R. R. Edeman and S. Warach: Medical progress: Magnetic resonance imaging. New England Journal of Medicine; First of two parts 328, 708 (1993); Second of two parts 328, 785 (1993). S. Gilman: Medical progress: Imaging of the brain. New England Journal of Medicine; First of two parts 338, 812 ( 1998); Second of two parts 338, 889 (1998). S. J. Knoury and H. L. Weiner: Multiple sclerosis. Archives of Internal Medicine 158, 565 (1998). L. A. Moulopoulos and M. A. Dimopoulos: Magnetic resonance imaging of the bone marrow in the hematologic malignancies. Blood 90, 2127 (1997). A. Schulze, T. Hess, R. Wevers, et al.: Creatine deficiency syndrome caused by guanidinoacetate methyltransferase deficiency: Diagnostic tools for a new inborn error of metabolism. Journal of Pediatrics 131,626 (1997). S. St6ckler-Ipsiroglu: Creatine deficiency syndromes: A new perspective on metabolic disorders and a diagnostic challenge. Journal of Pediatrics 131, 510 (1997).

CHAPTER

2

Amino Acids

Proteins are the most abundant class of organic compounds in the healthy, lean human body, constituting more than half of its cellular dry weight. Proteins are polymers of amino acids and have molecular weights ranging from approximately 10,000 to more than one million. Biochemical functions of proteins include catalysis, transport, contraction, protection, structure, and metabolic regulation. Amino acids are the monomeric units, or building blocks, of proteins joined by a specific type of covalent linkage. The properties of proteins depend on the characteristic sequence of component amino acids, each of which has distinctive side chains. Amino acid polymerization requires elimination of a water molecule as the carboxyl group of one amino acid reacts with the amino group of another amino acid to form a covalent amide bond. The repetition of this process with many amino acids yields a polymer, known as a polypeptide. The amide bonds linking amino acids to each other are known as peptide bonds. Each amino acid unit within the polypeptide is referred to as a residue. The sequence of amino acids in a protein is dictated by the sequence of nucleotides in a segment of the DNA in the chromosomes and the uniqueness of each living organism is due to its endowment of specific proteins.

proteins have an amino group, a carboxyl group, a hydrogen atom, and an R-group attached to the a-carbon (Figure 2-1). Proline is an exception because it has a cyclic structure and contains a secondary amine group (called an imino group) instead of a primary amine group (called an amino group). Amino acids are classified according to the chemical properties of the R-group. Except for glycine (R = H), the amino acids have at least one asymmetrical carbon atom (the a-carbon). The absolute configuration of the four groups attached to the a-carbon is conventionally compared to the configuration of L-glyceraldehyde (Figure 2-2). The D and L designations specify absolute configuration and not the dextro (right) or levo (left) direction of rotation of planepolarized light by the asymmetrical carbon center. In organic chemistry, the assignment of absolute configurations of an asymmetrical center is made by the R and S classification of isomers. This system prioritizes the substituents linked to the asymmetrical carbon atom (e.g., decreasing atomic number or valence density) and the assignment is based upon the clockwise (R) or the counterclockwise (S) positioning of the three higher priority groups.

2.2 Classification 2.1 L-(x-Amino Acids: Structure Almost all of the naturally occurring amino acids in proteins are L-a-amino acids. The principal 20 amino acids in

A useful classification of the amino acids is based on the solubility (i.e., ionization and polarity) characteristics of the side chains (R-groups). The R-groups fall into four classes:

17

18

CHAPTER2 Amino Acids

COOH

CHO

(~-Carbon

CHO

I

R

OH H

CH2OH

FIGURE 2-1 Basic structureof an or-aminoacid.

H OH

CH2OH

CHO 1. 2. 3. 4.

Nonpolar (hydrophobic), Polar, negatively charged (acidic), Polar, positively charged (basic), and Polar, neutral (un-ionized).

Within each class, R-groups differ in size, shape, and other properties. Figure 2-3 shows the structure of each amino acid according to this classification with the Rgroup outlined. Ionizable structures are drawn as they would exist at pH 7.0. The three-letter and one-letter abbreviations for each amino acid are given in Table 2-1. A -yl ending on amino acid residue indicates that the carboxyl group of an amino acid is linked to another functional group (e.g., in a peptide bond). The eight essential amino acids (Table 2-1) are those which humans cannot synthesize and which must be supplied in the diet. The remaining amino acids are synthesized in the body by various biochemical pathways (Chapter 17).

I

H~C~OHI

CHO

I

OH--?~H

CH2OH D-Glyceraldehyde

CH2OH L-Glyceraldehyde

CHO

CHO

9

9

H " - C--- OH 9

H0 - - C.,--'.H 9

CH,OH

CH,OH

Fischer perspective formulas CHO

CHO OH

H O .......I

CH2OH

H

CH2OH

Fischer projection formulas Nonpolar Amino Acids

COOH

COOH

9

Glycine Glycine is the smallest amino acid and has an H atom as its R-group. It is the only u-amino acid that is not optically active. The small R-group provides a minimum of steric hindrance to rotation about bonds; therefore, glycine fits into crowded regions of many peptide chains. Collagen, a rotationally restricted fibrous protein, has glycyl residues in about every third position in its polypeptide chains. Glycine is used for the biosynthesis of many nonprotein compounds, such as porphyrins and purines. Glycine and taurine are conjugated with bile acids, products derived from cholesterol, before they are excreted into the biliary system. Conjugated bile acids are amphipathic and are important in lipid absorption (Chapter 12). Glycine also is a neurotransmitter; it is inhibitory in the spinal cord and excitatory in the cerebral cortex and other regions of the forebrain. Nonketotic hyperglycinemia (NKH) is an inborn error of glycine degradation in which a large amount of glycine accumulates throughout the body. NKH causes

~

9

H--C-=" NH2

H,N-- r

~

(-,H3 D-Alanine

CH3 L-Alanine

FIGURE 2-2 Different representations of the configurational stereoisomersof glyceraldehydeand of alanine.

severe consequences in the central nervous system (CNS) and leads to death (Chapter 17).

CO0 I +H3N--C--H I H Glycine

SECTION2.2 Classification

FIGURE 2-3 Classification of commonly occurring L-a-amino acids based on the polarity of side chains (R-groups) at pH 7.0.

19

20

CHAPTER2 Amino Acids

TABLE 2-1 Amino Acid Abbreviations and Nutritional Property

Amino acid

Abbreviation

Designation letter

Nutritional property*

Alanine Arginine

Ala Arg

A R

Asparagine Aspartic acid Cysteine Glutamic acid Glutamine

Asn Asp Cys Glu Gln

N D C E Q

non-essential conditionally essential non-essential non-essential non-essential non-essental conditionally essential non-essential conditionally essential essential essential essential essential essential non-essential non-essential essential essential non-essential essential

Glycine Histidine

Gly His

G H

Isoleucine Leucine Lysine Methionine Phenylalanine Proline Serine Threonine Tryptophan Tyrosine Valine

lie Leu Lys Met Phe Pro Ser Thr Trp Tyr Val

I L K M F P S T W Y V

*The eight essential amino acids are not synthesized in the body and have to be supplied in the diet. The conditionally essential amino acids, although synthesized in the body, may require supplementation during certain physiological conditions such as pregnancy. The non-essential amino acids can be synthesized from various metabolites.

Valine, Leucine, and Isoleucine These branched-chain aliphatic amino acids contain bulky nonpolar R-groups and participate in hydrophobic interactions. All three are essential amino acids. A defect in their catabolism leads to maple syrup urine disease (Chapter 17). Isoleucine has asymmetrical centers at both the o~- and/3-carbons and four stereoisomers, only one of which occurs in protein. The bulky side chains tend to associate in the interior of water-soluble globular proteins. Thus, the hydrophobic amino acid residues stabilize the three-dimensional structure of the polymer.

COOCOO-

l

+H3N~C~H

I

I

+H3N~C~H

CH2

I

/ \

I

CH

H3C

OH 3 H3C

Valine

/ \

COO-

I

+H3N~C~H

I

H~C~CH 3

I

CH

CH2 OH3

Leucine

i

OH 3

Isoleucine

Phenylalanine A planar hydrophobic phenyl ring is part of the bulky R-group of phenylalanine. It is an essential amino acid whose metabolic conversion to tyrosine is defective in phenylketonuria (Chapter 17). Phenylalanine, tyrosine, and tryptophan are the only a-amino acids that contain aromatic groups and consequently are the only residues that absorb ultraviolet (UV) light (Figure 2-4). Tryptophan and tyrosine absorb significantly more energy than phenylalanine at 280 nm, the wavelength generally used to measure the concentration of protein in a solution.

CO0 -

Alanine

I

The side chain of alanine is a hydrophobic methyl group,--CH3. Other amino acids may be considered to be chemical derivatives of alanine, with substituents on the fl-carbon. Alanine and glutamate provide links between amino acid and carbohydrate metabolism (Chapter 22).

+H3N~C~H

I

COO -

Phenylalanine

I

+H3N~C~H Tryptophan

,~CarrbOn.~CiH3

Alanine

A bicyclic nitrogenous aromatic ring system (known as an indole ring) is attached to the /~-carbon of alanine to form the R-group of tryptophan. Tryptophan is a precursor of serotonin, melatonin, nicotinamide, and many

21

SECTION 2.2 Classification

FIGURE 2-4 UV absorption spectra of phenylalanine, tyrosine, and tryptophan.

naturally occurring medicinal compounds derived from plants. It is an essential amino acid. The indole group absorbs UV light at 280 n m - - a property that is useful for spectrophotometric measurement of protein concentration (Figure 2-4). Tryptophan and tyrosine both show fluorescence; however, tryptophan absorbs more intensely. When molecules are raised to a higher energy state by absorption of radiant energy, they generally are unstable and return to the ground state. The energy released in this process manifests as heat (radiation energy) or light. The process of light emission is known as fluorescence. The quantum of energy re-emitted as fluorescence is always less than that of absorbed energy. Thus, the fluorescent light always appears at a longer wavelength (lower energy) than the original absorbed light energy (Figure 2-5). Tryptophan fluorescence studies can provide valuable information regarding protein and protein-ligand conformations due to the effects of surrounding amino acid residues. COO -

I + H3N---C--- H

I CH2 I

N/CH Tryptophan

Methionine This essential amino acid contains an R-group with a methyl group attached to sulfur. Methionine serves as donor of a methyl group in many transmethylation reactions, e.g., in the synthesis of epinephrine, creatine, and

F I G U R E 2-5 Tryptophan fluorescence spectrum. The emission spectrumappears at longer wavelengths as compared to the absorption spectrum.

melatonin. Almost all of the sulfur-containing compounds of the body are derived from methionine. COO -

I +H3N~C---H I CH2 I CH2 I S I CH3

Methionine Proline Proline contains a secondary amine group, called an imine, instead of a primary amine group. For this reason, proline is called an imino acid. Since the three-carbon R-group of proline is fused to the ol-nitrogen group, this compound has a rotationally constrained rigid-ring structure. As a result, prolyl residues in a polypeptide introduce restrictions on the folding of chains. In collagen, the principal protein of human connective tissue, certain prolyl residues are hydroxylated (Figure 2-6). The hydroxylation occurs during protein synthesis and requires ascorbic acid (vitamin C) as a cofactor. Deficiency of vitamin C causes formation of defective collagen and scurvy (Chapters 25 and 38). COO -

I C~H I /OH2

+H2N ! H2C ~CH2

Proline

22

CHAPTER2 Amino Acids COOH

1COOH

HN"

COOH

H2N--C--H

2~C--H

H2C~ 4~CH2 ~C~ H OH 4-Hydroxyproline

CH2 HC--OH CH2

Present in collagen, a fibrous protein. COOH

I H2N--C--H I CH2 I CH~_ I CH2 I

O O-Phosphoserine

NH2 5-Hydroxylysine

COOH

I I

H=N--C--H COOH

CH=

I I CH2 I

r

H2N--C--H

C

H3C--N~~N

I

t

-C

I

ECH2

I

CH2 ,

I

(H3C),N--C--H

N

H a-Methylhistidine Present in myosin, a muscle protein.

H ~CH3 ~-N-Methyllysine The N-trimethyl derivative of lysine is involved in the synthesis of carnitine.

COOH

I I CH2 I

COO -

I H=N ~ C ~ o Diphthamide

A novel derivative of histidine present only in the eukaryotic protein elongation factor 2 (EF-2), which participates in the elongation step of protein biosynthesis. Diphtheria toxin inhibits eukaryotic protein synthesis by catalyzing a covalent modification of diphthamide (see Chapter 25).

H2N--C--H

I

HN - - - - - - - C - - H

I

Phosphorylationand dephosphorylation of selectedserine residuesin a variety of proteinsplayan importantrole in the regulationof metabolismand are mediatedby some hormones.

I I CH2 I O I HO--P--OH II

H2N--C--H

CH2

I

O---G~ ~CH2 C H2

CH ,OOC "~ ~COOH 7-Carboxyglutamic acid Present in certain blood-clotting proteins

Pyrrolidone car boxylate (pyroglutamate) Present in some proteins and peptides as N-terminal amino acid residue. H2N~

/COOH

c~ ;H~3 H2N~

fNH2

4z=.~~

HOoc/CH-- (CH,)~I

~3

NH2

(CRy#__ CH ~ C O O H

HOOc./CH -- (CH=)

(OH=)E--CH--COOH (CH=)F--CH--COOH 'I"

,H~,

(?H=),

H2NfC%ooH Desmosine

H~C%ooH Isodesmosine Desmosineand isodesmosineare formedfrom lysylresidues of the polypeptidechainsof elastin,a fibrousprotein.

FIGURE 2-6 Modified derivatives of certain amino acids are found in proteins.

I

NH2

23

SECTION2.2 Classification

Acidic Amino Acids

Aspartic Acid The ]3-carboxylic acid group of aspartic acid has a pK' of 3.86 and is ionized at pH 7.0 (the anionic form is called aspartate). The anionic carboxylate groups tend to occur on the surface of water-soluble proteins, where they interact with water. Such surface interactions stabilize protein folding. COO -

I

residues are present in a number of blood coagulation proteins (factors II, VII, IX, and X) and anticoagulant proteins C and S (Chapter 36). Osteocalcin, a protein present in the bone, also contains v-carboxyglutamate residues (Chapter 37). A cyclic, internal amide derivative of glutamic acid is pyrrolidone carboxylic acid (also known as pyroglutamic acid or 2-oxoproline). Some proteins (e.g., heavy chains of immunoglobulins; Chapter 35) and peptides (e.g., thyrotropin-releasing hormone; Chapter 33) have pyroglutamic acid as their N-terminal amino acid residue.

+ H3N---~C~H COO -

I

~CH2

I

I +H3N--(zC--H

I ~CH2 I

yCH2

Aspartate

I O//'C~o Glutamate

Glutamic Acid The v-carboxylic acid group of glutamic acid has a pK' of 4.25 and is ionized at physiological pH. The anionic groups of glutamate (like those of aspartate) tend to occur on the surfaces of proteins in aqueous environments. Glutamate is the primary excitatory neurotransmitter in the brain. Its levels are regulated by clearance that is mediated by glutamate transfer protein in critical motor control areas in the CNS. In amyotrophic lateral sclerosis (ALS) glutamate levels are elevated in serum, spinal fluid, and brain; glutamate excitotoxicity is implicated in the progression of the disease. ALS is a progressive disorder affecting motor neurons in the spinal cord, brain stem, and cortex. The precise molecular basis of the disease is unknown; however, factors involved are glutamate excitotoxicity, genetics, oxidative stress, and diminished neurotrophic factors. Two drugs that provide neuroprotection against glutamate excitotoxicity are riluzole and gabapentin. Gabapentin is an amino acid structurally related to the neurotransmitter v-aminobutyrate (GABA). GABA, an inhibitory neurotransmitter in the CNS, is produced by the decarboxylation of glutamate by glutamate decarboxylase, a pyridoxal phosphate dependent enzyme. GABA, when bound to its receptors, causes an increase in permeability to chloride ions in neuronal cells. A group of tranquilizing drugs known as benzodiazopines enhance the membrane permeability of chloride ions by GABA. In some proteins, the v-carbon of glutamic acid contains an additional carboxyl group. Residues of v-carboxyglutamic acid (Gla) bear two negative charges and can strongly bind calcium ions. v-Carboxylation of glutamic acid residues is a posttranslational modification and requires vitamin K as a cofactor, v-Carboxyglutamate

COO -

I

+H3NmCmH

COO -

I C--H I OH2

HN I

O---C

~c~

H2 Pyrrolidone carboxylate (pyroglutamate)

NH2 I

I

CH2

CH 2

I CH2 I

I

CH I O--c/C~c=o l I OO-

CH2 I

COO-

},-amino butyrate (GABA)

y - C a r b o x y glutamate

Basic Amino Acids

Lysine Lysine is an essential amino acid. The long side chain of lysine has a reactive amino group attached to the s-carbon. The s-NH2 ( p K ' = 10.53) is protonated at physiological pH. The lysyl side chain forms ionic bonds with negatively charged groups of acidic amino acids. The s-NH2 groups of lysyl residues are covalently linked to biotin (a vitamin), lipoic acid, and retinal, a derivative of vitamin A and a constituent of visual pigment. In collagen and in some glycoproteins, g-carbons of some lysyl residues are hydroxylated (Figure 2-6), and sugar moieties are attached at these sites. In elastin and collagen, some s-carbons of lysyl residues are oxidized to reactive aldehyde (-CHO) groups, with elimination of NH3. These aldehyde groups then react with other s NH2 groups to form covalent cross-links between polypeptides, thereby providing tensile strength and insolubility to

24

CHAPTER2 Amino Acids

protein fibers. Examples of cross-linked amino acid structures are desmosine, isodesmosine (Figure 2-6), dehydrolysinonorleucine, lysinonorleucine, merodesmosine, and dehydromerodesmosine (Chapter 10). Lysyl R-groups participate in a different type of cross-linking in the formation of fibrin, a process essential for the clotting of blood. In this reaction, the s-NH2 group of one fibrin polypeptide forms a covalent linkage with the glutamyl residue of another fibrin polypeptide (Chapter 36).

by diphenhydramine and pyrilamine. Hi-receptor antagonists are used in the treatment of allergic disorders. Secretion of HC1 by the stomach (Chapter 12) and an increase in heart rate are mediated by H2 receptors. Examples of Hz-receptor antagonists of histamine action are cimetidine and ranitidine, agents used in the treatment of gastric ulcers.

Arginine COO -

The positively charged guanidinium group attached to the 6-carbon of arginine is stabilized by resonance between the two NH2 groups and has a pK' value of 12.48. Arginine is utilized in the synthesis of creatine and it participates in the urea cycle (Chapter 17). The nitrogen of the guanidino group of arginine is converted to nitric oxide (NO) by nitric oxide synthase. NO is unstable, highly reactive, and has a life span of only a few seconds. However, NO affects many biological activities, including vasodilation, inflammation, and neurotransmission (Chapter 17).

+H3N~=C~H /YCH2 yCH2 6CH2 sCH2 NH3 + Lysine

Histidine

COO-

The imidazole group attached to the/~-carbon of histidine has a pK' value of 6.0. The pK' value of histidyl residues in protein varies depending on the nature of the neighboring residues. The imidazolium-imidazole buffering pair has a major role in acid-base regulation (e.g., hemoglobin). The imidazole group functions as a nucleophile, or a general base, in the active sites of many enzymes and may bind metal ions. Histidine is nonessential in adults but is essential in the diet of infants and individuals with uremia (a kidney disorder). Decarboxylation of histidine to yield histamine occurs in mast cells present in loose connective tissue and around blood vessels, basophils of blood, and enterochromaffin-like (ECL) cells present in the acid-producing glandular portion (oxyntic cells) of the stomach. The many specific reactions of histamine are determined by the type of receptor (HI or H2) present in the target cells. The contraction of smooth muscle (e.g., gut and bronchi) is mediated by Hi receptors and antagonized

l

+H3N--CmH

I

CH2

I

CH2

I

6CH2

I

NH

I

C~'NH2 ~ Guanidinium I

,

NH~.. . . . . !

Neutral

Amino

Acids

Serine The primary alcohol group of serine can form esters __.:*.L

wit.

._1_

I . . . .

." _

p.u~p.ull~

~

_

". _1

[11 ~'.

. . . . . .

,~l

,

+H3N--C~H

I I

I

C

I

I

I

N,.

~c / H Histidine

NH

N,.

"~c j H Histamine

_

1_

_

"_ _1 . . . . .

:a_l_

-

I

I

CH

_1

Hm C - - O H

I

CH2

C

._

+H3NmC--H

I

H3N~C~H

CH2

~

I

H

I

f'~

a~lu t l " l g u l c z.-U) allu gly~u~luc~ wluJ COO

COO -

group

Arginine

CH

I

NH

H Serine

sugars. The phosphorylation and dephosphorylation processes regulate the biochemical activity of many proteins. Active centers of some enzymes contain seryl hydroxyl

SECTION2.2 Classification

25

groups and can be inactivated by irreversible derivatization of these groups. T h e - O H group of serine has a weakly acidic pK' of 13.6.

Threonine This essential amino acid has a second asymmetrical carbon atom in the side chain and therefore can have four isomers, only one of CO0 -

I

+H3N~C~H

I

H~C~OH

I

CH3 Threonine

which, L-threonine, occurs in proteins. The hydroxyl group, as in the case of serine, participates in reactions with phosphoric acid and with sugar residues.

Cysteine The weakly acidic ( p K ' = 8.33) sulfhydryl group (-SH) of cysteine is essentially undissociated at physiological pH. Free-SH groups are essential for the function of many enzymes and structural proteins. Heavy metal ions, e.g., Pb 2+ and Hg 2+, inactivate these proteins by combining with their-SH groups. Two cysteinyl-SH groups can be oxidized to form cystine. A covalent disulfide bond of cystine can join two parts of a single polypeptide chain or two different polypeptide chains through cross-linking of cysteine residues. These - S - S - bonds are essential both for the folding of polypeptide chains and for the association of polypeptides in proteins that have more than one chain, e.g., insulin and immunoglobulins.

Tyrosine The phenolic hydroxyl group of this aromatic amino acid has a weakly acidic pK' of about 10 and therefore is un-ionized at physiological pH. In some enzymes, the hydrogen of the phenolic hydroxyl group can participate in hydrogen bond formation with oxygen and nitrogen atoms. The phenolic hydroxyl group of tyrosine residues in protein can be sulfated (e.g., in gastrin and cholecystokinin; see Chapter 12) or phosphorylated by a reaction catalyzed by the tyrosine-specific protein kinase that is a product of some oncogenes (Chapter 26). Tyrosine kinase activity also resides in a family of cell surface receptors that includes receptors for such anabolic polypeptides as insulin, epidermal growth factor, plateletderived growth factor, and insulin-like growth factor type 1. All of these receptors have a common motif of an external ligand binding domain, a transmembrane segment, and a cytoplasmic tyrosine kinase domain (Chapter 22). Tyrosine accumulates in tissues and blood in tyrosinosis and tyrosinemia, which are due to inherited defects in catabolism of this amino acid. Tyrosine is the biosynthetic precursor of thyroxine, catecholamines, and melanin. Tyrosine and its biosynthetic precursor, phenylalanine, both absorb UV light (Figure 2-4). COO -

I

+H3N--C--H

I

CH2

C) OH Tyrosine

Asparagine The R-group of this amide derivative of aspartic acid has no acidic or basic properties but is polar and participates in hydrogen bond formation. It is hydrolyzed to aspartic acid and ammonia by the enzyme asparaginase. In glycoproteins, the carbohydrate side chain is often linked through the amide group of asparagine.

CO0 -

I

+H3N~C~H

I

CH2

I

SH Cysteine COO

-

I

+H3N~C~H CO0 -

COO-

I

I

+H3N~C~H

+H3N~C~H

I

CH2~S~S Cystine

I

CH2

I

CH2

I O//'C~NH 2 Asparagine

2fi

CHAPTER 2 AminoAcids Glutamine

This amide of glutamic acid has properties similar to those of asparagine. The 7-amido nitrogen, derived from ammonia, can be used in the synthesis ofpurine and pyrimidine nucleotides (Chapter 27), converted to urea in the liver (Chapter 17), or released as NH3 in the kidney tubular epithelial cells. The last reaction, catalyzed by the enzyme glutaminase, functions in acid-base regulation by neutralizing H + ions in the urine (Chapter 39). Glutamine is the most abundant amino acid in the body. It composes more than 60% of the free amino acid pool in skeletal muscle. It is metabolized in both liver and gut tissues. Glutamine, along with alanine, are significant precursors of glucose production during fasting (Chapter 15). It is a nitrogen donor in the synthesis of purines and pyrimidines required for nucleic acid synthesis (Chapter 27). Glutamine is enriched in enteral and parenteral nutrition to promote growth of tissues; it also enhances immune functions in patients recovering from surgical procedures. Thus, glutamine may be classified as a conditionally essential amino acid during severe trauma and illness. CO0 -

I

+H3N---C~H

I CH2

I

CH2

I O~/'C~NH 2 Glutamine

Unusual Amino Acids

Several L-amino acids have physiological functions as free amino acids rather than as constituents of proteins. Examples are as follows: 1. ~-Alanine is part of the vitamin pantothenic acid. 2. Homocysteine, homoserine, omithine, and citrulline are intermediates in the biosynthesis of certain other amino acids. 3. Taurine, which has an amino group in the ~-carbon and a sulfonic acid group instead of COOH, is present in the CNS and as a component of certain bile acids participates in digestion and absorption of lipids in the gastrointestinal tract. 4. y-Aminobutyric acid is an inhibitory neurotransmitter. 5. Hypoglycin A is present in unripe akee fruit and produces severe hypoglycemia when ingested.

6. Some D-amino acids are found in polypeptide antibiotics, such as gramicidins and bacitracins, and in bacterial cell wall peptides.

Amino Acids Used as Drugs D-Penicillamine (D-fl,/3-dimethylglycine), a metabolite of penicillin, was first isolated in the urine specimens from patients treated with penicillin with liver disease. It is an effective chelator of metals such as copper, zinc, and lead. It is used in the chelation therapy of Wilson's disease, which is characterized by excess copper accumulation leading to hepatolenticular degeneration (Chapter 37).

CH3

I I

H3C ~ C ~ CH - - CO0"

I

SH NH + D-penicillamine

N-Acetylcysteine is administered in the acetoaminophen toxicity. It replenishes the hepatic stores of glutathione (Chapter 17). N-Acetylcysteine is also used in the treatment of pulmonary diseases including cystic fibrosis (Chapter 12). In patients with chronic renal insufficiency, prophylactic oral administration of N-Acetylcysteine have been used in the prevention of further renal impairment due to administration of radiographic contrast agents. In this setting presumably N-Acetylcysteine functions as an antioxidant and augments the vasodilatory effect of nitric oxide via the formation of S-nitrosothiol (Chapter 17). COOH

I

O

II

HS~CH2---CH--NH ~C~CH

3

N-acetylcysteine

Gabapentin is y-aminobutyrate covalently linked to cyclohexane to make it lipophilic and to facilitate its transport across the blood-brain barrier. It is used as an anticonvulsant and in amyotrophic lateral sclerosis (ALS).

H3N'~COO Gabapentin

SECTION2.3 Electrolyte and Acid-Base Properties

2.3 Electrolyte and Acid-Base Properties

I

H /o I

Nonpolar neutral

Amino acids are ampholytes, i.e., they contain both acidic and basic groups. Free amino acids can n e v e r occur as neutral nonionic molecules:

H

27

0

II

R--C--C--OH

I

= R--C--CI

R-group

I NH3

~"0

Zwitterion

Zwitterions are electrically neutral and so do not migrate in an electric field. In acidic solution (below pH 2.0), the predominant species of an amino acid is positively charged and migrates toward the cathode: H

O

I

II

R--C--C--OH

I NH3 +

FIGURE 2-7 Titration profile of glycine, a monoaminomonocarboxylicacid.

Resonance,-- stabilized carboxylate anion

Ammonium cation

NH2

Instead, they exist as neutral z w i t t e r i o n s that contain both positively and negatively charged groups:

e

28

CHAPTER2 Amino Acids

In basic solution (above pH 9.7), the predominant species is negatively charged and migrates toward the anode:

H I

O H

RmC_C_O I NH2

FIGURE 2-8 Titration profile of histidine.

-

The isoelectric point (pI) of an amino acid is the pH at which the molecule has an average net charge of zero and therefore does not migrate in an electric field. The pI is calculated by averaging the pK' values for the two functional groups that react as the zwitterion becomes alternately a monovalent cation or a monovalent anion. At physiological pH, monoaminomonocarboxylic amino acids, e.g., glycine and alanine, exist as zwitterions.

SECTION 2.3

Electrolyte and Acid-Base Properties

That is, at a pH of 6.9-7.4, the oe-carboxyl group (pK' 2.4) is dissociated to yield a negatively charged carboxylate ion (-COO-), while the ol-amino group (pK' 9.7) is protonated to yield an ammonium group (-NH~-). The pK' value of the o~-carboxyl group is considerably lower than that of a comparable aliphatic acid, e.g., acetic acid (pK' 4.6). This stronger acidity is due to electron withdrawal by the positively charged ammonium ion and the consequent increased tendency of a carboxyl hydrogen to dissociate as an H +. The or-ammonium group is corre-

FIGURE 2-9 Titrationprofileof lysine.

29 spondingly a weaker acid than an aliphatic ammonium ion, e. g., ethylamine (pK' 9.0) because the inductive effect of the negatively charged carboxylate anion tends to prevent dissociation of H +. The titration profile of glycine hydrochloride (Figure 2-7) is nearly identical to the profiles of all other monoaminomonocarboxylic amino acids with nonionizable R-groups (Ala, Val, Leu, Ile, Phe, Ser, Thr, Gln, Asn, Met, and Pro). The titration of glycine has the following major features. The titration is initiated with glycine hydrochloride,

30

CHAPTER2 Amino Acids

F I G U R E 2-11} Titration profile of aspartic acid.

CI-(H~-NCH2COOH), which is the fully protonated form of the amino acid. In this form, the molecule contains two acidic functional groups; therefore, two equivalents of base are required to completely titrate 1 mol of glycine hydrochloride. There are two pK' values: pK'1 due to reaction of the carboxyl group and pK~ due to reaction of the ammonium group. Addition of 0.5 eq of base to 1 mol of glycine hydrochloride raises the pH 2.34 (PKtl), whereas addition of 1.5 eq further increases the pH to 9.66 (pK~). At low pH values (e.g., 0.4), the molecules are

predominantly cations with one positive charge; at pH values of 5-7, most molecules have a net charge of zero; at high pH values (e.g., 11.7), all of the molecules are essentially anions with one negative charge. The midpoint between the two pK' values [i.e., at pH = (2.34 + 9.66)/2 = 6.0] is the pI. Thus, pI is the arithmetic mean of pK'1 and pK~ values and the inflection point between the two segments of the titration profile. The buffering capacities of weak acids and weak bases are maximal at their pK values. Thus, monoaminomono-

SECTION 2.4

Chemical Reactions of Amino Acids

31

TABLE 2-2 pK'and p l Values of Selected Free Amino Acids at 25~ * Amino Acid

pI

pK' 1 (a-COOH)

pK' 2

pK' 3

Alanine

2.34

9.69 (a-NH3+)

Aspartic acid

2.09

3.86 (/3-COOH)

9.82 (a-NH 3+)

298(pKI+pK21) 2

Glutamic acid

2.19

4.25 (7-COOH)

9.67 (tx-NH3+)

3.22 ( pK'I +2pK'2)

Arginine

2.17

9.04 (a-NH3+)

Histidine

1.82

6.00 (Imidazolium)

Lysine

2.18

Cysteine

6.00

10.76

pK' + PK'3) 2

9.17 (NH3+)

7.59

(eKe+eKe) 2

8.95 (a-NH3+)

10.53 (e-NH3 +)

9.74

(PK'2 + PK'3) 2

1.71

8.33 (-SH)

10.78 (~-NH3+)

5.02 ( pK'I +2pK'2 )

Tyrosine

2.20

9.11 (a-NH3 +)

10.07 (Phenol OH)

5.66 ( pK'I +2pK'2 )

Serine

2.21

9.15 (a-NH3 +)

13.6 (Alcohol OH)

5.68 ( pK'I +2pK'2)

12.48 (Guanidinium)

*ThepK' valuesfor functionalgroupsin proteinsmay varysignificantlyfromthe valuesfor free aminoacids.

carboxylic acids exhibit their greatest buffering capacities in the two pH ranges near their two pK' values, namely, pH 2.3 and pH 9.7 (Figure 2-7). Neither these amino acids nor the a-amino or ot-carboxyl groups of other amino acids (which have similar pK' values) have significant buffering capacity in the neutral (physiological) pH range. The only amino acids with R-groups that have buffering capacity in the physiological pH range are histidine (imidazole; pK' 6.0) and cysteine (sulfhydryl; pK' 8.3). The pK' values for R-groups vary with the ionic environment. The titration profile of histidine is shown in Figure 2-8. The pI is the mean of pK~ and pKg. Titration profiles of the basic and acidic amino acids lysine and aspartic acid are shown in Figures 2-9 and 2-10. The R-groups are ionized at physiological pH and have anionic and cationic groups, respectively. The pI value for aspartic acid is the arithmetic mean of pK'1 and pK~, whereas for lysine and histidine the pI values

are given by the arithmetic mean of pK~ and pKg. The pK' and pI values of selected amino acids are listed in Table 2-2.

2.4 Chemical Reactions of Amino Acids The reactions of amino acids with ninhydrin, carbon dioxide, metal ions, and glucose are described below. The last three are of physiological importance. Ninhydrin (triketohydrindene hydrate) reacts with o~amino acids to produce CO2, NH3, and an aldehyde with one less carbon than the parent amino acid. In most cases, a blue or violet compound (proline and hydroxyproline give a yellow color) is formed owing to reaction of the liberated NH3 with ninhydrin, as shown in Figure 2-11. Color and CO2 production provide a basis for the quantitative determination of amino acids.

32

CHAPTER2 AminoAcids O

II

H

C\ /OH

I

RmCRCOOH

7~OH

I

NH2

II

o Ninhydrin

Amino acid H +

CO2

+

H20

O

II

7 ~NH 2 I! o

o II Ninhydrin

II

o o

o-

QL I!

I

cT=N-\

§ 2.20

II

II

o o Purple-coloredion FIGURE 2-11 Reaction of an o~-aminoacid with ninhydrin.Two moleculesof ninhydrin and the nitrogenatomof the aminoacid are involvedin the productionof the purpleproduct.

Ammonia, some amines, and some proteins and peptides will also yield a colored product but will not generate CO2. Thus, the colorimetric analysis is not specific for amino acids unless CO2 release is measured or the amino acids are purified and freed from interfering materials (the usual procedure). The color reaction with ninhydrin is used extensively in manual and automated procedures. CO2 adds reversibly to the un-ionized amino group of an amino acid. The product is a carbamate (or carboxyamino) derivative.

0

0

C/LO ~C

H2NaC--H

I j

R

H =

Cl[110N--C--H

I O / / , C . o _R

+

H+

This type of reaction accompanies transport of C02 in the blood (Chapter 1). In tissue capillaries, CO2 combines with free o~-amino groups of hemoglobin to form carbaminohemoglobin; in pulmonary capillaries, this reaction is reversed to release CO2 into the alveoli. This mode of transport is limited to only about 10% of the carbon dioxide transported in the blood. Metal ions can form complexes with amino acids. Metal ions that function in enzymatic or structural biochemical systems include those of iron, calcium, copper, zinc, magnesium, cobalt, manganese, molybdenum, nickel, and chromium. The functional group that binds a metal ion is called a ligand. Ligands are electron donors that form noncovalent bonds with the metal ions, usually two, four, or six ligand groups per ion. When four ligand groups bind a metal ion, the complex is either a plane or a tetrahedron; when six ligand groups participate, octahedral geometry results. The term chelation is applied to a metal-ligand interaction when a single molecule provides two or more ligands (e.g., chelation of iron with four nitrogens in one porphyrin molecule; see Chapter 14). Metal ions can also react with amino acid functional groups to abolish the biological activity of proteins. Heavy metal ions that form highly insoluble sulfides (e.g., HgS, PbS, CuS, AgzS) characteristically react with sulfhydryl groups of cysteinyl residues. If the reactive-SH group is involved in biological activity of the protein, the displacement of the hydrogen and the addition of a large metal atom to the S atom usually cause a major change in protein structure and loss of function. Hence, heavy metals are often poisons. In contrast, amino acid residues in proteins may undergo nonenzymatic chemical reactions that may or may not alter biological activity. An example of this type of reaction is the formation of glycated proteins. The amino groups of proteins combine with carbonyl groups of sugars (glucose) to form labile aldimines (Schiff bases), which are isomerized (Amadori rearrangement) to yield stable ketoamine (fructosamine) products (Figure 2-12). The degree of glycation achieved in a protein is determined by the concentration of sugar in the environment of the protein. In glycated hemoglobin, a Schiff-base adduct forms between the sugar and the N-terminal group of the /~-chains of hemoglobin. The Amadori sugar-amino acid residue adducts in proteins are produced with prolonged hyperglycemia and undergo progressive nonenzymatic reactions involving dehydration, condensation, and cyclization. These compounds are collectively known as advanced glycosylation end products and are involved in the chronic complications of diabetes mellitus (cataracts and nephropathy) (Chapter 22).

SupplementalReadingsand References

33

~2COH O

.of~ Glucose -H20

RNH2

Aminogroup of a protein

H OH

1[

H2COH

H2COH OH HR

H(

H pNR

-_ ~

H

H

H

/

1

, ,

OH Glucosylamine H+[

OH Schiff base [ Amadorirearrangement

H2COH H2R H(

H

H2c~O"~ ' ~

OH

OH

[02]

H~//(~H2NHR

OH 1-Amino-1-deoxy2-keto compound

HN

I (?H2)3 CH

I

((~H2)4 CH I~

I~ H2N COON H2N COOH Pentosidine

F I G U R E 2-12 Nonenzymatic reaction between the glucose and a free amino group of a protein.

Supplemental Readings and References D. G. Dyer, J. K. Blackledge, S. R. Thorpe, and J. W. Baynes: Formation of pentosidine during nonenzymatic browning of proteins by glucose. Journal of Biological Chemistry 266, 11654 (1991). M. E. Gurney, E B. Curring, E Zhai, and others: Benefit of vitamin E, riluzole, and gabapentin in a transgenic model of familial amyotrophic lateral sclerosis. Annals of Neurology 39, 147 (1996). R. G. Hankard, M. W. Haymond, and D. Darmaun: Role of glutamine as a precursor in fasting humans. Diabetes 46, 1535 (1997). S. J. Kuhl and H. Rosen: Nitric oxide and septic shock. West Journal of Medicine 168, 176 (1998). L. Lacomblez, G. Bensimon, E N. Leigh, et al.: Dose-ranging study of riluzole in amyotrophic lateral sclerosis. Lancet 347, 1425 (1996). J. Loscalzo and G. Welch: Nitric oxide and its role in the cardiovascular system. Progress in Cardiovascular Diseases 38, 87 (1995).

R. G. Miller, D. Moore, L. A. Young, et al.: Placebo-controlled trial of gabapentin in patients with amyotrophic lateral sclerosis. Neurology 47, 1383 (1996). R. Safirstein, L. Andrade, and J. M. Vieira: Acetylcysteine and nephrotoxic effects of radiographic contrast agents--a new use for an old drug. The New England Journal of Medicine 343, 210 (2000). M. Tepel, M. van der Giet, C. Schwarzeld, and others: Prevention of radiographic-contrast-agent-induced reductions in renal function by acetylcysteine. The New England Journal of Medicine 343, 180 (2OOO). H. Vlassara: Recent progress on the biological and clinical significance of advanced glycosylation end products. J. of Laboratory and Clinical Medicine, 124, 19 (1994). T. R. Ziegler, K. Benfele, R. J. Smith, et al.: Safety and metabolic effects of 1-glutamine administration in humans. Journal of Parenteral and Enteral Nutrition 14, 1375 (1990).

CHAPTER

3

Protein Isolation and Determination of Amino Acid Sequence

To understand the structure and function of a protein, one must know the number and kinds of amino acids present in the protein and their order (called sequence or primary structure). This information is necessary to understand the effects of mutations, mechanisms of enzyme-catalyzed reactions, and chemical synthesis of species-specific peptides that may eliminate undesirable hypersensitivity reactions. Studies of amino acid sequences in proteins have aided in the understanding of the evolutionary development of living systems. Another application of amino acid sequence determination is in recombinant DNA technology. A desired DNA coding for a given polypeptide can be constructed from knowledge of the precise sequence of that polypeptide (Chapter 23). Through the use of available technologies of sequence determination and peptide synthesis, peptide fragments have been produced that are identical to part of large proteins present on the surface of a virus or other pathogen. In the appropriate host, such peptides elicit antibody production that is effective against the active pathogen and are potentially useful in the development of synthetic vaccines (Chapter 35). Viruses for which immunogenic peptides are being synthesized and clinically applied are hepatitis B, influenza, rabies, mouse leukemia, and hoof-and-mouth disease. Bovine insulin (M.W. 5,700) was the first protein to be completely sequenced (Sanger, 1955). Sanger was also the first to deduce the base sequence of a DNA molecule obtained

from phage 4~X 174 (a bacterial virus; see Chapter 23). Bovine insulin comprises two peptide chains of 21 and 30 amino acids each, linked by two interchain disulfide bonds. In 1960, Hirs, Moore, Stein, and Anfinsen described the first primary structure of the enzyme ribonuclease (M.W. 13,700), which has a single peptide chain of 124 amino acid residues and four intrachain disulfide bonds. These investigators established many of the techniques still used in sequence analysis, such as the use of ion exchange resins for separation of peptides and amino acids.

3.1 Quantitative Determination of Proteins UV absorption at 280 nm is an inaccurate method of protein determination because proteins have different amounts of tyrosine and tryptophan residues, and nucleic acids also absorb at 280 nm. In the biuret test, the sample is treated with an alkaline copper sulfate reagent that produces a violet color and requires a peptide with at least two peptide bonds. The violet color is produced through formation of a coordination complex (between peptide nitrogen atoms and cupric ion) that is analogous to the structure of the complex of biuret with cupric ion, as shown below:

35

36

CHAPTER3 Protein Isolation and Determination of Amino Acid Sequence NH2

I

O H

C=O

2

I

NH

, , . . ./ %/ ~\N

+

I

C--O

I

HH

NH2 Biuret

Cu 2+

,NH:

C--N"

J/J Un

H,O

\WL-d\ /

_..'% m

+.

H31~I -

l

A

il

B

1 I

1

C

COO -

I

;NH

"N--C MI-I m O

% .I \,,\\

Violet complex

In the Folin-Ciocalteu reaction, the protein reacts with the phosphomolybdotungstic acid reagent to give a blue color through reaction with tyrosyl residues. The sensitivity of this method depends on the amino acid composition of proteins in each sample. Protein content, particularly in urine or cerebrospinal fluid, may also be estimated by methods based on precipitation using sulfosalicylic acid (an anionic protein precipitant) or heat. The turbidity, which is a measure of protein concentration, can be quantitated by spectrophotometric absorbance methods or light scattering analysis. Absorbance of a hydrophobic indicator dye that binds to protein and changes color is also used. Specific proteins in biological fluids can be estimated by more discriminating methods, such as electrophoresis, specific binding techniques, or immunochemical methods.

Assume that cleavage of the above polypeptide with a specific cleaving agent yields four peptides, A, B, C, and D. These peptides are separated and sequenced but it is yet not known whether the correct order is ABCD or ACBD. A second hydrolytic procedure gives rise to three peptides: E, F, and G.

----E +

+---F .... i

A-B break

i B-C break

-+ I

Gl C-D break

--

_ COO

-

By comparison of the amino acid sequence of fragments E and G, the terminal fragments A and D can be matched with their respective adjacent peptides, B and C. Alternatively, the order of the peptides B and C can be deduced from the overlapping sequence in E Purification of a protein is indispensable for establishing its amino acid sequence. Mixtures of proteins will yield mixtures of peptides and ambiguous amino acid positions; a unique amino acid sequence can be obtained only from a pure protein. Protein purification techniques exploit differences in size, shape, charge, solubility, and specific binding affinity of the proteins. The optimal combination of techniques is usually reached by trial and error.

3.2 Determination of Primary Structure The determination of the primary structure of a protein consists of the following steps: 1. Obtain a pure protein. 2. Determine the amino acid composition and molecular weight of the pure protein. From amino acid composition and molecular weight data, calculate the number of residues of each amino acid present per protein molecule to the nearest whole number. 3. Reduce disulfide bonds to sulfhydryl groups. 4. Determine amino terminal (N-terminal) and carboxy terminal (C-terminal) amino acids. (A unique residue for each terminus suggests that the native protein contains only one peptide chain.) 5. Fragment aliquots of the polypeptide by enzymatic or chemical hydrolysis and separate the peptide mixtures into individual fragments. (This process will yield overlapping sets of smaller peptides.) 6. Sequence each fragment and, by analyzing the overlapping parts, assemble the sequence of the original protein. The logic for arranging overlapping peptides is as follows:

3.3 Separation of Proteins Proteins are separated on the basis of differences in their size, shape, charge, solubility, and binding affinity for other molecules. Most separations begin with proteins in solution. However, when proteins are an integral part of an organelle's structure, it is first necessary to extract them from membranous elements. Chemicals used to extract proteins from membranous particles include dissociating agents (e.g., urea and mercaptoethanol), chelating agents (e.g., ethylenediaminetetraacetic acid [EDTA]), and organic detergents (e.g., sodium deoxycholate, sodium lauryl sulfate, and Triton X-100). Since the total quantity (or activity) of a protein in a tissue sample is difficult to determine, the original (100%) quantity of protein at the start of a purification is usually based on measurements made on an aliquot of the initial homogenate. Each step in a purification process should remove extraneous protein and retain most of the protein of interest. A pure protein preparation is operationally defined as one that maintains a high activity per gram of protein following several purification steps, i.e., the optimum

SECTION3.3 Separation of Proteins

specific activity of the protein. Alternatively, a pure protein cannot be further subdivided by the methods described below, e.g., chromatography or electrophoresis. The initial purification steps usually separate proteins according to general classification, e.g., fibrous (insoluble) or globular (soluble). The fibrous and globular designations are related to shape and solubility. Globular proteins are spherical or ellipsoidal and make up the majority of known proteins. Fibrous proteins contain one or more polypeptide chains. Their molecules are elongated and asymmetrical with lengths that may be many times their diameters. Lateral cross-linking between adjacent polypeptides, by a variety of types of chemical bonding, confers mechanical strength and water insolubility to fibrous proteins; consequently, they are found in connective, elastic, and contractile tissues as well as in hair and skin. An initial aqueous extraction procedure tends to partition globular proteins into the soluble fraction and fibrous proteins into the "insoluble pellet" remaining after centrifugation.

Separation by Molecular Size Protein can be separated on the basis of their size by g e l f i l t r a t i o n , and m e m b r a n e f i l t r a t i o n . Small molecules (e.g., NaC1, amino acids, and sucrose) originally present or added during the separation of organelles can be removed by dialysis through a semipermeable membrane. Dialysis membranes are prepared from cellophane or collodion and contain pores that permit passage of solute molecules whose molecular weight is less than ~5000. Thus, proteins of high molecular weight are retained within a dialysis bag, whereas low-molecularweight solutes diffuse through the pores into the fluid (dialysate) outside of the bag. Complete removal of lowmolecular-weight solutes requires repeated changes of dialysate. Rapid removal of low-molecular-weight solutes and simultaneous concentration of the protein solution can be accomplished by applying pressure to the dialysis solution (or vacuum to the dialysate). Such a process is known as u l t r a f i l t r a t i o n (Figure 3-1). The principles of membrane filtration are the same as those of dialysis except that synthetic membranes with specified pore sizes are used. In gel filtration (or molecular sieving), a column is packed with hydrated, insoluble gel particles with known pore sizes; the protein solution is passed through the column and the effluent solution is collected in fractions that will contain solutes of different sizes (Figure 3-2). The volume of the column is essentially divided into two phases: the gel phase (within the pores) and the solvent phase (outside the gel particles). As a solution migrates in the column, the solute molecules that can penetrate the pores of the gel are distributed both within the gel and outside it. The particles that are larger than the pore size are excluded

37 Hydrodynamic force [

Solvent Large,

--impermeable molecules :::!~ ::!:!::ii:::::~i:::ii~.:iii'~.:i

Small,

. . . . . . . . ............... 9 ~i:!~!.::~!;::~:@: Semi. ::i~.?~-:..~)# :.~:L~"~ - - permeable :i::::i:: i iI :i:i:i:iil ::i!::i:::iiiiiii: membrane

6

i . 9.....

6

..:. ~....."

!:i#iiii!iiiii!iiiii',iiiii:i :;::!::;::i~i!!Oi::~iii!!O::iiiiiii::~:~:#l

J

.....::::i:!.........!~!~~::~:::~:i i~:~::~i........ :~

F I G U R E 3-1 Separation and concentration of high-molecular-weight solutes from low-molecular-weight solutes by application of hydrodynamic force over the solution above a semipermeable membrane (ultrafiltration).

dialysis,

from the gel phase flow through the column more rapidly and appear first in the effluent. Commonly used gel particles are inert cross-linked dextrans or agarose, which are commercially available with a wide range of exclusion limits. Molecular sieving is effective in the purification of macromolecules and if the column has been calibrated by elution of solutes of known molecular weight, it can also be used in the estimation of molecular weights of proteins or other solutes.

Separation by Chromatography Chromatographic separations of proteins are based upon the differential partitioning of solute molecules due to their differences in affinity between a moving solvent phase and a fixed or supportive phase. In gas chromatography, the mobile phase is a gas, whereas in liquid chromatography it is a liquid. Gas chromatography is not useful in protein purification because proteins cannot be converted to gases without decomposition. Liquid chromatography of proteins is performed on a variety of mechanically different stationary phases, e.g., paper, finely divided particles coated onto a glass or a plastic surface (thin-layer chromatography), or beads packed in a column. Many chemically different stationary phases are used in liquid chromatography of proteins. Ion exchange c h r o matography uses an ion exchange resin, and the proteins are eluted with buffer solutions differing in ionic strength

38

CHAPTER 3

OOOo 9_oJ Sam~,eI O gO0 O O'WOq~' 9 9 O 9

zone

DOg O e,_,_[ :ii !i !i i !~;ii!Qii !i i i !I!i~i~:

Protein Isolation and Determination of Amino Acid Sequence

,..,0 0

ilJi!iiiiiiiiiiiiiiilQiiiiii!iiiii~il il

ii!i!ii!iiii!! !ii!i] i

.ii :.:,:.,~::?.!',!',!i....... i i i ,.,,i, i !i',i':~!~:..[

i~~~i 9 !!ii i!~i~lid ', G:~!i ::.i ",i!i i::~i%!i'.':....... ~:::.'.::

Ge,

,~i~.:!::,::Gi;~.!,,ii!!ii:,..:,:.,::.~ pa,~de~ ,

:~i!i;i:,ii~:~:~:!:iiiiiil;i!iiii',i;iiiil :~i!ii~:::Qi:iUIiI',Q .

.

.

.

.

The pores of the gel particle exclude the larger solutes but admit smaller ones. Individual solute molecules are represented by the open circles.

......................................

iii!i; 9~;i::::;~iiii:ii!i!~i~!i',~i:!iii'i i ,!

iii!iiiiiiiii;iiii!iiiiiiii !;i !iii!ill ii!!iiiii;i!ii!ii iii!iiiiiiiii!i i

!ii!i!!i!ii!iiii!i!i! F I G U R E 3-2 Gel filtration for separation of solutes by size. Solute molecules smaller than the diameter of the pores of the gel particles enter them and are retained for a longer time. Larger solute molecules cannot penetrate the pores of the gel particles and are eluted off the column first.

and pH. The resins are inert polymers to which ionizable groups have been attached; resins with negative charges are cation exchangers, and those with positive charges are anion exchangers. Two ion exchange resins frequently used in protein purification are diethylaminoethylcellulose (DEAE-cellulose, an anion exchanger) and carboxymethylcellulose (CM-cellulose, a cation exchanger). A protein with a net positive charge at a given pH will combine with the negative groups of a cation exchange resin, and its flow will be retarded; a protein with a net negative charge will migrate through the cation exchange resin unimpeded. Cationic proteins in a mixture going through the column will compete with one another for binding to the negatively charged groups of the resin. The relative migration rates of different molecules depend on three factors: their individual affinities for the charged sites on

the resin, the degree of ionization of the functional groups attached to the resin, and the chemical properties and concentrations of competing low-molecular-weight ions, e.g., potassium and sodium. For example, NaC1 at the appropriate concentration can displace a cationic protein through competition between sodium ions (Na+) and the positively charged groups of the protein for the negatively charged sites on the resin (Figure 3-3). Changing the pH of the eluting buffer can also bring about desorption of proteins from the resin through neutralization of the ionizable R-groups of amino acid residues. Decreasing pH will elute proteins in the order of their decreasing is 9 point values during anion exchange chromatography. Affinity chromatography takes advantage of specific affinities between protein molecules and analogues of biological molecules that are covalently bound to the column

SECTION 3.3

39

S e p a r a t i o n of Proteins Adsorption

(':h

Affinity Tag Chromatography ~.i -. '~. .~J~: ~ . ~

F I G U R E 4-2 Geometry of a peptide (amide) linkage. For the peptide bond, bond angles and bond lengths indicate that carbon-nitrogen bonds have a significant amount of double-bond character and that the C, O, N, and H atoms all lie in the same plane. The 05 and ~ refer to rotations about the single bonds connecting the e-carbon with the o~-nitrogen and the o~-carbonyl carbon, respectively.

The folding of polypeptide chains into ordered structures maintained by repetitive hydrogen bonding is called secondary structure. The chemical nature and structures of proteins were first described by Linus Pauling and Robert Corey who used both fundamental chemical principles and experimental observations to elucidate the secondary structures. The most common types of secondary structure are the right-handed c~-helix, parallel and antiparallel ~-pleated sheets, and ~-turns. The absence of repetitive hydrogen-bonded regions (sometimes erroneously called "random coil") may also be part of secondary structure. A protein may possess predominantly one kind of secondary structure (a-keratin of hair and fibroin of silk contain

~

CHAPTER4 Three-Dimensional Structure of Proteins

mostly u-helix and/3-pleated sheet, respectively), or a protein may have more than one kind (hemoglobin has both or-helical and non-hydrogen-bonded regions). Globularproteins usually have mixed and fibrous proteins have predominantly one kind of secondary structure. c~-Helix The rod-shaped right-handed or-helix, one of the most common secondary structures found in naturally occurring proteins, consists of L-or-amino acids (Figure 4-4). In the right-handed c~-helix the helix turns counterclockwise (C-terminal to N-terminal) and in the left-handed it turns clockwise. The left-handed c~-helix is less stable than a right-handed or-helix because its carbonyl groups and the R-groups are sterically hindered. The helical structure is stabilized by intrachain hydrogen bonds involving each - N H a n d - C O group of every peptide bond. These hydrogen bonds are parallel to the axis of the helix and form between the amido proton of the first residue and the carbonyl oxygen of the fourth residue, and so on, producing 3.6 amino acid residues per turn of the helix. The rise per residue is 0.15 nm and the length of one turn is 0.54 nm (Figure 4-5). In some proteins, or-helices contribute significantly to the secondary structure (e.g., ct-keratin, myoglobin, and hemoglobin), whereas in others, their contribution may

F I G U R E 4-4 Hydrogen bonds in the a-helix. (a) Each peptide group forms a hydrogen bond with the fourth peptide group in each direction along the amino acid chain. (b) Coiling of an amino acid chain brings peptide groups into juxtaposition so that the hydrogen bonds shown in (a) can form. The multiple hydrogen bonds (indicated by the three dots) stabilize the helical configuration.

0.51 nm

26 ~

m1 . . . . 0.54-nm pitch (3.6 residues)

...... l 0.15 nm

Rise per amino acidresidue

f F I G U R E 4-5 Average dimensions of an a-helix. Only the atoms of the a-carbon, the carbonyl carbon and the nitrogen of the peptide bonds are shown. The rise per residue and the length of one turn are 0.15 and 0.54 nm corresponding to minor and major periodicity, respectively.

be small (e.g., chymotrypsin and cytochrome c) or absent (e.g., collagen and elastin). Whether a polypeptide segment forms an c~-helix depends on the particular R-groups of the amino acid residues. Destabilization of an or-helix may occur for a variety of reasons: electrostatic repulsion between similarly charged R-groups (Asp, Glu, His, Lys, Arg); steric interactions due to bulky substitutions on the/3-carbons of neighboring residues (Ile, Thr); and formation of side-chain hydrogen or ionic bonds. Glycine residues can be arranged in an a-helix; however, the preferred and more stable conformation for a glycine-rich polypeptide is the/3-pleated sheet because the R-group of glycine (-H) is small and gives rise to a large degree of rotational freedom around the a-carbon of this amino acid. Prolyl and hydroxyprolyl residues usually create a bend in an a-helix because their a-nitrogen atoms are located in rigid ring structures that cannot accommodate the helical bonding angles. Moreover, they do not have an amido hydrogen and therefore can form neither the necessary hydrogen bond nor the usual planar peptide bond. However, some proteins such as rhodopsin do contain proline residues embedded in a-helical segments. In some proteins, the a-helices twist around each other to form rope-like structures (coiled coils) to give rise to a supersecondary structure. Examples of such proteins are the ot-keratins, which are major protein components of

SECTION4.3 Secondary Structure

/c-.

SS

/c-.

/c-.

/

99 9 ..

9

.9 9

R--Ck/ 9

. H ~.

9

7%0 9 .H./%a \ .

/C%o H~N\

.

CmR

C~R

/

.-c\ 9

9.

N/'H"

"

R--C\/

"

R--C\/

"" . H . / ' O .

N / H "

R--C(

/~O.

.
0).

favorable and that the reaction can occur spontaneously. If a chemical reaction has a positive A G value (as in Figure 5-2), the reaction is thermodynamically unfavorable and can occur only with the assistance of an external source of energy (e.g., coupling of an unfavorable reaction with a favorable one); such a reaction is termed

endergonic. An enzyme accelerates a chemical reaction by decreasing AG', which is usually accomplished by providing an alternate pathway for the reaction (Figure 5-3). While A G' is decreased by the enzyme, the A G value (i.e., free-energy difference between reactant and product) is unchanged. In enzyme catalysis, the reactant is termed substrate. The ability of an enzyme to decrease the AG' value is illustrated by hydrogen peroxide decomposition (2H202 --+ 2H20 -k- O2). In the absence of catalase, AG' for the reaction is 18 kcal. mo1-1 (79.3 kJ. mol-1), whereas in the presence of catalase, AG' is 7 kcal. mo1-1 (29.2 kJ. mo1-1). A calorie is the amount of heat required to raise the temperature of 1 g of water by I~ (from 14.5~ to 15.5~ One kilocalorie (kcal) is 1000 calories. A joule (J) is the amount of energy required to apply a force of 1 N (newton) over a distance of 1 m. One kilojoule (kJ) is 1000 J (1 kcal = 4.184 kJ). For a reversible chemical reaction, the dependence of A G on the temperature and concentration of reactants and products is given by the expression AG = AG ~ + RT In

[products] [reactants]

(5.1)

F I G U R E 5-3 Energy diagram showing the reduction in free energy of activation ( A G ' ) in an enzyme-catalyzed reaction. The free-energy difference between reactant and product (AG) is unchanged.

where AG ~ is the standard free-energy change in calories per mole of reactant consumed; R is the gas constant (1.987 x 10 .3 kcal. mo1-1, deg-1); T is the absolute temperature in degrees Kelvin (-- ~ + 273); In is the natural logarithm (base e); and [products] and [reactants] are the corresponding molar concentrations (for solvents and solutes) or pressures in atmospheres (for gases). Since at equilibrium A G = 0, this equation can be written [products] AG ~ -- - R T i n Keq where Keq = [reactants]" Note that the standard free-energy change is proportional to the absolute temperature and the equilibrium constant, Keq. The equilibrium constant depends only on the temperature of the system. This equation provides a way of determining AG ~ from equilibrium concentration data. Another commonly used form is AG ~ -- - R T (ln 10) (log Keq) -- - 2 . 3 0 3 R T log Keq (5.2) where log means logarithm (base 10). The standard state for AG ~ is one in which the temperature is 25~ all gases are present at pressures of 1 atm, all liquids and solids are pure (i.e., not mixtures), and all solutes have unit activity. Activity is similar to concentration but takes into account interactions of solute molecules with each other and with the solvent. Activities are difficult

CHAPTER5 Thermodynamics, Chemical Kinetics, and Energy Metabolism

70

to measure, however, and molar concentrations are commonly used in their place. Since concentration and activity are almost equal in dilute solutions and many biological reactions occur at low concentrations, this approximation is valid. A physical interpretation of A G ~ can be obtained by noting that if [products] / [reactants] -- 1, then A G = A G ~. A G ~ is the amount of useful work that can be obtained by conversion of 1 mol of each reactant in its standard state to 1 mol of each product in its standard state. The standard free-energy change for a reaction is also useful for predicting whether a reaction will occur under standard conditions. If AG ~ < 0, the reaction will occur spontaneously provided standard conditions prevail; if A G ~ > 0, the reaction will not occur by itself. It is A G, however, that determines whether or not a reaction will occur under conditions different from the standard state, such as those existing within a cell, for two reasons. 1. Most solutes are present at fairly low concentrations (< 0.1 M) and water is present at high concentration (55.6 M for pure water); the water concentration is thus assumed to be constant. If water enters into a reaction, it is incorporated into Keq; if Keq is determined at pH 7.0, it is indicated by K'eq" 2. If H + or O H - participates in a reaction, its concentration influences AG ~. Since most biological reactions occur in systems buffered to pH "- 7, it is useful to define AG ~ as the value of AG ~ measured at pH -- 7.0, i.e., where [H +] -- 10 -7 M. AG ~ values can be compared to each other but not to values of AG ~ Therefore, for biochemical systems, Equation (5.2) can be written as !

AG ~ -- - 2 . 3 0 3 R T log Keq

(5.3)

Substituting for R -- 1.987 x 10 -3 kcal. mol - l . deg-l (8.314 x 10 .3 kJ. mol-1 d e g - l ) and T -- 298 K (i.e., 273 + 25~ .

!

~

'

.

[in kcal/mol]AG ~ -- - 1.36 log Keq, t __ 0-AG~ o r Keq 1

[in kcal/mol]AG ~ -- - 5 . 6 9 log Keq, o r g e 'q

m

10 -AG~

(5.4)

From Equation (5.4), at 25~ each - 1.36 kcal/mol value of A G ~ corresponds to a factor of 10 in the equilibrium constant in favor of product formation (Table 5-1). As Keq increases, the value for A G ~ decreases and the tendency for reactions to occur increases spontaneously. Thus, we I can calculate A G ~ by knowing Keq and the temperature at

T A B L E 5-1

Relationship between Standard Free Energy Change (AG ~ and Equilibrium Constant (Keq ) at p H 7.0 and 25~ * AG O"

K' eq

kcal/mol

kJ/mol

10 -6 10 -5 10 -4 10 -3 10 -2 10 - i

8.18 6.82 5.46 4.09 2.73 1.36

1

0

0

-1.36 -2.73 -4.09 -5.46 -6.82 - 8.18

-5.69 -11.42 -17.11 -22.84 -28.53 -34.22

10 102 103 104 105 106 ,

34.22 28.53 22.84 17.11 11.42 5.69

1.36 K , eq = 1 0 - A G 0 ' when AG O' is expressed in kcal/mol at 25~ K,eq = 10_AGO"5.69 when AG o" is expressed in kJ/mol at 25~

I which Keq is determined. In living systems, chemical reactions rarely reach complete equilibrium states but do attain near-equilibrium and nonequilibrium conditions. These conditions are maintained by utilizing energy from the external source. In a metabolic pathway, one of the nonequilibrium reactions usually becomes the rate-determining step of that pathway and its regulation provides directionality to the pathway. In a reaction sequence

A ~-~-B ~- C ~- . . . the reaction A --+ B can be prevented from reaching near-equilibrium by removing B (converting it to C) as fast as it can be made from A. However, the conversion of B to C may attain near-equilibrium. The nearequilibrium reactions are reversible and allow for reversal of the biochemical pathway. All reactions in the body are interrelated and the system as a whole is in a steady-state condition, with individual reactions operating in either near- or nonequilibrium conditions. A change in concentration of any component (product of one reaction used as reactant in another reaction) shifts the concentration of all other components linked to it by means of a sequence of chemical reactions, resulting in the attainment of a new steady state. The value of A G for a nonequilibrium system can be determined from A G ~ , and the actual concentrations of

SECTION 5.2

Thermodynamics

71

products and reactants in the cell can be determined by use of the common logarithm analogue of Equation (5.1)" AG -- AG ~ + 2.303R T log [products]. [reactants] If A G is negative, then u n d e r c e l l u l a r c o n d i t i o n s the reaction proceeds to form products. The AG ~ value for a given chemical reaction may be positive suggesting immediately that the reaction cannot occur (without external assistance), yet the AG value may be negative indicating that the reaction can occur under the prevailing concentrations of reactant and products. The reaction of glycolysis, catalyzed by the enzyme aldolase (Chapter 13), furnishes a good illustration. Fructose 1,6-bisphosphate ~- glyceraldehyde 3-phosphate + dihydroxyacetone phosphate The Ke~qfor this reaction at 25~ is 6.7 x 10 -5 M. We can calculate AG ~ from Equation (5.3)"

!

AG ~ -- - 2 . 3 0 3 R T log Keq = - 2 . 3 0 3 x 1.98 x 10 -3 x 298 x log (6.7 x 10 -5) -- +5.67 kcal/mol (or + 23.72 kJ/mol). The positive value for AG ~ indicates that the reaction is not favored in the forward direction but can proceed in the reverse direction. Now we calculate the A G value using the concentration of 50/zmol/L (or 50 x 10 -6 M) for both reactant and product, which is close to their physiological concentrations: AG-

AG ~ + 2.303RT

glyceraldehyde] [ dihydroxyacetone] 3-phosphate I I phosphate j • log i

~

of reactants and products (formation of fructose 1,6bisphosphate from glyceraldehyde 3-phosphate and dihydroxyacetone phosphate during gluconeogenesis). The same enzyme catalyzes both the forward and the reverse reactions. The concept of A G is further clarified by an example. Consider a self-operating heat engine that functions by taking in an agent (e.g., steam) at temperature T1 and releasing it at T2 (T1 > T2). The heat extracted (Q) is converted as efficiently as possible into useful work. If W represents the maximum useful work available, W-

rl

= - 0 . 1 8 kcal/mol(or - 0.75 kJ/mol). The negative value of A G suggests that under physiological conditions the forward reaction is favored, despite the fact that AG ~ is positive. Thus, under physiological conditions A G values predict better than AG ~ values whether a reaction can occur spontaneously. Similarly, whereas under certain conditions of reactants and products the conversion of reactant to product is favored (e.g., the conversion of fructose 1,6-bisphosphate to glyceraldehyde 3-phosphate and dihydroxyacetone phosphate during glycolysis), the direction of this reaction can be reversed when appropriate changes occur in the concentrations

T2

-e-eg

where 7'i and Te are absolute (Kelvin) temperatures. Unless T2 = 0 or T1 = oo, the useful work is always less than the total energy supplied by a factor of Q ( T 2 / T 1 ) = (Q / T1) T2. The ratio Q / T1 is the entropy (S) of the system, and the amount of energy unavailable for useful work (T2 S) is that which is lost in the process of energy transfer; it can be thought of as the amount of randomness or disorder introduced into the system during the transfer. For chemical systems, G can be defined by the equation G = H - TS. Here, G (free energy) is analogous to W, the maximum useful work available; H (enthalpy) is analogous to Q, the heat content of the system at constant pressure; S (entropy) is analogous to ( Q / T 1 ) , the wasted heat energy; and T is again the absolute temperature. This relationship G = H - T S is more commonly used to describe c h a n g e s in these quantities. If a system goes from state I (with G = GI, H = HI, S = SI) to state II (G = GII, H -- HII, S -- Sn) at constant temperature,

i]~55b~i

= +5.67 kcal/mol + 2.303 • 1.98 • 10 -3 • 298 [(50 • 10 -6) X (50 X 10-6)] • log (50 • 10 -6) -- 5.67 kcal/mol - 5.85 kcal/mol

Q (T1 - T2)

GII - GI = (HII - HI) - T (Sn - SI) or, simply, A G-

AH-

T AS.

Since entropy (S) is a measure of disorder (randomness), it is the energy that is unavailable for useful work. Entropy values of denatured molecules are high relative to those of native structures (as in protein denaturation). In a living system, entropy is kept at a minimum by utilization of free energy (G) from outside and by increase in entropy of the surroundings. Enthalpy (H) is related to the internal energy of a system by the following equation, where E is the internal energy, P the external pressure on the system, and V the volume of the system. In terms of changes between states, the equation becomes AH

-- A E + A ( P V ) -

AE + PAV

+ VAP.

~

CHAPTER5 Thermodynamics, Chemical Kinetics, and Energy Metabolism

At constant pressure, A P - - 0 and A H -- A E 4- P A V. If, in addition, the volume is constant, then A V -- 0 and A H - A E . In many biological reactions, A V - A P = 0. Consequently, for a change of state, the maximum amount of energy that can be released as heat ( A H ) is equal to A E , the change in the internal energy of the system. A H can be measured by burning the particular compound in a bomb calorimeter at constant pressure. The heat released is measured as the rise in temperature of a large water bath surrounding the combustion chamber. The values so obtained indicate the total energy available in a compound when it is completely oxidized. Factors that usually contribute to A H under these conditions are the heat of fusion (melting), the heat of vaporization (boiling), and the heat of combustion (bond making and breaking). The first two factors are important, for example, if at the standard temperature (25~ a solid is combusted to a combination of liquids and gases, as in the examples below. Just as with A G, A H for a process is equal to - A H for the reverse process: A H (liquid--+ g a s ) - - A H

(gas--~ liquid)

A H (liquid --+ solid) -- - A H (solid --+ liquid)

Nemst equation:

Eh = E ~ 4-

2.303 R T nF

log

(electron acceptor) (electron donor)

where Eh = observed potential, E ~ = standard redox potential, R = gas constant (8.31J deg -1. mol), T = temperature in Kelvin, n = number of electrons being transferred, and F = the Faraday constant (96,487 J/V). It is evident from the Nernst equation that Eh = E ~ when [electron acceptor] -- [electron donor]. Thus, the E ~ value represents the midpoint in a redox reaction, which is analogous to the measurement of the pK' value of a weak acid, HA, when [A-] = [HA] (see discussion of the HendersonHasselbalch equation in Chapter 1). In this manner, E ~ values for many redox reactions of biochemical importance have been determined (Chapter 14). A half-reaction is a reaction in which electrons or hydride ions are written explicitly. Values of E ~ and E ~ are tabulated with the half-reactions for which they are measured. For example, NADH --+ NAD + 4- H -

E ~ = -0.32 V

and 1 H20---+ ~ 0 2 4- 2H + + 2e-

E ~ -- - 0 . 8 1 6 V

A H (making a bond) - - A H (breaking the same bond). A H values for various food substances and their application to human energy metabolism are discussed later in this chapter. A number of processes in living systems result in the transfer of electrons (oxidation and reduction) and can be understood in thermodynamic terms. Oxidation is the loss of electrons or hydride ( H - ) ions (but not hydrogen [H +] ions) by a molecule, atom, or ion. Reduction is the gain of electrons or hydride ( H - ) ions by a molecule, atom, or ion. Transfer of one hydride ion results in the transfer of two electrons. The amount of work required to add or remove electrons is called the electromotive potential or force (ernf ) and is designated E. It is measured in volts (joules per coulomb, where a coulomb is a unit of electric charge or a quantity of electrons). The standard emf, E ~ (or, at pH 7, E~ is the emf measured when the temperature is 25~ and the materials being oxidized or reduced are present at concentrations of 1.0 M. In biological systems, E ~ is most commonly used. 1 The emf measured for an oxidation-reduction (redox) reaction under nonstandard conditions, Eh, is mathematically related to the emf measured for the same reaction under standard conditions, E ~ through the !

l Some texts use the notation E0 for the standard oxidation-reduction potential.

are half-reactions because they show electrons (e-) or hydride ions (H-). NADH and NAD + are abbreviations for reduced and oxidized forms of nicotinamide adenine dinucleotide (whose structure is given in Chapter 6 and whose metabolic functions are discussed elsewhere). Alternatively, these half-reactions can be written as NAD + 4 - H - ~ N A D H E~ 1 2e- + 2H + + ~ 0 2 ~ H20 E ~ -- +0.816 V. Reversing the direction of a reaction changes the sign of the potential charge (just as with AG and AH). The oxidation potential is a measure of the ease of removal of electrons from a material compared to the ease of removal of electrons from hydrogen in the half-reaction: H2 ~ 2H + 4- 2e-

E ~ -- 0.00 V.

E ~ in this reaction is defined as zero when H2 gas is at 1.0 atm, [H +] is 1.0 M (i.e., pH 0), and the temperature is 25~ thus fixing the scale of E ~ value for other reactions. If it is easier to remove electrons from a material, E ~ will be negative; if it is harder, E ~ will be positive. E ~ for the hydrogen half-reaction is used as the zero point even when reference is made to E ~ values. Thus, H2 ~ 2H + 4- 2e-

E ~ ---=- 0 . 4 2 V.

It is easier to remove electrons from hydrogen (and produce H +) when [H + ] -- 10 -7 M than when [H +] - 1.0 M.

SECTION 5.3

Standard Free Energy of Hydrolysis of ATP

The choice of a negative sign for the standard emf to signify "easier to remove electrons" is arbitrary. 2 Since free electrons combine rapidly with whatever is at hand, half-reactions never occur by themselves. Something must accept electrons as fast as they are released. The substance releasing electrons (or H - ) is the reductant, or reducing agent (because it is oxidized), and the substance accepting electrons is the oxidant, or oxidizing agent (because it is reduced). Two half-reactions when combined give a redox reaction. When balanced, such reactions never show free (uncombined) electrons. For example, 1 H + + NADH + ~02 ~ NAD + + H20

A E ~ -- - 0 . 3 2 - (+0.816)-- - 1 . 1 3 6 V. A E ~ for this reaction is calculated accordlng to AE ~

A E ~ ( r e d u c t a n t ) - A E ~ (oxidant).

Under standard conditions (and pH -- 7.0), the reaction will occur as written if A E ~ < 0. Otherwise the reaction will proceed from right to left (the reverse of the way it is written). When A E ~ in volts is converted to calories per mole, the result is A G ~ for the redox reaction being considered. This conversion is accomplished by means of the equation AG~

nF

9A E ~

4.184

= (23.061)n. A E ~

where, as before, F is Faraday's constant -- 96,487 J/V; n is the number of electrons transferred per mole of material oxidized or reduced; A E ~ is E ~ (reductant) - E ~ (oxidant) in volts; and AG ~ is the standard free energy change in calories per mole of material oxidized or reduced (since there are 4.184 J/cal). In the case of the reaction just considered 1 H + + NADH + ~02 ---> NAD + + H20 AE ~

23.061n x ( - 1 . 1 3 6 ) = -26.197 x 2-

As in all reactions, the actual value of AG ~ depends on the reactant or product for which it is calculated. The significance of free-energy changes and redox reactions lies in the fact that life on earth depends on the redox reaction in which CO2 is reduced by H20 to yield (CH20), using sunlight as the energy source: Reduction (photosynthesis)

CO2 + H20 + energy
02 + (CH20)

Oxidation (respiration)

where (CH20) represents carbohydrates. The A E ~ for the forward reaction is about + 1.24 V, or AG ~ is about 114.3 kcal/(CH20). Thus, for glucose formation, AG ~ is +685.8 kcal. In plants, energy from the sun is absorbed by the chloroplasts of green plants, causing water to be oxidized and carbon dioxide reduced, producing oxygen and carbohydrate [represented by ( C H 2 0 ) m ] . In oxidation reactions, carbohydrate (or lipids) and oxygen are consumed, and energy, water, and carbon dioxide are released. The energy is captured primarily as adenosine triphosphate (ATP), which is used as an immediate source of energy in most cellular endergonic processes.

5.3 Standard Free Energy of Hydrolysis of ATP In the living organism, ATP functions as the most important energy intermediate, linking exergonic with endergonic processes. Exergonic processes (e.g., oxidation of glucose, glycogen, and lipids) are coupled to the formation of ATR and endergonic processes are coupled to the expenditure of ATP (e.g., biosynthesis, muscle contraction, active transport across membranes). ATP is called a "high-energy compound" because of its large negative free energy of hydrolysis

- 1 . 1 3 6 V,

n equals 4 gram-equivalents of electrons per mole of O2 or 2 gram-equivalents of electrons per mole of H20, NAD +, or NADH. Then, AG ~

73

-26.197n

-52.394

calories/mol of H20 or NAD + formed or NADH converted -- - 2 6 . 1 9 7 x 4 -- - 1 0 4 . 7 8 8 calories/mol of 02 transformed 2Although the International Union of Pure and Applied Chemistry recommends the opposite convention, the older convention is retained here because it is commonly encountered in medically and biologically related publications.

ATP 4- + H20 --+ ADP 3- + HPO 2- (or Pi) + H + AG~

kcal/mol ( - 3 0 . 5 kJ/mol).

The reason for the large release of free energy associated with hydrolysis of ATP is that the products of the reaction, ADP and Pi, are much more stable than ATR Several factors contribute to their increased stability: relief of electrostatic repulsion, resonance stabilization, and ionization. At pH 7.0, ATP has four closely spaced negative charges with a strong repulsion between these charges (Figure 5-4). Upon hydrolysis, the strain in the molecule due to electrostatic repulsion is relieved by formation of less negatively charged products (ADP s- and Pi2 - ). Furthermore, since these two products are also negatively charged, their

74

CHAPTER 5

Thermodynamics, Chemical Kinetics, and Energy Metabolism

NH,

Adenine I

II

II

O-

-

N

O-

O---P-tO--P-r-O--P-

oIII /

II

o

II

o

D-Ribose

I

OH

I

OH

Adenosine Adenosine monophosphate(AMP 2-) I

Adenosine diphosphate (ADP ~) FIGURE 5-4 Structural formula of adenosine triphosphate (ATP) at pH 7.0. The three phosphate groups are identified by Greek letters o~,/5, and y. The y- and E-phosphate groups are linked through phosphoanhydride bonds and their hydrolysis yields a large negative AG ~ whereas the u-phosphate linked by a phosphate ester bond has a much lower negative AG ~ In vivo most ATP is chelated to magnesium ions via two of the anionic oxygens (Mg 9ATP 2-).

tendency to approach each other to re-form ATP is minimized. The number of resonance forms of ADP and Pi exceeds those present in ATP so that the products are stabilized by a larger resonance energy. Ionization of HzPO 4 ~- HPO2-+ H + has a large negative AG ~ contributing the AG ~ for ATP hydrolysis. The free energy of hydrolysis of ATP is also affected by Mg 2+ and the physiological substrate is Mg 2+ ATP 9 4(or Mg. ATP 2-). The presence of magnesium ions favors ATP hydrolysis because both ADP and Pi have higher affinity (about six times) for Mg 2+ than ATE In vivo, the A G value for ATP hydrolysis is probably much higher than AG ~ owing to the prevailing intracellular concentration of the reactants and products (in erythrocytes, - 12.4 kcal/mol). In some energy-consuming reactions, ATP is hydrolyzed to AMP and pyrophosphate, with a value of free energy of hydrolysis comparable to that of ATP to ADP and Pi ATP4- + H20--+ AMP 2- + HP20~- + H + AG ~ = - 7 . 7 kcal/mol ( - 3 2 . 2 kJ/mol) In vivo, pyrophosphate is usually hydrolyzed to inorganic phosphate, which also has a large negative A G ~ HP2073- + H20--+ 2 H P O 2- + H § AG ~ = - 7 . 1 7 kcal/mol ( - 3 0 kJ/mol)

The pyrophosphate hydrolysis, although not coupled to any particular endergonic reaction, still ensures completion of the forward reaction or process (e.g., activation of fatty acids and amino acids, synthesis of nucleotides and polynucleotides). The hydrolysis of AMP to adenosine and Pi does not yield a high negative AG ~ because the phosphate is linked in a normal ester bond, as opposed to the other two linkages (fl and y), which are phosphoanhydride bonds (Figure 5-4). The AG ~ values of hydrolysis of other nucleoside triphosphates (e.g., UTP, CTP, GTP) are similar to that of ATP and they are utilized in the biosynthesis of carbohydrates, lipids, and proteins (discussed elsewhere). In addition to undergoing hydrolysis, ATP may act as a donor of phosphoryl (e.g., in the formation of glucose 6-phosphate; see Chapter 13), pyrophosphoryl (e.g., in the formation of phosphoribosylpyrophosphate; see Chapter 27), adenylyl (e.g., in the adenylylation of glutamine synthetase; see Chapter 17), or adenosyl groups (in the formation of S-adenosylmethionine; see Chapter 17). Since ATP synthesis from ADP and Pi is energydependent, it requires compounds or processes that yield larger negative AG ~ values than ATP hydrolysis does. Some of these compounds are organic phosphates (Table 5-2). Other high-energy compounds are thioesters,

75

SECTION5.4 Chemical Kinetics TABLE

t2 is the slope,

5-2

Standard Free Energy o f Hydrolysis (AG O") o f Some OrganophosphaCes A G O'

Compound / Hydrolyzed

kcal/mol (kJ/mol)

Product

Phosphoenolpyruvate --->Pyruvate 1,3-Bisphosphoglycerate ---> 3-Phosphoglycerate Phosphocreatine --->Creatine ATP --->ADP ATP --->AMP ADP ~ AMP Glucose 1-Phosphate --->Glucose Fructose 6-Phosphate --->Fructose AMP --->Adenosine Glucose 6-Phosphate --->Glucose Glycerol 3-Phosphate ~ Glycerol

v (-49.3) (-43.1) (-30.5) (-32.2) (-27.6) (-20.9) ( - 15.9) ( - 14.2) (-13.8) ( - 9.2)

[B]2 - [B]l

At

t2 - - t l

The instantaneous velocity, V, at some time t' (7~ to) is expressed as follows"

-14.8 (-61.9) -11.8 -10.3 -7.3 -7.7 -6.6 -5.0 -3.8 -3.4 -3.3 -2.2

A[B]

d[B (t)]

dt

evaluated at t'.

The equation d[B(t)]/dt is the first derivative of [B(t)] with respect to t. It is the limit (lim) of the slope A[B]/At as At --+ 0, or v--

lim ~ - -

A[B]

lim

At--,0

At~O At

derivatives,

and

sugar

5.4 Chemical Kinetics The study of chemical reaction rates is called chemical kinetics. Whereas thermodynamics deals with the relative energy states of reactants and products, kinetics deals with how fast a reaction occurs and with the chemical pathway (mechanism) it follows. The conversion A ~ B (where k is the rate constant) can be represented diagrammatically by the increase in concentration of product, [B], with respect to time (Figure 5-5). The average velocity, ~, for the period tl to

A[B (t)]

dt

.

Alternatively, one can consider the change in concentration of the reactants. Thus, d[B (t)]

d[A(t)] dt

dt

aminoacyl esters, sulfonium nucleoside diphosphates.

--

since the reactant concentration, [A], decreases at the same rate at which the product concentration, [B], increases. The expressions [B(t)] and [A(t)] imply a detailed knowledge of the way in which the concentrations of A and B vary with time and the way in which the reaction progresses. Since the preceding is generally not the case, [B] and [A] will be used instead of [B(t)] and [A(t)]. The rate of formation of [B] should increase if [A] increases. For the reaction n A ~ B, it is appropriate to write A[B] =

k[A]"

At

where k is again the rate constant, and n -- 0, 1, 2, . . . is the order of the reaction with respect to A. (The order is said to be zero, first, second, etc., with respect to A.) The rate constant expresses the proportionality between the rate of formation of B and the molar concentration of A and is characteristic of a particular reaction. The units of k depend on the order of the reaction. For zero order, they are moles liter -1 time -1 (time is frequently given in seconds). For first order, they are time -1 , and for second order, liters moles -1 time -1, etc. The units of k are whatever is needed for d[B]/dt to have the unit of moles liters -1 time -1. If n - 0, v - k; this means that in a zero order reaction, [B] changes at a constant rate independent of the concentration of reactants, which is especially important in enzyme kinetics. A plot of [B] versus t for such a reaction is a straight line. A somewhat more complicated example is A+B--~ C+D

F I G U R E 5-5 Time course of a chemical reaction.

d[C] v

--

dt

diD] =

dt

d[products] =

dt

=

k[A][B]

~6 This reaction is first order with respect to A, first order with respect to B, and second order overall. Most reactions are zero, first, or second order. The concept of reaction order can be related to the number of molecules that must collide simultaneously for the reaction to occur. In a first-order reaction, no collisions are required (since only one reactant molecule is involved). Every molecule with sufficient free energy to surmount the activation barrier may spontaneously convert to products. In a second-order reaction, not only must two molecules have enough free energy but they also must collide with each other for the products to form. A third-order reaction requires the simultaneous meeting of three molecules, a less likely event. Reactions of orders higher than second are seldom encountered in simple chemical conversions. Higher apparent orders are encountered, however, in some cases in which an overall rate equation is written for a process that proceeds in several steps via one or more intermediates. In such situations, the individual steps seldom involve a third- or higher order process. Therefore, for a chemical reaction consisting of multiple steps, the order of the reaction provides information about the overall rate equation but does not provide information regarding the number of atoms or molecules that must actually collide to form products during each step. This information is obtained by knowing the molecularity of the reaction, which is defined as the number of molecules that come together in a reaction. In a single-step reaction, the molecularity (which is the same as the order of reaction) determines the exponents in the rate equation. In a multistep reaction sequence, each step has unique molecularity; knowing this provides information about the reaction mechanism. Zero-order reactions can be explained in two ways. In a process that is truly zero order with respect to all reactants (and catalysts, in the case of enzymes), either the activation energy is zero or every molecule has sufficient energy to overcome the activation barrier. This kind of reaction is rare in homogeneous reactions in gases or solutions. Alternatively, a reaction can be zero order with respect to one or more (but not all) of the reactants. This kind of reaction is important in enzyme kinetics and assays of enzymes of clinical importance (Chapter 8). If one of the reactants (or a catalyst, such as an enzyme) is limited, then increasing the availability (concentrations) of the other reactants can result in no increase in the velocity of the reaction beyond that dictated by the limiting reagent. Hence, the rate is independent of the concentrations of the nonlimiting materials and zero-order with respect to those materials.

CHAPTER5 Thermodynamics, Chemical Kinetics, and Energy Metabolism

Theoretically all reactions are reversible. The correct way to write A --+ B, then, is ki

AZB k-1

where kl is the rate constant for the conversion of A to B, and k_l is the rate constant for the reverse reaction, i.e., conversion of B to A. The reaction is kinetically reversible if kl ~ k_l; it is irreversible for all practical purposes (i.e., the reverse reaction is so slow that it can be ignored) if k-1 6CO2(g) nt- 6HzO(e)

At 20~

A H ~ = - 6 7 3 kcal/mol or - 2 8 1 6 kJ/mol.

Oxidation of 1 mol of palmitic acid (a fatty acid) to carbon dioxide and water: C16H3202(s) + 2302(8)----> 16CO2(g) q-- 16H20(1)

At 20~ A H ~ 2380 kcal/mol or - 9 . 9 6 MJ/mol (1 MJ -- 1 megajoule -- 1000 kJ). The average A H values in kilocalories per gram are as follows: carbohydrate, 4.1; lipid, 9.4; and protein, 5.6. These values vary within each class of food depending on chemical structure. For example, the value for starch is 4.1 kcal/g, whereas for glucose it is 3.8. Furthermore, in humans the end product of protein metabolism is urea instead of CO2 and nitrogen, which are the end products obtained after complete oxidation in the bomb calorimeter. Therefore, a realistic estimate of energy derived from protein in the body, taking into account incomplete oxidation and specific dynamic action, is 4.1 kcal/g. In human nutrition, the commonly used values for energy yield in kilocalories (or kilojoules) per gram are as follows: carbohydrate, 4 (16.7); lipid, 9 (37.7); and protein, 4 (16.7). During the metabolism of food substances in the body, oxygen is consumed and CO2 is produced. The molar ratio of CO2 produced to oxygen consumed is known as the respiratory quotient (RQ) and is characteristic of a given substrate. For example, in the complete oxidation of glucose presented above, RQ-

volume of CO2 produced volume of 0 2 consumed

=

6CO2 602

= 1.0.

For the corresponding oxidation of palmitic acid, RQ -

16CO2 2302

= 0.7.

The RQ for protein is difficult to measure because it is not oxidized completely in the body and part of its carbon, hydrogen, and oxygen is lost in the urine and feces. However, the RQ is usually taken to be 0.8. The measurement of RQ thus provides a means for assessing the type of food that is being metabolized. An overall RQ of 0.85 is obtained in a normal adult who consumes a mixed diet. This value is enhanced by carbohydrate feeding and decreased by lipid feeding. Heat production can be calculated from oxygen consumption and RQ as follows. For glucose oxidation: C 6 H 1 2 0 6 -Jr- 6 0 2 -- 6 C O 2 -Jr- 6 H 2 0 180 g 192 g (134L)

Since the energy yield of 1 g of glucose is 3.8 kcal, 180 g of glucose yields 180 x 3.8 or 684 kcal. The energy yield per liter of oxygen consumed equals (180 x 3.8)/134 5.1 kcal (21.3 kJ). Again, for palmitate oxidation, C16H3202 + 2302 --+ 16CO2 + 16H20. 256 g

736 g

(513.7 L)

~8 Since the energy yield of 1 g of fat is 9 kcal, 256 g of fat yields 256 x 9, or 2304 kcal. The energy yield per liter of oxygen consumed equals (256 x 9)/513.7 = 4.5 kcal (18.8 kJ). In the body, the energy derived from food is released as body heat and also used in the synthesis of ATP. The energy captured in ATP is then transformed into other forms, i.e., chemical (synthesis of new compounds), mechanical (muscle contraction), electrical (nerve activity), electrochemical (various ion pumps), thermal (maintenance of body temperature), and informational (base sequences in nucleic acids, amino acids in proteins). In general, the energy of food provides for the specific dynamic action of food, the maintenance of the body's basal metabolism, and the energy expenditure associated with various types of activity. The term specific dynamic action (SDA) describes the thermogenic effect of food, which consists of the rise in the metabolic rate during the assimilation of food above the metabolic rate during fasting. The mechanism of the thermogenic effect is not understood, but it may be due to the work performed in the assimilation of food and in its preparation for energy storage or energy yield. These processes require energy, which is derived from the body's energy stores. The values for SDA vary with the type of food substance ingested. The SDA value for protein is about 30% of its energy value because protein must undergo many changes before the carbon skeletons of its constituent amino acids can be used appropriately. SDA values for carbohydrates and lipids are, respectively, 5% and 4% of their energy value. The energy utilized as a result of SDA is wasted and appears as heat. It is maximal with protein intake. Thermodynamically, all energy derived from the metabolism of substrates to CO2 and H20 must of course eventually appear as heat. Basal metabolism (determined by the basal metabolic rate [BMR]) is an expression of the body's vital energy needs during physical emotional, and digestive rests. It represents the energy required for the maintenance of body temperature, muscle tone, the circulation of blood, the movement of respiration, and the glandular and cellular activities of a person who is awake and not involved in physical, digestive, or emotional activities. The BMR is measured in an individual at rest after a 12-hour fast and in comfortably warm, pleasant, quiet surroundings. Although the BMR could be obtained directly by measurement of the amount of heat produced, an indirect method that measures the volume of oxygen consumed and carbon dioxide evolved per unit time is less cumbersome and provides acceptable values. Oxygen consumption is measured in a respirometer. For every liter of oxygen consumed, the energy equivalent is 4.83 kcal

CHAPTER5 Thermodynamics, Chemical Kinetics, and Energy Metabolism

(20.2 kJ). This value is based on the average value for the oxidation of carbohydrates and lipids (see above). BMR values are standardized and usually expressed as kilocalories per square meter of body surface per hour. The metabolic rate that occurs during sleep is approximately equal to the BMR. Less stringent conditions often used for hospitalized patients provide the resting metabolic rate, which is about 3% less than the BMR. The normal values for the resting rate of energy expenditure are shown in Table 5-3. It is clear that the resting metabolic energy requirements are closely related to lean body mass (as the body fat increases, the energy requirement decreases). Muscle, endocrine glands, and organs such as the liver are metabolically more active (i.e., consume a larger amount of oxygen per unit weight) than adipose tissue and bone. The B MR for women (who have a greater proportion of fat per total body weight than men) is lower than that for men. In both sexes, the development of muscle tissue as a result of increased exercise increases the metabolic rate and the basal metabolic needs. Body fat is rapidly determined with calipers by skinfold measurement at several different sites. In normal young men and women, the average body fat constitutes about 12% and 26% of total body weight, respectively; if these values are exceeded by more than 20% and 30% of the average values respectively, they indicate the presence of obesity. The BMR changes with age. It is high at birth, increases up to 2 years of age, then gradually decreases except for a rise at puberty. This general decline in the BMR (approximately 2% per decade after age 21 years) is proportional to the reduction in muscle tissue (or lean body mass) and the accompanying increase in body fat. B MR is also affected by menstruation, pregnancy, and lactation. The B MR is altered in a number of pathological states. The BMR is increased in hyperthyroidism, fever (approximately 12% elevation for each degree Celsius above normal body temperature), Cushing's syndrome, tumors of the adrenal gland, anemia, leukemia, polycythemia, cardiac insufficiency, and injury. B MR is decreased in hypothyroidism, starvation, malnutrition, hypopituitarism, hypoadrenalism (e.g.,Addison's disease), and anorexia nervosa. To calculate energy requirements in an individual, it is necessary to take into account BMR, physical activity (muscular work), age, sex, height, and weight (Appendix I). The energy requirements of muscular work can be measured. Table 5-4 lists the energy expenditure for various physical activities. Maintenance of desirable body weight at any age depends on the balance between energy intake and energy output or requirement. During growth, pregnancy, or recovery from illness, energy demands are greater; hence,

SECTION5.5 Energy Metabolism

79

TABLE 5-3 Normal Values for the Resting Rate of Metabolic Energy Expenditure of Adults Expressed in kcal/min (kJ/min)*

Men

Percent Body Women Fat

Thin Average

Weight in kg (lbs) 45(99)

50(110)

55(121)

60(132)

65(143)

70(154)

75(165)

80(176)

5

0.98 (4.10)

1.05 (4.39)

1.12 (4.69)

1.20 (5.02)

1.27 (5.31)

1.32 (5.52)

1.40 (5.86)

10

0.94 (3.93)

1.00 (4.18)

1.08 (4.52)

1.15 (4.81)

1.22 (5.10)

1.27 (5.31)

1.34 (5.61)

Plump

Thin

15

0.82 (3.42)

0.89 (3.72)

0.96 (4.02)

1.04 (4.35)

1.11 (4.64)

1.18 (4.94)

1.22 (5.10)

1.30 (5.44)

Fat

Average

20

0.79 (3.30)

0.84 (3.51)

0.90 (3.77)

0.98 (4.10)

1.05 (4.39)

1.12 (4.69)

1.18 (4.94)

1.24 (5.19)

Plump

25

0.79 (3.31)

0.86 (3.60)

0.94 (3.93)

1.00 (4.18)

1.08 (4.52)

1.12 (4.69)

1.20 (5.02)

Fat

30

0.82 (3.43)

0.89 (3.72)

0.96 (4.02)

1.04 (4.35)

1.08 (4.52)

1.15 (4.81)

*Data from S. Davidson, R. Passmore,J. F. Brock, and A. S. Truswell:HumanNutrition and Dietetics, 7th ed. (ChurchillLivingstone, 1979).

intake should be higher than output. Obesity and cachexia (a disorder characterized by general physical wasting and malnutrition) are extreme examples of problems affecting the energy stores of the body. The appetite and satiety centers in the central nervous system normally are

sensitive regulators that adjust energy consumption to energy requirements. Obesity is rarely caused by damage to those centers. Obesity results from an excess of energy intake over physiological needs. Factors predisposing or contributing to the development of obesity are heredity,

TABLE 5-4

Examples of Rate of Energy Expenditure during Various Types of Physical Activities of Adults* Rate of Energy Expenditure, kcal/min (kJ/min) Type of Activity

Man, 70 kg

Woman, 58 kg

Very light: Sitting, standing, painting, driving, working in a laboratory, typing, playing musical instrument, sewing, ironing.

up to 2.5 (up to 10.5)

up to 2.0 (up to 8.3)

Light: Walking on level (2.5-3 mph), tailoring, pressing, working on automobiles, washing, shopping, golfing, sailing, playing table tennis or volley ball.

2.5-4.9 (10.5-20.5)

2.0-3.9 (8.37-16.3)

Moderate: Walking (3.5-4 mph), plastering, weeding and hoeing, loading and stacking bales, scrubbing floors, shopping with heavy load, cycling, skiing, playing tennis, dancing.

5.0-7.4 (20.9-31.0)

4.0-5.9 (16.7-24.7)

Heavy: Walking uphill with load, tree felling, working with pick and shovel, basketball, swimming, climbing, playing football.

7.5-12.0 (31.4-50.2)

6.0-10.0 (25.1-41.8)

*Adapted from RecommendedDietaryAllowances, 9th ed. (Food and Nutrition Board, National Research CouncilINational Academy of Sciences, Washington, D.C., 1980).

~0

CHAPTER5 Thermodynamics, Chemical Kinetics, and Energy Metabolism

endocrine disorders, behavior, lifestyle and physical activity, eating habits, and culture. Obesity is the most common nutritional disorder in developed countries. A sedentary lifestyle is a significant contributing factor in insidious weight gain. Sedentary lifestyles may not only predispose individuals to obesity but also lead to degenerative arterial diseases. A sound exercise program coupled with good nutrition can help maintain health and quality of life. Obese individuals who wish to lose weight by dietary restriction and increased physical activity should be under medical supervision. The human body needs a wide variety of nutrients to function optimally. Recommended Dietary Allowance (RDA), published by the Food and Nutrition Board of the National Academy of Sciences National Research Council (USA), provides a broad guideline of the amounts of some of the well-established nutrients (protein, vitamins, and minerals) that should be consumed daily to maintain normal health (RDA values are given in Appendix IV). To use the RDA properly, keep in mind the following: 1. RDA values are overestimates of nutrient requirements; minimum quantities of nutrients required to maintain normal function and health in humans are either unknown or incompletely known. 2. RDA values relate primarily to populations of developed countries and are intended to meet the needs of virtually all healthy individuals. Thus, the allowances are far higher than demanded by physiological needs, and for many populations of the world, the RDA estimates may be inappropriately high. If the intake of a given nutrient is below the RDA value, nutritional inadequacy does not necessarily result. However, if a particular deficiency is accompanied by biochemical and clinical abnormalities, then corrective action is needed.

Clinical conditions include trauma and stress, prematurity, inherited metabolic disorders, disease states and rehabilitation, and use of certain drugs. 3. Since the RDA applies to healthy people, it does not provide requirements in disease states or in drug-nutrient interactions. 4. RDA does not address the fact that some nutrients possess pharmacological activities unrelated to nutrient function. Excessive intake of such nutrients can have toxic effects. 5. RDA does not provide values for all essential nutrients, and therefore the daily requirement should be met by selection from a wide variety of foods. No single food source is nutritionally complete. Foods are categorized into four groups: 1) milk and milk products, 2) meat and meat alternatives (e.g., legumes, nuts, and milk products), 3) fruits and vegetables, and 4) breads and cereals. Table 5-5 lists the key nutrients in some representative foods in each group. Not only is consumption of adequate amounts of nutrients essential but so is consumption of appropriate quantities of foods that provide amounts of energy commensurate with physical activity and the physiological state. Knowledge of a given nutrient concentration (also known as nutrient density) per unit of energy (e.g., 1000 kcal) in a specific food is useful in adjusting the diet to individual need. Dietary fiber, although not nutritionally essential, is important in maintaining good health. Dietary fiber consists of polymers of sugars and other indigestible substances. Fiber affects nutrient absorption and makes the contents of the gut bulky and soft. In terms of the four food groups (Table 5-4), fiber can be obtained from the second group by selection of dry beans, peas, and nuts in place of (or in addition to) meat, poultry, and fish

TABLE 5-5

Food Groups*

Group 1. Milk and milk products (e.g., milk, yogurt, cheese, ice cream) 2. Meat (e.g., beef, pork, poultry, fish) and meat alternatives (e.g., legumes, nuts, milk products) 3. Fruits and vegetables (e.g., green leafy, deep yellow, or orange vegetables, citrus fruits) 4. Breads, cereals, and grains

Major Nutrients Provided Calcium, riboflavin, protein Protein, niacin, iron, thiamine (vitamin B l), Vitamins A and C Carbohydrate, thiamine, iron, niacin

*A well-balanced diet includes a variety of foods on a daily basis, selected from each of the four food groups. With proper planning a vegetarian diet can be nutritionally adequate. Special foods consumed by ethnic groups should be considered in diet planning. Basic principles of a good diet include consumption of a variety of foods; consumption to maintain ideal body weight; moderation in intake of foods or beverages rich in saturated fats, cholesterol, sugar, and sodium salts; and consumption of adequate quantities of fruits, vegetables, and whole grains.

SECTION5.5 Energy Metabolism

81

TABLE 5-6

BMI Values Corresponding to Different Height-Weight Combinations* BMI -->

19

20

21

22

23

24

25

Weight**

Height* ft.in,

m

lb

kg

lb

kg

lb

kg

lb

kg

lb

kg

lb

kg

lb

kg

4'10" 4'11" 5'0" 5'1" 5'2" 5'3" 5'4" 5'5" 5'6" 5'7" 5'8" 5'9" 5'10" 5'11" 6'0" 6'1" 6'2" 6'3" 6'4"

1.473 1.499 1.524 1.549 1.575 1.600 1.626 1.651 1.676 1.702 1.727 1.753 1.778 1.803 1.829 1.854 1.880 1.905 1.930

91 94 97 100 104 107 110 114 118 121 125 128 132 136 140 144 148 152 156

41.28 42.64 44.00 45.36 47.17 48.54 49.90 51.71 53.52 54.89 56.70 58.06 59.88 61.69 63.50 65.32 67.13 68.95 70.76

96 99 102 106 109 113 116 120 124 127 131 135 139 143 147 151 155 160 164

43.55 44.91 46.27 48.08 49.44 51.26 52.62 54.43 56.25 57.61 59.42 61.24 63.05 64.86 66.68 68.49 70.31 72.58 74.39

100 104 107 111 115 118 122 126 130 134 138 142 146 150 154 159 163 168 172

45.36 47.17 48.54 50.35 52.16 53.52 55.34 57.15 58.97 60.78 62.60 64.41 66.23 68.04 69.85 72.12 73.94 76.20 78.02

105 109 112 116 120 124 128 132 136 140 144 149 153 157 162 166 171 176 180

47.63 49.44 50.80 52.62 54.43 56.25 58.06 59.88 61.69 63.50 65.32 67.59 69.40 71.22 73.48 75.30 77.57 79.83 81.65

110 114 118 122 126 130 134 138 142 146 151 160 162 165 169 174 179 184 189

49.90 51.71 53.52 55.34 57.15 58.97 60.78 62.60 64.41 66.23 68.49 72.58 73.48 74.84 76.66 78.93 81.19 83.46 85.73

115 119 123 127 131 135 140 144 148 153 158 162 167 172 177 182 186 192 197

52.16 53.98 55.79 57.61 59.42 61.24 63.50 65.32 67.13 69.40 71.67 73.48 75.75 78.02 80.29 82.56 84.37 87.09 89.36

119 124 128 132 136 141 145 150 155 159 164 169 174 179 184 189 194 200 205

53.98 56.25 58.06 59.88 61.69 63.96 65.77 68.04 70.31 72.12 74.39 76.66 78.93 81.19 83.46 85.73 88.00 90.72 92.99

BMI --~

Weight**

Height* ft.in,

m

40

35

30

29

28

27

26

lb

kg

4'10" 1.473 4'11" 1.499 5'0" 1.524 5'1" 1.549 5'2" 1.575 5'3" 1.600 5'4" 1.626 5'5" 1.651 5'6" 1.676 5'7" 1.702 5'8" 1.727 5'9" 1.753 5'10" 1.778 5'11" 1.803 6'0" 1.829 6'1" 1.854 6'2" 1.880 6'3" 1.905 6'4" 1.930

124 128 133 137 142 146 151 156 161 166 171 176 181 186 191 197 202 208 213

56.25 58.06 60.33 62.14 64.41 66.23 68.49 70.76 73.03 75.30 77.57 79.83 82.10 84.37 86.64 89.36 91.63 94.35 96.62

*Without shoes

**Withoutclothes

lb

kg

129 58.51 133 60.33 138 62.60 143 64.86 147 66.68 152 68.95 157 71.22 162 73.48 167 75.75 172 78.02 177 80.29 182 82.56 188 85.28 193 87.54 199 90.27 204 92.53 210 95.26 216 97.98 221 100.25

lb

kg

134 60.78 138 62.60 143 64.86 148 67.13 153 69.40 158 71.67 163 73.94 168 76.20 173 78.47 178 80.74 184 83.46 189 85.73 195 88.45 200 90.72 206 93.44 212 96.16 218 98.88 224 101.61 230 104.33

lb

kg

138 62.60 143 64.86 148 67.13 153 69.40 158 71.67 163 73.94 169 76.66 174 78.93 180 81.65 185 83.92 190 86.18 196 88.91 202 91.63 208 94.35 213 96.62 219 99.34 225 102.06 232 105.24 238 107.96

lb

kg

lb

kg

lb

kg

143 148 153 158 163 169 174 180 186 191 197 203 209 215 221 227 233 240 246

64.86 67.13 69.40 71.67 73.94 76.66 78.93 81.65 84.37 86.64 89.36 92.08 94.80 97.52 100.25 102.97 105.69 108.86 111.59

167 173 179 185 191 197 204 210 216 223 230 236 243 250 258 265 272 279 287

75.75 78.47 81.19 83.92 86.64 89.36 92.53 95.26 97.98 101.15 104.33 107.05 110.22 113.40 117.03 120.20 123.38 126.55 130.18

191 198 204 211 218 225 232 240 247 255 262 270 278 286 294 302 311 319 328

86.64 89.81 92.53 95.71 98.88 102.06 105.24 108.86 112.04 115.67 118.84 122.47 126.10 129.73 133.36 136.99 141.07 144.70 148.78

82

CHAPTER 5

from the third group; and from the fourth group by selection of whole-grain products (See also Chapter 9).

5.6 Obesity The first law of thermodynamics states that the amount of stored energy equals the difference between energy intake and energy expenditure. The principal storage of energy is that of triglycerides in adipose tissue. Energy stores are essential for survival during times of energy deprivation (Chapters 18 and 22). Energy stores in an adult are maintained at a relatively constant level throughout life. However, even a small imbalance in energy intake over long periods of time will have a significant effect on energy storage. For example, suppose a non obese adult's energy intake exceeds expenditure by about 1% daily for one year. This amounts to an excess of 9,000 kcal and corresponds to a weight gain of about 2.5 lb (1.15 kg) per year. 3,500 kcal of chemical energy is equivalent to 1 lb (0.45 kg) of adipose tissue. Weight gain in most people is attributable to overconsumption of palatable, energy-dense foods (e.g., lipids) and a sedentary lifestyle. Childhood obesity is a risk factor for obesity in adulthood. Obesity is a consequence of a positive energy balance, i.e., input is greater than output. Body mass index (BMI) is the most useful parameter in assessing the magnitude of obesity (Table 5-6). A BMI of 19-25 is considered healthy, 26-29.9 is moderately overweight, and 30 or greater is obese. Obesity is the most common health problem in the developed world, and it is estimated that obesity affects about one third of the U.S. adult population. In developed nations, the prevalence of obesity is higher among economically deprived people, whereas in developing nations, the relatively affluent have a higher risk. A BMI greater than 28 is an independent risk factor (3-4 times higher than the general population) for cardiovascular diseases (Chapter 20), diabetes mellitus type 2 (Chapter 22), and stroke. The prevalence of obesityassociated morbidity depends on the location of fat distribution in the body. Intra-abdominal or visceral fat deposits are associated with higher health risks than gluteofemoral adipose tissue fat accumulation. In experimental animals and possibly in humans, energy intake influences the aging process as well as the onset of aging-associated diseases. Energy restriction in animals, without altering their optimal nutritional status, increases the average life span but not the maximal life

Thermodynamics,Chemical Kinetics, and Energy Metabolism

span. This beneficial biological response to energy restriction has been attributed to the attenuation of oxidative damage to proteins, lipids, and DNA (Chapter 14). During aging, despite increased body fat, there is a linear decrease in food intake and metabolism over the life span.

Biochemical Mediators of Obesity The regulation of energy intake and expenditure is achieved by coordinating the effects of endocrine mediators and neural signals that arise from adipose tissue, endocrine glands, neurological and gastrointestinal systems. All of the information finally is integrated by the central nervous system (Figure 5-6). One of the most significant mediators of the energy store in the adipose tissue is leptin (from the Greek leptos, meaning "thin"). Leptin is a protein of 167 amino acid residues that is synthesized in adipocytes. Its synthesis is increased by insulin, glucocorticoids, and estrogens and is decreased by/3-adrenergic agonists. The role of leptin in obesity comes from studies on rodents. In genetically obese mice (Ob/Ob), the observed gross obesity is due to absence of leptin production in the adipocytes. Leptin's action on energy metabolism is mediated by receptors in many cells and it binds specifically to a receptor in the hypothalamus. The action of leptin involves at least two pathways. During starvation and weight loss, adipose tissue is decreased with consequent low levels of leptin. The low level of leptin leads to production of neuropeptide Y, which is synthesized in the arcuate nucleus of the hypothalamus and is transported axonally to the paraventricular nucleus. Neuropeptide Y binds to its receptor and functions as a potent appetite stimulant. The overall effect is increased appetite, decreased energy expenditure and temperature, decreased reproductive function (infertility), and increased parasympathetic activity. An opposite set of events occurs when the leptin levels rise, except that the effects are mediated by melanocyte-stimulating hormone (MSH) that binds to the melanocortin 4 receptor (MC4-R). The MSH binding to MC4-R initiates several biological responses, including decreased appetite, increased energy expenditure, and increased sympathetic activity (Figure 5-7). In humans, obesity is a complex disease because of the redundancy of systems that regulate energy storage. There are inherited disorders of hyperphagia leading to obesity with associated clinical features such as hypogonadism and mental retardation. One hereditary disorder is PraderWilli syndrome (Chapter 26), which is the most prevalent form of dysmorphic genetic obesity (1 in 10,000-20,000

SECTION 5.6

Obesity

83 Afferent signals increasing appetite or decreasing energy expenditure

Afferent signals decreasing appetite or increasing energy expenditure

Gastrointestinal tract Opioids,neurotensin, growthhormone-releasing hormone,somatostatin

Gastrointestinal tract Glucagon,cholecystokinin, glucagon-likepeptides, bombesmpeptides,glucose

Endocrine system l" Epinephrine(o~-adrenergiceffect), androgens,glucocorticoids, insulin,peptideYY,progesterone

Endocrine system Epinephrine([3-adrenergiceffect), estrogens

~

t ' ~ ~ ] ~ : . 4 ' Adipose . ~ . . = ~ . ;tissue .

~

l

Per,0hera, nervous system Norepinephrine (cz-adrenergiceffect))

Peripheral nervous system l Norepinephrine (13-adrenergiceffect)

} - - ~ -~

Centra, oe~oos s~stem Dopamine,y-aminobutyricacid, serotonin,cholecystokinin

Centra' ne ous system Oa,anin,opioi~s,

~ " ' ~ ' ~ #

Hypothalamic tracts Norepinephrine, serotonin, neuropeptide Y, melanocyte-concentratinghormone, glucagon-like peptide I, corticotropin-releasing hormone

.

growthhormone-releasing hormone,somatostatin . . . .

@

Efferents

Sympathetic nervous system parasympathetic nervous system, thyroid hormones

@

Energy intake and expenditure

FIGURE 5-6 Afferent and efferent pathways of regulation of energy intake and expenditure. [Reproduced with permission from M. Rosenbaum, R. L. Leibel, and J. Hirsch. Obesity. N. Engl. J. of Med. 337:396 (1997).]

Starvation

Overeating (hyperphagia) obesity

Weight loss and decreased adipose tissue

Weight gain and increased adipose tissue

keptin

keptin

I Transport across blood-brain barrier I

I Transport across blood-brain barrier I

Leptin receptor-hypothalamus

Leptin receptor-hypothalamus

Neuropeptide Y and Y5 receptor

Melanocyte-stimulating hormone-

Biological responses: Food intake Energy expenditure Fertility TemperatureS, Parasympathetic activity

Melanocortin receptor

Biological responses: Food intake Energy expenditure Sympathetic activity

FIGURE 5-7 An overview of the action of leptin on food intake, energy expenditure, and biological responses (1" increased and ,l, decreased).

.

84

births). Deletions in chromosome 15 account for a majority of cases of Prader-Willi syndrome. In other cases, a novel genetic mechanism of uniparental disomy is the cause of this syndrome. Congenital human leptin deficiency is associated with early-onset obesity. Homozygous mutations in the human leptin receptor gene result in a truncated leptin receptor lacking both transmembrane and intracellular domains; such mutations are associated with early-onset obesity, absence of pubertal development, and pituitary dysfunction. In most obese humans, the plasma levels of leptin are high due to excess adipose tissue. In spite of abundant leptin, there is continued overeating. This puzzling observation may ultimately be explained by as yet-to-be identified defects in leptin metabolism.

Supplemental Readings and References P. Bj6rntorp: Obesity. Lancet 350, 423 (1997). J. E Caro, J. W. Kolaczynski, M. R. Nyce, et al.: Decreased cerebrospinalfluid/serum leptin ratio in obesity: A possible mechanism for leptin resistance. Lancet 348, 159 (1996).

CHAPTER5 Thermodynamics, Chemical Kinetics, and Energy Metabolism K. Clement, C. Vaisse, N. Lahlou, et al.: A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature 392, 398 (1998). J. M. Friedman and J. L. Halaas: Leptin and the regulation of body weight in mammals. Nature 395, 763 (1998). C. T. Montague, I. S. Farooqi, J. E Whitehead, et al.: Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387, 903 (1997). J. E. Morley: Anorexia of aging: Physiologic and pathologic. American Journal of Clinical Nutrition 66, 760 (1997). Recommended Dietary Allowances, 10th ed. Food and Nutrition Board, National Research Council-National Academy of Sciences, Washington, DC, (1989). M. Rosenbaum, R. L. Liebel, and J. Hirsch: Obesity. New England Journal of Medicine 337, 396 (1997). M. W. Schwartz, E. Peskind, M. Raskind, et al.: Cerebrospinal fluid leptin levels: Relationship to plasma levels and to adiposity in humans. Nature Medicine 2, 589 (1996). J. Stevens, J. Cai, E. R. Parnuk, et al.: The effect of age on the association between body-mass index and mortality. New England Journal of Medicine 338, 1 (1998). L. A. Tartaglia, M. Dembroski, X. Weng, et al.: Identification and expression of cloning of a leptin receptor, OB-R. Cell 83, 1263 (1995). R. Weindruch and R. S. Sohal: Caloric intake and aging. New Journal of Medicine 337, 986 (1997).

CHAPTER

6

Enzymes I: General Properties, Kinetics, and Inhibition

This chapter deals with enzyme catalysis, kinetics, inhibition, and mechanisms. Enzymes are catalysts and are functional units of cellular metabolism. Enzymes are usually proteins but may include RNA molecules as well. This chapter and the following two chapters discuss protein enzymes. RNA molecules are ribonucleases (ribozymes) that recognize specific nucleotide sequences in the target RNA and hydrolyze phosphodiester bonds. Ribosomal RNA functions as a peptidyl transferase in protein biosynthesis (Chapter 25). Some RNA molecules also undergo a self-splicing process. One example is the transformation of pre-mRNA to mature, functional mRNA by the splicing out of intervening sequences (introns) and ligation of the coding sequences (exons). The splicing-ligation process requires small nuclear ribonucleoprotein particles (snRNPs) and other proteins. Enzymatic RNA also can act on other RNA molecules. An example is ribonuclease P (RNase P), which is involved in the conversion of precursor tRNA to functional tRNA by generating 5'-phosphate and 3'hydroxyl termini. RNase P contains both a catalytic RNA moiety and an associated protein (Chapter 25). Many enzymes are bound either covalently or noncovalently to nonprotein components essential for enzymatic activity. The term prosthetic group is generally reserved for those nonprotein components that are bound tightly, whereas the term coenzyme applies to less tightly bound nonprotein components. The coenzymes

are complex organic compounds, frequently derived from vitamins. The term eofaetor is used for metal ions and simple organic compounds that partipate in enzyme catalysis. The protein portion of an enzyme is the apoenzyme, and the fully functional enzyme with its attached nonprotein component is the holoenzyme (i.e., apoenzyme + coenzyme = holoenzyme).

6.1 Nomenclature Enzymes are generally named for the substrate or chemical group on which they act, and the name takes the suffix -ase. Thus, the enzyme that hydrolyzes urea is named urease. Examples of exceptions to this terminology are trypsin, pepsin, and papain, which are trivial names. Systematic nomenclature for the enzymes has been developed by the Enzyme Commission of the International Union of Biochemistry. This system provides a rational and practical basis for identification of all enzymes currently known as well as for new enzymes. The systematic name describes the substrate, the nature of the reaction catalyzed, and other characteristics. A unique numerical code consisting of four numbers separated by periods (e.g., EC. 4.2.1.1) is designated. The prefix "EC" denotes "Enzyme Commission." The first number in this designation specifies the class to which the enzyme belongs. All enzymes are assigned to one

85

86

CHAPTER6 Enzymes I: General Properties, Kinetics, and Inhibition

of six classes on this basis of the type of reaction they catalyze: 1. 2. 3. 4.

oxidoreductases (oxidation-reduction), transferases (transfer of groups), hydrolases (hydrolysis), lyases (nonhydrolytic and nonoxidative cleavage of groups), 5. isomerases (isomerization), and 6. ligases or synthetases (joining of two molecules with the breaking of a pyrophosphate bond).

The next two numbers in the code indicate the subgroup and the sub-subgroup; the last number is the special serial number given to each enzyme in its sub-subgroup. Consider the systematic nomenclature for the enzyme with the trivial name carbonic anhydrase, which facilitates the transport of CO2 by catalyzing the following reaction (Chapter 1):

(D-glucose, 2-deoxy-D-glucose, D-fructose, and D-mannose) at almost equivalent rates. 9 Phosphatase, which hydrolyzes phosphate groups from a large variety of organic phosphate esters. 9 Esterase, which hydrolyzes esters to alcohols and carboxylic acids, with considerable variation in chain length in both the alkyl and acyl portions of the ester. 9 Proteinase, which hydrolyzes peptide bonds irrespective of the chemical nature of the substrate. Many enzymes show stereoisomeric specificities. For example, human or-amylase catalyzes the hydrolysis of glucose from the linear portion of starch but not from cellulose. Starch and cellulose are both polymers of glucose, but in the former the sugar residues are connected by or(1 --+ 4) linkages, whereas in the latter they are connected by/3(1 --+ 4) linkages (Chapter 9).

H2CO3 (or H + + HCO3) ~-- H 2 0 Jr- CO2

Active Site and Enzyme-Substrate Complex

or H2CO3(or H § -t- HCO3- ) ~ H20 + CO2

or

O II

The systematic name for carbonic anhydrase is carbonate hydro-lyase, and its numerical code is EC. 4.2.1.1. The first number identifies it as a lyase; the second as an enzyme that catalyzes the breakage of a carbon-oxygen bond, leading to unsaturated products; and the third as a hydro-lyase, participating in a reaction involving the elimination of water. The last number is the specific serial number assigned to this enzyme. In this text, the trivial names are used. Names of a selected list of clinically useful enzymes with their EC codes, systematic names, other common names, and abbreviations are given in Appendix V.

6.2 Catalysis Specificity of Enzyme Catalysis Enzymes are highly specific and usually catalyze only one type of reaction. Some enzymes show absolute specificity. For example, pyruvate kinase catalyzes the transfer of a phosphate group only from phosphoenolpyruvate to adenosine diphosphate during glycolysis (Chapter 13). Examples of enzymes that show less specificity are: 9 Hexokinase, which transfers the phosphate group from adenosine triphosphate to several hexoses

An enzyme-catalyzed reaction is initiated when the enzyme binds to its substrate to form an enzyme-substrate complex. In general, enzyme molecules are considerably larger than the substrate molecules. Exceptions are proteinases, nucleases, and amylases that act on macromolecular substrates. Irrespective of the size of the substrate, binding to the enzyme occurs at a specific and specialized region known as the active site, a cleft or pocket in the surface of the enzyme that constitutes only a small portion of the enzyme molecule. Catalytic function is accomplished at this site because various chemical groups important in substrate binding are brought together in a spatial arrangement that confers specificity on the enzyme. Thus, the unique catalytic property of an enzyme is based on its three-dimensional structure and on an active site whose chemical groups may be brought into close proximity from different regions of the polypeptide chain. The stereospecificity of an enzyme for a substrate has been compared to a lock-and-key relationship. This analogy implies that the enzyme has an active site that fits the exact dimensions of the substrate (Figure 6-1). However, after attachment of substrate, the enzyme may undergo conformational changes that provide a more perfect fit between it and the substrate. This process has been described as inducedfit (Figure 6-2).

Factors Governing the Rate of Enzyme-Catalyzed Reactions The overall reaction involving conversion of substrate (S) to product (P) with formation of the enzyme-substrate

SECTION 6.2

Catalysis

8? to the same extent. Thus, enzymes affect only the rate at which equilibrium is established between reactants and products. However, under steady-state conditions, which is the normal state of affairs in the human body, the net effect of the enzyme is to convert substrates to products as rapidly as the products are removed (Chapter 5).

Effect of Temperature

FIGURE 6-1

Schematic diagramof a lock and key relationshipbetween an enzymeand its substrate. The shape of the active site is complementaryto that of the substrate molecule.

complex, ES can be written: kl

E+S

k2

~-- ES ~ E + R k-1

k-2

where kl, k_l, k2, and k_2 are rate constants for the designated steps; the positive subscripts indicate forward reactions, and the negative subscripts indicate backward reactions. Because an enzyme increases the rate of a particular reaction by decreasing the free energy of activation without itself being consumed or permanently altered, a small number of enzyme molecules can convert an extremely large number of substrate molecules to products very rapidly. Enzymes do not alter the equilibrium constant of a chemical reaction because they catalyze the forward and backward reactions

FIGURE 6-2

Schematic diagramof the induced-fitmodel for the relationshipbetween an enzymeand its substrate. The shape of the active site of the enzyme conforms to that of the substrate onlyafter substrate binding induces appropriate conformationalchangesin the enzyme.

The rates of almost all chemical reactions increase with a rise in temperature, which causes both the average kinetic energy and the average velocity of the molecules to increase, resulting in a higher probability of effective reaction collisions. Enzymes, however, undergo denaturation and are inactivated at high temperatures. Below the denaturation temperature, the reaction rate will approximately double for every rise of 10~ The ratio by which the rate changes for a 10~ increase in temperature is known as Q 10; this ratio varies from 1.7 to 2.5. The optimal temperature for most enzymes is close to the normal temperature of the organism at which catalysis occurs at the maximum rate. In humans, most enzymes have an optimal temperature of 37~

Effect of pH The activity of most enzymes depends on pH. The p H enzyme activity profile of most enzymes delineates a bellshaped curve (Figure 6-3), exhibiting an optimal pH at which activity is maximal. This pH is usually the same as the pH of the fluid in which the enzyme functions. Thus, most enzymes have their highest activity between pH 6 and pH 8 (the pH of human blood is about 7.4). However, pepsin, which must function at the low pH of gastric juice, has maximal activity at about pH 2. The pH dependence of enzyme activity is the result of several effects. Ionizable groups in the active site of the enzyme (or elsewhere), in the substrate, or in the enzyme-substrate complex can affect catalysis depending on whether the protons on the reactive groups are dissociated or undissociated. Ionization of these groups depends on their pK values, the chemical properties of surrounding groups, and the pH of the reaction medium. Changes in pH affect the binding of the substrate at the active site of the enzyme and also the rate of breakdown of the enzyme-substrate complex. Thus, it may be possible to infer the identity of an ionizable group that participates at the active site from the pH-activity profile for a given enzyme. The enzymes in living systems function at nearly constant pH because they are in an environment that contains buffers (Chapter 1).

CHAPTER6 Enzymes I: General Properties, Kinetics, and Inhibition

~8

F I G U R E 6-4 Plot of substrate concentration versus initial velocity of an enzymecatalyzed reaction. Segment A: At low substrate concentration, the reaction follows first-order kinetics with respect to substrate concentration; i.e., v = k[S], where k is a reaction rate constant. Segment B: At high substrate concentration, maximum velocity (Vmax) is attained (saturation kinetics), and any further increase in substrate concentration does not affect the reaction rate; the reaction is then zero-order with respect to substrate but first-order with respect to enzyme. Km is the value of [S] corresponding to a velocity of 1 Vmax.

F I G U R E 6-3 pH dependence of the rates of enzyme-catalyzed reactions. (a) Optimal activity at alkaline pH. (b) Optimal activity of acidic pH.

derived for a single substrate-enzyme-catalyzed reaction. In the reaction k~

k2

E + S +--'--ES--+E + P k-i

Effect of Concentration of Enzyme and Substrate The reaction rate is directly proportional to the concentration of the enzyme if an excess of free substrate molecules is present. Thus, enzyme-substrate interactions obey the mass-action law. For a given enzyme concentration, the reaction velocity increases initially with increasing substrate concentration. Eventually, a maximum is reached, and further addition of substrate has no effect on reaction velocity (v) (Figure 6-4). The shape of a plot of v versus [S] is a rectangular hyperbola and is characteristic of all nonallosteric enzymes (Chapter 7). At low substrate concentrations, the reaction rate is proportional to substrate concentration, with the reaction following firstorder kinetics in terms of substrate concentration.

Michaelis-Menten Treatment of the Kinetic Properties of an Enzyme A model for enzyme kinetics that has found wide applicability was proposed by Michaelis and Menten in 1913 and later modified by Briggs and Haldane. The MichaelisMenten equation relates the initial rate of an enzymecatalyzcd reaction to the substrate concentration and to a ratio of rate constants. This equation is a rate equation,

an enzyme E combines with substrate S to form an ES complex with a rate constant kl. The ES complex formed can dissociate back to E and S with a rate constant k-l, or it can give rise to a product P and regenerated enzyme with a rate constant k2. In the latter process, the formation of product is essentially irreversible; the reverse reaction k_'~ (E + P --4 ES) does not occur to any appreciable extent. Thus, k-2 is much less than k2, which corresponds to a large, negative AG in going from ES to E + E In the Michaelis-Menten expression, [ET] represents the total concentration (in moles per liter) of enzyme, equal to [E] + [ES]. Therefore, the concentration of free enzyme [E] is equal to [ET] -- [ES]. The following assumptions are made: 1. In the reaction mixture, [S] is greater than [E]. However, [S] is not so large that all enzyme molecules are in the ES form, but [S] must be sufficiently large that [S] does not rapidly become so small that [S] < [E]. Excess concentration of substrate is the most common condition in enzymatic studies in which the catalyst is present at very low concentration. Under these conditions, [ES] rapidly becomes constant, so that the rate of formation of ES

SECTION 6.2

Catalysis

89

equals its rate of breakdown, and steady-state kinetics then applies. 2. Only initial velocities are measured. Hence, during the period of measurement, [S] is not significantly depleted as [P] increases. 3. [ES] is constant during the period of measurement, which means that (a) enough time has elapsed since mixing E and S for [ES] to build up; (b) not enough time has passed for the rate of formation of ES to decrease owing to substrate depletion; and (C) k2 < k - l , so that [ES] can build up. If k2 > k - l , ES breaks down as rapidly as it is formed, and a steady-state can never be established. Rate of formation of ES --

d[ES]

dt

These expressions can both be solved for k2" k2 --

d[ES]

= k-1 [ES] 4- k2 [ES]

kl ([ET] -- [ES])[S] -- (k_l 4- k2)[ES].

(6.1)

Rearranging the above Equation yields

[ES]

k-1 4- k2 kl

= Km.

(6.2)

Note that Km (the Michaelis constant) is not an equilibrium constant but a ratio of rate constants. Equation (6.2) can be rearranged in the following ways" [S][E7] - [S][ES] -- Km[ES] or

[S][ET] - [ES] (Km + [S]) or

[ S ] - [ES] [ET] (Km 4- [S]).

[ET]

Vmax

V

[ES]

--

[ET]

v or

Wmax

[ES] --

[E7]

.

Substituting the latter equation for [ES]/[ET] in Equation (6.3), we obtain [S] --

V

Vmax

(Km 4- [S]).

Rearranging and solving for v,

Under steady-state conditions, the rate of formation of ES equals its rate of breakdown, so that

=

[gs]

- kl [E] [S]

dt -- (k-1 4- k2)[ES].

([ET] - [ES])[S]

Vmax

and k2 -

Therefore,

v --

-- kl ([ET] -- [ES])[S].

Rate of breakdown of ES --

V

(6.3)

It is usually difficult to measure [ES] in a reaction mixture. Consequently, Equation (6.3) is not useful experimentally. On the other hand, the velocity (v) and the maximum velocity (Vmax) are readily determined by a variety of methods. As indicated in Figure 6-4, Vmaxis the limiting value that v approaches as [S] --+ oc. In that situation, all enzyme molecules have substrate bound to them, and so [E ] = 0 and [ET] = [ES]. Thus, v = k2 [ES], and Vmax = k2 [ET].

[ S ] Vmax

Km 4- [S]

.

(6.4)

Equation (6.4) is known as the Michaelis-Menten equation, and the following points should be noted. 1. Km is a constant for a particular enzyme and substrate and is independent of enzyme and substrate concentrations. 2. Vmaxdepends on enzyme concentration, and at saturating substrate concentration, it is independent of substrate concentration. 3. Km and Vmax may be influenced by pH, temperature, and other factors. 4. A plot of v versus [S] fits a rectangular hyperbolic function (Figure 6-4). 5. If an enzyme binds more than one substrate, the Km values for the various substrates can be used as a relative measure of the affinity of the enzyme for each substrate (the smaller the value of Km, the higher the affinity of the enzyme for that substrate). 6. In a metabolic pathway, Km values for enzymes that catalyze the sequential reactions may indicate the rate-limiting step for the pathway (the highest Km corresponds roughly to the slowest step). 7. When k_l >> k2, [ES] is assumed to be at equilibrium with [E] and [S], ES is dissociating more often to yield E and S than to yield product. Under this condition, the dissociation constant of the enzyme-substrate complex Ks for this equilibrium (ES ~ E + S) is Ks--

[E] [S]

k-1

[ES]

kl

But K m

k-1 + k e kl

90

CHAPTER 6

so that

Enzymes I General Properties, Kinetics, and Inhibition

T A B L E 6-1

Turnover Numbers of Some Enzymes

k-1 Km

=

kl

=

Ks.

Turnover Number (per second)

Enzyme 8. When k2 ) ) k_l, the rate of dissociation of ES to E and S is low, so that products are usually formed. For practical purposes, the reaction sequence can be thought of as consisting of two irreversible reactions: E + S-~ES-~ E+ P The overall reaction rate is always determined by [ES]. At low values of [S], ES is formed by a second-order reaction whose rate is proportional to [E][S]. At high values of [S], [ES] is constant and the rate is determined by the first-order breakdown of ES to product. 9. When [S] >> Km, the characteristic property of the turnover number for an enzyme can be invoked. This number provides information on how many times the enzyme performs its catalytic function per unit time, or how many times it forms the ES complex and is regenerated (turned over) by yielding product. The turnover number can be determined from Vmax and [ET]. We know that d (P)

dt

- - Vmax - - k 2 [ E S ] -

k2[ET]

under saturation conditions. Therefore, if [ET] is increased n-fold while [S] > [ET] is maintained, then Vmax will also increase n-fold. The rate is proportional to (or first order with respect to) [ET]. The kinetic constant ke, denoted as kcat under saturation conditions, provides the value for the turnover number. For example, if a reaction mixture contains an enzyme at a concentration of x molar, and the product is formed at a rate such that its concentration increases by y molar per second when the enzyme is saturated, Y per second. x

kcat - - -

This means that y/x substrate molecules are converted to product molecules every second. Correspondingly, the time required for a single conversion is x / y seconds. Turnover numbers for most enzymes usually range from 1 to 104 per second. A few enzymes have turnover numbers above 10 5 (Table 6-1). The ability of a cell to produce a given amount of product by an enzymatic reaction during its life span is proportional to the turnover number and the number of molecules of that enzyme in the cell. Because turnover numbers

Catalase Carbonic anhydrase Acetylcholinesterase a-Amylase Lactate dehydrogenase /3-Galactosidase Chymotrypsin Phosphoglucomutase Tryptophan synthase Lysozyme

5 x 10 6 6 x 10 5 2.5 x 10 5 1.9 x 104 1 x 10 3 2 x 10 2 10 2 21 2 0.5

can be measured only for pure enzymes, the activity of an enzyme is expressed as specific activity, defined as micromoles (#mol) of substrate converted to product per minute per milligram (mg) of enzyme protein. The International Union of Biochemistry recommends use of the unit known as katal (kat); one katal is the amount of enzyme that converts one mole of substrate to product per second. In clinical disorders, the activity of a variety of enzymes is measured in biological fluids. For this purpose, the activity is usually defined as that quantity of enzyme which catalyzes the conversion of 1 #mol of substrate to product per minute under a defined set of optimal conditions. This unit, referred to as the International Unit (U), is expressed in terms of U/mL of biological specimen (e.g., serum), or U/L. 10. When [S] Product C + Product D

The enzyme-substrate interactions can proceed by either

single-displacement or double-displacement reactions (commonly known as "ping-pong" reactions). A substrate reaction proceeding by way of a single-displacement reaction can be shown by E+A~-EA

or

EA+B~-EAB

or

Enzyme inhibition is one of the ways in which enzyme activity is regulated experimentally or naturally. Most therapeutic drugs function by inhibition of a specific enzyme. Inhibitor studies have contributed much of the available information about enzyme kinetics and mechanisms. In the body, some of the processes controlled by enzyme inhibition are blood coagulation (hemostasis), blood clot dissolution (fibrinolysis), complement activation, connective tissue turnover, and inflammatory reactions. Enzyme inhibitors are reversible or irreversible.

E+B~-EB EB+A~-EAB

EAB ~- C + D Note that the ternary complex EAB can be formed in two different ways. If the formation of EAB can occur with either substrate binding first, the reaction is known as a random single-displacement reaction. Many reactions catalyzed by phosphotransferases are of this type. If a particular substrate must bind first with the enzyme before the second substrate can bind, the reaction is known as an ordered single-displacement reaction. Many reactions catalyzed by dehydrogenases are of this type. The values for Km and Vmax for each substrate can be obtained from experiments in which the concentration of one substance is held constant at saturating levels while the concentration of the second substrate is varied. Kinetic analyses can distinguish between these types of reactions. In a double-displacement reaction, at first only one substrate is bound; the release of one product and covalently modified enzyme (E*) follows. E* then combines with the second substrate to form a second product. Reactions catalyzed by aminotransferases are of this type. In this mechanism, no ternary complex EAB is formed. The double displacement reaction sequence is shown below: (Substrate 1)

(Product 2)

Reversible Inhibition In reversible inhibition, which is further subdivided into competitive, noncompetitive, and uncompetitive types, the activity of the enzyme is fully restored when the inhibitor is removed from the system (by dialysis, gel filtration, or other separation techniques) in which the enzyme functions. In reversible inhibition, equilibrium exists between the inhibitor, I, and the enzyme, E: E+I~-EI. The equilibrium constant for the dissociation of the enzyme-inhibitor complex, known as the inhibitor constant Ki, is given by the Equation Ki = [E] [I] [El] Thus, Ki is a measure of the affinity of the inhibitor for the enzyme, somewhat similar to the way in which Km reflects the affinity of the substrate for the enzyme. In competitive inhibition, the inhibitor is a structural analogue that competes with the substrate for binding at the active site. Thus, two equilibria are possible: E+S and E+I~-EI. A modified Michaelis-Menten equation that relates the velocity of the reaction in the presence of inhibitor to the concentrations of substrate and inhibitor can be derived: V

EA

(Product 1)

~- ES--~ E + P

Vmax [S] [S] + Km (1 + x~J

E*B

(Substrate 2)

In this relationship, Km is modified by a term that includes the inhibitor concentration, [I], and the inhibitor constant,

SECTION 6.4 Inhibition

93 COO -

COO -

I

C H2 I + FAD CH2

I

Succinate

dehydrogenase ,

C~ H

II

~

+ FADH2

H--C

I

I

COO-

COO-

Succinate

Fumarate

In this reaction, flavin adenine dinucleotide (FAD), a coenzyme, serves as a hydrogen acceptor. This enzyme is competitively inhibited by malonate, oxalate, or oxaloacetate, all structural analogues of succinate. COO -

coo I CH2

cooI

I c---o I

I

COO-

CH 2

I

coo-

COO-

Malonate

Oxalate

Oxaloacetate

Inhibition of Folate Synthesis Competitive inhibition of a biosynthetic step in folate synthesis accounts for the antimicrobial action of sulfonamides, which are structural analogues of p-aminobenzoic acid (PABA): F I G U R E 6-6 Effects of three types of reversible inhibitors on the velocity-substrate plots and on the Lineweaver-Burk double-reciprocal plots.

O H2N

O --OH

H2N

S--NH2

II

Ki, but Vmax is unchanged. Thus, the profiles of plots of v against [S] show the same Vmax in the presence and the absence of inhibitor but with an apparent change in Kin, to a larger value (Kmapp), because S and I compete for binding at the same site and a higher concentration of S is required to achieve half-maximal velocity. Therefore, the value of Km is increased in the presence of inhibitor and depends on the concentration of I. The Lineweaver-Burk plot for competitive inhibition shows that the lines for [S] and (IS] + [I]) intersect at the same point on the ordinate (because Vmax is unaltered) but have different slopes and intercepts on the abscissa (because of differences in Km). Figure 6-6 illustrates these plots, and Table 6-2 shows the values for apparent Kin. Competitive inhibition occurs in four different circumstances. (1) Structural analogues compete with the substrate for binding at the active site of the enzyme. Several specific examples are described below.

Inhibition of Succinate Dehydrogenase A classic example of competitive inhibition occurs with the succinate dehydrogenase reaction"

O p-Aminobenzoic acid

Sulfanilamide

PABA is used by bacteria in the synthesis of folic acid (pteroylglutamic acid), which functions as a coenzyme in one-carbon transfer reactions that are important in amino acid metabolism, in the synthesis of RNA and DNA, and thus in cell growth and division. Sulfonamides inhibit the bacterial enzyme responsible for incorporation of PABA into 7,8-dihydropteroic acid (Figure 6-7) and lead to the inhibition of growth (bacteriostasis) of a wide range of gram-positive and gram-negative microorganisms. Microorganisms susceptible to sulfonamides are those which synthesize their own folic acid or which cannot absorb folic acid derived from the host. Sulfonamides, however, have no effect on host cells (or other mammalian cells) that require preformed folic acid.

Inhibition of Dihydrofolate Reductase Folatedependent reactions in the body are inhibited by folate analogues (or antagonists, e.g., methotrexate). Before it can function as a coenzyme in one-carbon transfer reactions, folate (F) must be reduced by dihydrofolate reductase to tetrahydrofolate (FH4). Dihydrofolate

CHAPTER6 Enzymes I: General Properties, Kinetics, and Inhibition

94

TABLE 6-2 Effects o f Competitive Inhibitors on K m and Vmax (Michaelis-Menten Kinetic Parameters)

Inhibitor Type

Vmax

Competitive

Km Kmapp- K m

None

1+

( Vmaxapp - Vmax)

V

Noncompetitive

Wmaxapp =

Uncompetitive

Wmaxapp --

[E][I] [ES][I] Note: Ki= [EI] or [ESI]

max

gmax

I

II

H

T--

O

~o

,#'--~

I

COOH

II

N.,. 4 / 0 ~ .,,~.C--CH2--N - ~ ~ ~ "~C N~ [

H

I

C-- N--C--OH2CH2OOOH

CH 3

NH2

K m

In uncompetitive inhibition, the inhibitor binds equally well to E and ES.

reductase is competitively inhibited by methotrexate. Since FH4 is needed for the synthesis of DNA precursors, a deficiency causes most harm to those cells which synthesize DNA rapidly. Certain types of cancers (e.g., the leukemias) exhibit an extremely high rate of cell division and are particularly susceptible to folate antagonists. The reactions of one-carbon metabolism are discussed in Chapter 27. H2N~c//N"~'C/%C

None

N

N N

H Hypoxanthine

H AIIopurinol

Allopurinol inhibits the formation of xanthine and of uric acid, catalyzed by xanthine oxidase, and is itself transformed into alloxanthine by the enzyme. Alloxanthine bears the same structural relationship to xanthine that allopurinol does to hypoxanthine:

Methotrexate

(4-amino-Nl~

folic acid)

OH

OH

N

Inhibition of Xanthine Oxidase Uric acid, the end product of purine catabolism in humans, is formed by the serial oxidation of hypoxanthine and of xanthine, catalyzed by xanthine oxidase. Hypoxanthine

xanthine oxidase

> xanthine

xanthine oxidase

> uric acid

Allopurinol, a structural analogue of hypoxanthine, is a competitive inhibitor as well as a substrate for xanthine oxidase:

H Xanthine

H AIIoxanthine

Alloxanthine, however, remains tightly bound to the active site of the enzyme by chelation with Mo 4+. Since xanthine oxidase is a molybdenum-dependent enzyme whose catalytic cycle requires the reversible oxidation and reduction of Mo 4+ to Mo 6+ (Chapter 27), the reoxidation of Mo 4+ to Mo 6+ in the presence of alloxanthine is very

SECTION6.4 Inhibition

95 H

H,N,~N~ /N,~ ~H

Guanosine 5"triphosphate

"H~"

o

H

N

o

II

II

CH'mO--I~ mOmP(OH)'

O OH 6-Hydroxymethyldihydmptedn diphosphate fH2N @

COOH

p-Aminobenzoic acid (PABA)

H H,N,,,,,,~ N'~,,,~ N",,,,,,f H HN

N

CH,---N

COOH + PPi

O 7,8-Dihydropteroicacid fATP Glutamic acid ADP + Pi H H H ' N ~ c ~ N ~ c f N ~ c I --H

,

,,

COOH

,

- - N - - CH- - CH,CI-IzCOOH

HN~c'IC~N~C~CH2~NH II O 7,8-Dihydrofolate

NADPH + H+ Dihydrofolate NADP+ ,..,/~Jreductase H

I

H

H2N ~,-~N~-,/N ~r'/-IH ~',2 ~ 7~

/7"~'~

13 1.,.C~N,.S II 61/H _~ N~,...,C 4 jC--CH9 =m NH10 I

OH

r~ ~"

COOH

~~; mNH~ CgCH2CH~COOHH I

I

H 5,6,7,8-Tetrahydrofolate

FIGURE 6-7 Folate biosynthetic pathwaysin bacteria. The incorporation of p-aminobenzoic acid into 7,8-dihydropteroic acid is competitively inhibited by sulfanilamide.

slow, thereby affecting the overall catalytic process. Thus, in this type of inhibition, an inhibitor bearing a particular structural similarity to the substrate binds to the active site of the enzyme and, through the catalytic action of the enzyme, is converted to a reactive compound that can form a covalent (or coordinate covalent) bond with a functional group at the active site. This type of inhibition, known as mechanism-based enzyme inactivation, depends on both the structural similarity of the inhibitor to the substrate and the mechanism of action of the target enzyme. The substrate analogue is called a suicide substrate because the enzyme is inactivated in one of the steps of the

catalytic cycle. The suicide substrate, by virtue of its high selectivity, provides possibilities for many in vivo applications (e.g., development of rational drug design). Suicide substrates with potential clinical applications for several enzymes (e.g., penicillinase, prostaglandin cyclooxygenase) have been synthesized. Examples of suicide substrates harmful to the body are given in Chapter 27. Allopurinol, which affects both the penultimate and ultimate steps in the production of uric acid, is used to lower plasma uric acid levels in conditions associated with excessive urate production (e.g., gout, hematologic disorders, and antineoplastic therapy). Sodium urate has a

CHAPTER 6 Enzymes I' General Properties, Kinetics, and Inhibition

96

low solubility in biological fluids and tends to crystallize in derangements of purine metabolism that result in hyperuricemia. The crystalline deposits of sodium urate are responsible for recurrent attacks of acute arthritis or of renal colic (pain in kidney due to either stone formation or acute inflammation; see also discussion of purine catabolism in Chapter 27). (2) In two-substrate enzyme-catalyzed reactions with a double-displacement reaction sequence, high concentrations of the second substrate may compete with the first substrate for binding. For example, in the reaction catalyzed by aspartate aminotransferase, L-Aspartate + ot-ketoglularate ~_- L-glutamate + oxaloacetate, this enzyme is inhibited by excess concentrations of o~-ketoglutarate. The inhibition is competitive with respect to L-aspartate. (3) Competitive inhibition can occur in freely reversible reactions owing to accumulation of products. Even in reactions that are not readily reversible, the product can function as an inhibitor. In the alkaline phosphatase reaction, in which hydrolysis of a wide variety of organic monophosphate esters into the corresponding alcohols (or phenols) and inorganic phosphates occurs, the inorganic phosphate acts as a competitive inhibitor. Both the inhibitor and the substrate have similar enzyme binding affinities (i.e., Km and Ki are of the same order of magnitude). (4) In reactions that require metal ions as cofactors, similar metal ions can compete for the same binding site on the enzyme. For example, Ca 2+ inhibits some enzymes that require Mg 2+ for catalytic function. Pyruvate kinase catalyzes the reaction

Alcohol CH30H dehydrogenas~

HCOOH Formic acid

Major toxic effects are caused by formaldehyde and formic acid. The former is responsible for damage to retinal cells that may cause blindness, while the latter produces severe acidosis that may eventually lead to death. A minor effect of methanol is depression of the central nervous system (CNS). Retardation of the first step in the oxidation of methanol is accomplished by administration of ethanol, the oxidation products of which are not as toxic as those of methanol. Other therapeutic modalities include removal of methanol by gastric lavage (to prevent further absorption), hemodialysis (to remove absorbed methanol), and administration of exogenous bicarbonate (for treatment of severe acidosis). Ethylene glycol, which is widely used as an antifreeze for automobile radiators, upon ingestion causes depression of the CNS, metabolic acidosis, and severe renal damage. Its oxidation in the body requires the action of alcohol dehydrogenase: Ethylene glycol OH OH

I

I

H --C--C--H

I

I

H

H

NAD+ Inhibited by ethanol Q....Alcohol dehydrogenase or 4-methylpyrazole r ~ NADH + H + Glycoaldehyde OH O I II H--C--C--H

I

H Glycolic acid OH O I II H--C--C--OH

I

H

for which K + is an obligatory activator, whereas Na + and Li + are potent competitive inhibitors. Formic acid HCOOH = f

Competitive inhibition is the basis for the treatment of some intoxications (e.g., methyl alcohol, ethylene glycol). Methanol, which is widely used industrially as a solvent, is added to ethanol (ethyl alcohol) to make it unsuitable for human consumption. Such adulterated alcohol is commonly known as denatured alcohol. Methanol is metabolized primarily in the liver and kidney by oxidation to formaldehyde and formic acid:

---*

ethanol

Phosphoenolpyruvate + ADP--+ ATP + pyruvate

Competitive Substrates in Treatment of Some Intoxications

HCHO

Methanol Inhibited ~ by Formaldehyde

CO 2'~

Glyoxylic acid O O II II H--C--C--OH Oxalic acid 0 0

II

II

HO-- C -- C--OH

Kidney damage results from precipitation of oxalate crystals in the convoluted tubules. The elevated anion-gap metabolic acidosis is caused by glycolic acid and lactic acid. The latter is formed from pyruvate due to a shift in the redox potential favoring the production of lactate. The treatment is the same as that for methanol intoxication.

SECTION 6.4

97

Inhibition

4-Methylpyrazole (fomepizole), a more effective inhibitor of alcohol dehydrogenase with none of the adverse effects of ethanol, has been used in the treatment of ethylene glycol toxicity. Isopropanol, present in rubbing alcohol, hand lotions, and antifreeze preparations can be ingested accidentally by young children and intentionally by alcoholics or suicidal adults. Isopropanol is oxidized to acetone, a toxic, nonmetabolizable product, by alcohol dehydrogenase. It should be noted that acetone is also present in diabetic ketoacidosis (Chapter 22). However, diabetic ketoacidosis is accompanied by glucosuria, whereas isopropanol ingestion shows the presence of acetone without glucose. Toxicity of isopropanol includes profound CNS depression, with coma as the most common presentation, gastritis, vomiting, and hemorrhage are also common. Acetone is also a CNS depressant and can be detected in the urine. The treatment of isopropanol toxicity is supportive and is removed by hemodialysis. In evaluating individuals for toxicity to substances such as methanol, ethylene glycol, and isopropanol, the following serum components are measured: Na +, K +, CI-, HCO 3, glucose, urea nitrogen, blood osmolality, blood gases, and urine ketones. The severity of the osmolality gap, anion gap, and metabolic acidosis (Chapter 39), along with pertinent clinical history, will aid in the identification of the offending substance. In particular, the assessment of osmolality gap, which represents the difference between the measured serum osmolality and the calculated osmolality is useful in the diagnosis of low-molecularweight toxic substances. Elevated serum levels of lowmolecular-weight toxic chemicals contribute to serum osmolality significantly. Serum osmolal gap -- measured osmolality -

calculated osmolality

Calculated osmolality = 2 x Na + (mM/L) + glucose (mM/L) + urea nitrogen (mM/L). In noncompetitive inhibition, the inhibitor does not usually bear any structural resemblance to the substrate, and it binds to the enzyme at a site distinct from the substrate binding site. No competition exists between the inhibitor and the substrate, and the inhibition cannot be overcome by increase of substrate concentration. An inhibitor may bind either to a free enzyme or to an enzyme-substrate complex; in both cases, the complex is catalytically inactive: E + I ~ EI (inactive) ES § I ~-- ESI (inactive)

The value of Vmax is reduced by the inhibitor, but Km is unaffected because the affinity of S for E is unchanged. Thus, comparison of the double-reciprocal plots, with and without the presence of inhibitor (Figure 6-6), shows that both the slope and the intercept on the y axis (1 / Vmax) are altered, whereas the x intercept ( - 1 / K m ) is unchanged. Examples of noncompetitive inhibition are 1. Enzymes with sulfhydryl groups (-SH) that participate in maintenance of the three-dimensional conformation of the molecule are noncompetitively inhibited by heavy metal ions (e.g., Ag +, Pb 2+, and Hg2+): E-SH + Hg 2+ ~- E - S - H g + + H + Lead poisoning causes anemia (low levels of hemoglobin) owing to inhibition of heme synthesis at two sites at least: porphobilinogen synthase and ferrochelatase, both of which contain sulfhydryl groups (Chapter 29). Heavy metal ions react with S-containing, O-containing, and N-containing ligands, present as - O H , - C O O - , - O P O 3 H - , - C = O , -NH2, a n d - N H groups. 2. Enzymes that are dependent on divalent metal ion (e.g., Mg 2+ and Ca 2+) for activity are inhibited by chelating agents (e.g., ethylenediamine tetraacetate) that remove the metal ion from the enzyme. 3. Enolase catalyzes a step in the metabolism of glucose (see the discussion of glycolysis in Chapter 13), 2-Phospho-D-glycerate ~ phosphoenolpyruvate + H20 a reaction that has an absolute requirement for a divalent metal ions (e.g., Mg 2+ or Mn 2+) complexed to the enzyme before the substrate is bound. This reaction is inhibited by fluoride ion (F-) in a process involving the formation of a complex with phosphate giving rise to a phosphofluoridate ion that binds magnesium ions. Thus, addition of fluoride ions inhibits the breakdown of glucose in the glycolytic pathway. For this reason, F - is used as a preservative in clinical specimens (e.g., blood) in which glucose determinations are to be made. In uncompetitive inhibition, inhibitor, I, combines with ES to form an enzyme-substrateinhibitor complex: ES + I ~ ESI The double-reciprocal plot with the inhibitor yields parallel lines (i.e., the slope remains constant), but the intercepts on both the x and y axes are altered by the presence of the inhibitor (Figure 6-6). The apparent Vmax and the apparent Km are both divided by a factor

98

CHAPTER6 Enzymes I: General Properties, Kinetics, and Inhibition

of (1 + [I] /Ki) (Table 6-2). Uncompetitive inhibition is rarely observed in single-substrate reactions. A noteworthy example in clinical enzymology is the inhibition of intestinal alkaline phosphatase by L-phenylalanine. Uncompetitive inhibition is more common in two-substrate reactions with a double-displacement reaction mechanism.

H3C~H~CH3 C I O I Enzyme-CH2-OH + FBP---O Active seryl I residue O I C

/H\

H3C

Irreversible Inhibition

CH3

Diisopropylphosphofluoridate

Irreversible inhibition occurs when the inhibitor reacts at or near the active site of the enzyme with covalent modification of the active site or when the inhibitor binds so tightly that, for practical purposes, there is no dissociation of enzyme and inhibitor. The latter situation occurs in the case of proteinase inhibitors (see below). Thus, physical separative processes are ineffective in removing the irreversible inhibitor from the enzyme. Irreversible inhibitor reaction is written E + I ~ E1 (inactive enzyme) Examples of irreversible inhibitors of enzymes are: 1. Enzymes that contain free sulfhydryl groups at the active site (e.g., glyceraldehyde-3-phosphate dehydrogenase; see Chapter 13) react with an alkylating reagent, iodoacetic acid, resulting in inactivation of the enzyme. Enzyme-SH + ICH2COOH --+ Iodoacetic acid

enzyme-S - CH2COOH + HI Inactive covalent derivative of enzyme

The imidazole ring of histidine also undergoes alkylation on reaction with iodoacetate. In ribonuclease, two residues (His 12 and His 119) are alkylated with loss of activity when the enzyme is treated with iodoacetate at pH 5.5. 2. Enzymes with seryl hydroxyl groups at the active sites can be inactivated by organophosphorous compounds. Thus, diisopropylphosphofluoridate (DPF) inactivates serine hydrolases by phosphorylation at the active site: A specific example is inactivation of acetylcholinesterase (Table 6-1), which catalyzes hydrolysis of acetylcholine to acetate and choline. Acetylcholine is a neurotransmitter, a chemical mediator of a nerve impulse at a junctionmknown as a synapse--between two neurons or between a neuron and a muscle fiber. On arrival of a nerve impulse at the ending of the neuron, acetylcholine (which is stored in the vesicles of the presynaptic nerve terminal) is released. The released

(DPF)

~""~ HF H3C

\H/

CH3

C I O I Enzyme--CH2BOmP--O I O I C

/H\

H3C

CH3

acetylcholine acts on the postsynaptic membrane to increase the permeability of Na + entry across the membrane. Depolarization results in the inside of the membrane becoming more positive than the outside; normally, the inside of the membrane is more negative than the outside. This process may propagate an action potential along a nerve fiber, or it may lead to contraction of a muscle (Chapter 21). Acetylcholine is quickly destroyed by acetylcholinesterase present in the basal lamina of the neuromuscular junction (Figure 6-8). If, however, acetylcholine is not destroyed, as in the case of inactivation of acetylcholinesterase by DPF, its continued presence causes extended transmission of impulses. In muscle fibers, continuous depolarization leads to paralysis. The cause of death in DPF intoxication is respiratory failure due to paralysis of the respiratory muscles (including the diaphragm and abdominal muscles). Several organophosphorous compounds are used as agricultural insecticides, improper exposure to which can result in toxic manifestations and death. Knowledge of the mechanisms of action of acetylcholinesterase and of the reaction of organophosphorous compounds with esterases led to the development of drugs useful in the treatment of this kind of intoxication. The active site of acetylcholinesterase consists of two subsites"

SECTION6.4 Inhibition

FIGURE 6-8 The neuromuscular junction. (a) Connection between a nerve and a muscle fiber. (b) Invagination of a nerve terminal into the muscle fiber. The acetylcholine receptor-rich domains are located on the crest of the fold.

an esteratic site and a site consisting of a negative charge (Figure 6-9). The esteratic site contains a seryl hydroxyl group whose nucleophilicity toward the carbonyl carbon of the substrate is enhanced by an appropriately located imidazole group of histidine that functions as a general base catalyst. A side-chain carboxyl group that is suitably located apparently functions to hold the imidazole and imidazolium ion in place. The substrate is positioned on the enzyme so that its positively charged nitrogen atom is attracted to the negatively charged active site of the enzyme by both coulombic and hydrophobic forces. The acyl carbon of the substrate is subjected to nucleophilic attack by the oxygen atom of the serine hydroxyl group. This catalytic mechanism is similar to that of other serine hydrolases (esterases and the proteinases, i.e., chymotrypsin, trypsin, elastase, and thrombin). Although they have dif-

99

ferent functions, these enzymes have a common catalytic mechanism, which supports the view that they evolved from a common ancestor. The inactive phosphorylated acetylcholinesterase undergoes hydrolysis to yield free enzyme, but the reaction is extremely slow. However, a nucleophilic reagent (e.g., hydroxylamine, hydroxamic acids, and oximes) can reactivate the enzyme much more rapidly than spontaneous hydrolysis. Wilson and co-workers accomplished the reactivation by use of the active-site-specific nucleophile pralidoxime. This compound, oriented by its quaternary nitrogen atom, brings about a nucleophilic attack on the phosphorus atom, leading to the formation of an oxime-phosphonate-enzyme complex that dissociates into oxime-phosphonate and free enzyme (Figure 6-10). Pralidoxime has found use in the treatment of organophosphorous poisoning and is most active in relieving the inhibition of skeletal muscle acetylcholinesterase. The phosphorylated enzyme can also lose an isopropoxy residue; such an enzyme-inhibitor complex has been named an "aged" enzyme. The aged enzyme is resistant to regeneration by pralidoxime because the phosphorus atom is no longer an effective center for nucleophilic attack. The severity and duration of toxicity of organophosphorous compounds depend on their lipid solubility, the stability of the bond linking the phosphorus atom to the oxygen of the serine hydroxyl group, and the ease with which the aged enzyme complex is formed. Some organophosphorous compounds are so toxic that they can be fatal within a few seconds of exposure. Other manifestations (e.g., excessive mucus secretion and bronchoconstriction) are reversed by administration of atropine, which does not relieve enzyme inactivation but, by binding to some of the acetylcholine receptor sites, renders ineffective the accumulated acetylcholine. Organophosphorous compounds have been used in the identification of functional groups essential for catalysis. Another approach to identification of functional amino acid residues is affinity labeling. The labeling is produced by a synthetic substrate-like reagent designed to form a covalent linkage with some amino acid residue at or near the active site of the enzyme. After the labeling, the enzyme is subjected to classic techniques of degradative protein chemistry (Chapter 3) to determine the amino acid sequence of the structurally altered portion of the enzyme, which is inferred to be the active site.

Inactivation and Reactivation of Cytochrome Oxidase Cyanides are among the most rapidly acting toxic substances. Cyanide (CN-) inhibits cellular respiration to

CHAPTER6

100

EnzymesI: GeneralProperties,Kinetics,and Inhibition

H-- o, Anionic

~Histidine Serine,

site Esteraticsite Acetylcholinesterase Acetylcholine H3C~

_CH2

H3C~~CH3

O OH2

+

Choline

~_.j~-~N~--~ H,

H C\ CH,\

-O-

.~N.~ CH2 H3Cf +~CH3

C~CH 3 ?j-------

I

k-C-~-~ N'~.

Enzyme-substrate intermediate

C~ I CH3 ?J "

Acetylated enzyme Acetic acid

HOmC~cH 3 kSf--'-~N :~-~.-.H -- ?j

Regenerated enzyme FIGURE 6-9 Hydrolysis of acetylcholine by acetylcholinesterase.

cause tissue hypoxia by binding to the trivalent iron of cytochrome oxidase, a terminal component of the mitochondrial electron transport chain. This chain consists of electron-transferring proteins and other carriers arranged sequentially in the inner mitochondrial membrane. The reducing equivalents, obtained from a variety of substrates, are passed through the electron transport system to molecular oxygen with the formation of water and energy (Chapter 14). Thus, cyanide severely impairs the normal energy-generating functions of mitochondria by inhibition of mitochondrial respiration, leading to cell death, particularly affecting the central nervous system. Death in acute cyanide poisoning is due to respiratory failure. Cytochrome oxidase is a multienzyme complex that contains oxidation-reduction centers of iron-porphyrin prosthetic groups as well as centers of copper ion. Cyanide has a higher affinity for the oxidized form of cytochrome oxidase than for the reduced form. CN- probably forms a loose complex with Fe 2+ of porphyrin. When Fe 2+ is oxidized to Fe 3+, the latter forms a stable complex with CN-. This complex cannot be reduced, thus preventing electron flow and uptake of O2. Cyanide and cyanide precursors occur widely in nature. Foods that contain moderate to high levels of cyanogenic

glycosides include cassava (a dietary staple in several regions of Africa), kernels of some fruits (peach, cherry, plum, and apricot), lima beans, sorghum, linseed, sweet potato, maize, millet, and bamboo shoots. Amygdalin (Figure 6-11) is one of the principal cyanogenic glycosides of dietary origin. Hydrogen cyanide is released when amygdalin undergoes enzymatic hydrolysis in the gastrointestinal tract: C20H27NOll-+- 2H2 ~ Amygdalin

2C6H1206-4- C6HsCHO Glucose

+

Benzaldehyde

HCN

Hydrogencyanide

The toxicity of amygdalin is directly related to the release of hydrogen cyanide. The enzymes that catalyze the hydrolysis of amygdalin are supplied by the microbial flora of the intestine, which probably explains why amygdalin is many times more toxic when taken by mouth than when given intravenously. Amygdalin and the related compound laetrile have been at the center of a controversy regarding their efficacy as anticancer agents. Their anticancer activity is claimed to depend on selective hydrolysis at the tumor site by/3-glucuronidase or fl-glucosidase, with local release of HCN to cause cell death. Presumably

SECTION6.4 Inhibition

101

HO... //0 P

H,qO~ ~ ~ " "

,9

~o

...........+H20

~OqH,

H--~# l

Regenerated

enzyme

k..~.,)~-~NL Phosphorylated enzyme

,Oj I

~o

%

P

+H20 . , ~ "Aged"enzyme ~N~+.~ ? ~N--OH

I

I

CH3 Pralidoxime

H3C/N +

Oxime-phosphonate

C-~-N--O~i CH_N_ HTC30

~OC3H7

I*

H,C30

OC3H,

CH3

+

k.~

. ~ N . ~ " H - - ,C)j

I

I

Complex

Regenerated

enzyme

F I G U R E 6-10 Reactivation of alkylphosphorylated acetylcholinesterase. Spontaneous reactivation (dashed arrow) by water occurs at an insignificant rate; however, loss of one isopropoxy group occurs at a much more rapid rate, yielding the "aged" enzyme, which is resistant to reactivation. The regeneration of the enzyme by pralidoxime is shown at bottom.

normal cells are not affected by HCN because they contain CN--inactivating enzyme (rhodanese; see below). No objective evidence demonstrates any therapeutic value for these compounds as anticancer agents. Their ingestion has led to severe toxicity in some reported cases. Nondietary sources of cyanide include sodium nitroprusside (a hypotensive agent), succinonitrile (an antidepressant agent), acrylonitrile (used in the plastic industry and as a fumigant to kill dry-wood termites), and tobacco smoke. Chronic exposure to cyanogenic compounds leads to toxic manifestations such as demyelination, lesions of the optic nerves, ataxia (failure of muscle coordination), and depressed thyroid functions. This last effect arises from accumulation of thiocyanate, the detoxified product of cyanide in the body (see below). Thiocyanate inhibits the active uptake of iodide by the thyroid gland and, therefore, the formation of thyroid hormones (Chapter 33). Acute cyanide poisoning requires prompt and rapid treatment. The biochemical basis for an effective mode of treatment consists of creating a relatively nontoxic porphyrin-ferric complex that can compete effectively

with cytochrome oxidase for binding the cyanide ion. This is accomplished by the administration of nitrites (NaNO2 solution intravenously, and amylnitrite by inhalation), which convert a portion of the normal oxygen-carrying hemoglobin with divalent iron to oxidized hemoglobin (i.e., methemoglobin with trivalent iron, which does not transport oxygen). Methemoglobin binds cyanide to form cyanomethemoglobin, whose formation is favored because of an excess of methemoglobin relative to cytochrome oxidase. This process may lead to the restoration of normal cytochrome oxidase activity (Figure 6-12). Cyanomethemoglobin is no more toxic than methemoglobin and can be removed by normal processes that degrade erythrocytes and by the reaction catalyzed by rhodanese. Rhodanese (thiosulfatecyanide sulfurtransferase) catalyzes the reaction involving cyanide and thiosulfate to form thiocyanate: SSO~- + enzyme ~-- SO~- + Thiosulfate

Sulfite

Enzyme-S + CN-

(Cyanide)

enzyme-S Sulfur-substituted enzyme

--> SCN- + enzyme Thiocyanate

102

CHAPTER6 Enzymes I" General Properties, Kinetics, and Inhibition O

CH2OH HH

~

H H

o

HH

H

Oxyhemoglobin [Hb202(Fe2+)] ~ Nitrites(NO~)

CH2

OH

H I

Reducedform of nitrites Methemoglobin [MetHbOH(Fe3+)] ~Cyanide complex [Cytaa3(Fe3+)CN ] of cytochrome 0xidase

Hf~ CN

H

OH

H

Cytochrom.# oxidase [Cytaa3(FdT)] Cyanomethemoglobin [MetHbOH(Fe")CN]

Amygdalin ( D-mandelonitrile-13-D-glucosido-6-13-D-glucoside) COOH O

/--Thiosulfate ($2032-)

CN

Rhodanese ~"*Sulfite (SO~-) Methemoglobin + thiocyanate (SCN-)

C,H,

H

F I G U R E 6-12 Reactivation of cytochrome oxidase and inactivation of cyanide.

OH

Laetrile (1-mandelonitrile-13-glucuronicacid) CH2= CHCN Acrylonitrile

order to decrease the amount of cyanide available for binding with cytochrome oxidase.

NCCH2CH2CN Succinonitrile

2-

NO

"'''-

~176~176176176176176

(Na+)2

-Fe3+ , -..,....~

NC

_

i i i

"CN

CN-

Sodiumnitroprusside, Na2Fe(CN)sNO F I G U R E 6-11 Structures of some cyanogenic compounds.

Rhodanese is present in the mitochondria, particularly of liver and kidney cells. A double-displacement mechanism has been proposed for its biochemical action. The steps are as follows: The free enzyme reacts with a sulfane sulfur-containing compound (a sulfane sulfur is one that is divalent and covalently bonded only to other sulfurs), cleaving the S-S bond of the donor substrate (e.g., SSO 2-) to form the sulfur-substituted enzyme. The latter reacts with the cyanide (a thiophilic acceptor) to form thiocyanate in an essentially irreversible reaction. Nitrite administration has been augmented by thiosulfate administration (intravenously) in the treatment of cyanide poisoning. Cobalt-containing compounds (e.g., cobalt chloride and cobalt ethylenediaminetetraacetate) have also been used to form complexes with cyanide, in

Proteinase Inhibitors and Their Clinical Significance

In the body, enzymes are compartmentalized and function under highly restricted conditions. Some enzymes (e.g., proteinases) are not substrate-specific. When present in active form in an inappropriate part of the body, they act indiscriminately and cause considerable damage to the tissue. Inhibitors inactivate these enzymes at sites where their action is not desired. Proteinase inhibitors, which are themselves proteins, are widely distributed in intracellular and extracellular fluids. Protein inhibitors of enzymes other than proteinases are relatively rare. Such inhibitors are available for or-amylases, deoxyribonuclease I, phospholipase A, and protein kinases. Many proteinase inhibitors are present in blood plasma and participate in the control of blood coagulation (Chapter 36), dissolution of blood clots (Chapter 36), activation of the complement cascade (Chapter 35), formation and destruction of some peptide hormones, and inactivation of proteinases released from phagocytic cells (Table 6-3). The proteinase inhibitors rapidly combine with their target enzymes to form stable complexes that are practically nondissociable. Inhibition occurs through binding of the reactive site (a substrate-like region) of the inhibitor to the active site (the substrate binding region) of the proteinase. At or near the center of the reactive site of the inhibitor, a specific amino acid residue recognizes the primary binding site of the target proteinase. Some of the

SECTION 6.4

103

Inhibition

TABLE 6-3

Principal Proteinase Inhibitors in Human Blood Plasma*

Name c~1-Proteinase inhibitor 1-Antichymotrypsin Inter-a-trypsin inhibitor az-Antiplasmin Antithrombin III C 1-inactivator az-Macroglobulin

Concentration (mg/lO0 mL) 290_+45 49 _+7 50 7_+ 1 24 _+2 24_+ 3 260_+ 70

Mol. Wt.

Number of Polypeptide Chains

Heads t

52,000 69,000 160,000 70,000 65,000 70,000 720,000

1 1 1 1 1 1 4

1 1 2 1 1 1 1-2 or more

*Reproduced, with permission, from M. Lakowski, Jr., and I. Kato: Protein inhibitors of proteinases. Annu. Rev. Biochem., 49, 593 (1980). 9 1980 by Annual Reviews, Inc. tMaximal number of enzyme molecules simultaneously inhibited by one inhibitor molecule.

proteinase inhibitors share significant homology at their reactive site regions. Neutrophils and macrophages function protectively against foreign organisms in an inflammatory process. In this process, a number of proteinases escape to surrounding tissues. One such enzyme is elastase, which catalyzes the hydrolysis of elastin, a protein of connective tissue (Chapter 10). Normally, the activity of elastase is inhibited by the c~l-proteinase inhibitor (also known as 1-antitrypsin), which inhibits a broad spectrum of proteinases containing serine in their active sites (e.g., elastase, trypsin, and chymotrypsin). Genetic deficiency of an o~1-proteinase inhibitor strongly predisposes to pulmonary emphysema and liver disease. Emphysema (which means swelling or inflation) results from the breakdown of alveolar walls due to coalescence of alveoli resulting in formation of air sacs, which leads to a gradual decrease in the effectiveness of CO2 elimination and oxygenation of hemoglobin. A collection of high-molecular-weight proteinase inhibitors with broad specificity, known as o~2macroglobulins, are present in the plasma of all mammals and exhibit an interesting inhibition pattern. When o~2-macroglobulins combine with a wide variety of proteinases, they inhibit only the proteolytic activity of the enzyme toward large protein substances without significantly affecting the catalysis of low-molecular-weight substrates. Proteinases and their inhibitors play a major role in metastasis of cancer. Metastasis of tumors requires remodeling of extracellular matrix (ECM). Remodeling is a balance between proteolysis and respective proteinase inhibitors. This process is aided by proteolytic enzymes

synthesized by the tumor cells. Examples of proteolytic enzymes include serine proteinases, cathepsins, and matrix metalloproteinases (Table 6-4). Some of the cathepsins are cysteine proteinases and their inhibitors belong to the cystatin superfamily. The cystatin family consists of three subfamilies: stefins, cystatins, and rinogens. Progressive loss of expression of the proteinase inhibitors may be responsible for metastasis. The loss of control over proteinase expression and their respective inhibitors is influenced by a variety of biological response modifiers such as growth factors, cytokines, tumor promoters, and suppressor genes. Viral proteinases (also called proteases) offer unique targets for antiviral drugs. An example is in the treatment of human immunodeficiency virus (HIV) infections by HIV protease inhibitors (Figure 6-13). HIV is a retrovirus and its RNA undergoes reverse transcription in cells to produce double stranded DNA (Chapter 26). This step is inhibited by nucleoside analogues such as zidovuline, didanosine, zalcitabine, stavudine, and lambivudine. The viral DNA, upon integration into the host genome, produces a protease as well as polypeptides. The HIV protease is an aspartyl protease and the enzymatic activity resides in the homodimers. The HIV protease cleaves polypeptides at phenylalanine-proline or tyrosine-proline bonds, which are unusual sites of cleavage for mammalian proteases. That cleavage results in the production of three large proteins (p24, p17, and p7) and three smaller proteins (p6, p2, and p 1). The three large proteins are essential for RNA packaging and in the structure of the virion. Viral protease is essential in the production of functionally mature and infectious particles and thus is a prime target for inhibition by drugs.

104

CHAPTER 6

Enzymes I: General Properties, Kinetics, and Inhibition

TABLE 6-4 Proteinases and Their Putative Extracellular Matrix Targets

Proteinases

Functions/Targets of Degradation

Serine proteinases Urokinase plasminogen activator (uPA) Tissue-type plasminogen activator (tPA) Plasminogen Plasmin Thrombin Elastase

Activation of plasminogen Activation of plasminogen

Cathepsins

Collagens, laminin

Cathepsin Cathepsin Cathepsin Cathepsin

Laminin, type IV collagen

D (aspartic proteinase) B, L (cysteine proteinases) G (serine proteinase) 0 2 (similar to cathepsins S and L)

Integral membrane proteinases Matrix metalloproteinases (MMP)

Localized ECM degradation

MMP- 1 interstitial collagenase Neutrophil collagenase MMP-2 (type IV collagenase, gelatinase) (two major forms) MMP-9 (type IV collagenase) MMP-3 (Stromelysin- 1) MMP- 10 (Stromelysin-2, transin) MMP-I 1 (Stromelysin-3) MMP-4 MMP-5 MMP-6 Acid metalloproteinase MMP-7 (pump-l)

Degradation of interstitial collagen types I, II, III, VII Collagen I Types I, II, III, IV, V, VII collagens, fibronectin Types IV, IX collagens, laminin, fibronectin Types III, IV, V collagens, fibronectin a 1-chain of type I collagen Native 3/4 collagen fragments Cartilage proteoglycan Gelatin of types I, III, IV, V collagens, fibronectin

Reproduced with permission from The Genetics of Cancer, G. V. Sherbet and M. S. Lakshmi.Academic Press, 1997.

6.5 Kinetics of Ligand-Receptor Interaction The kinetics of enzyme-substrate interactions can be applied to ligand-receptor relationships. A ligand (e.g., a hormone or a drug) can saturate the receptor sites as well as dissociate from them, similar to enzyme-substrate interactions. Nonspecific ligand binding, on the other hand, shows a relatively low affinity to the receptor and nonsaturation kinetics. In ligand-receptor binding, no product is formed, but effects responsible for the ligand's biological action are produced (Chapter 30). Cells that contain a receptor protein specific for a particular hormone are targets for the action of that hormone. The activity of a ligand on a target cell depends on the concentration of ligand, the binding affinity of the ligand to the receptor sites, the number of receptors bound, and the duration of binding. Ligands that compete for binding to a receptor at a specific site and produce the same biological

response are known as agonists. A ligand that competes with an agonist for binding to a receptor and causes a blockage or inhibition of the biological response of the cell is known as an antagonist. Antagonists are similar to competitive inhibitors of enzymes. Consider a ligand-receptor interaction, L + R ~ LR

(6.8)

where L is the ligand, R is the free receptor site, and LR is the ligand bound to the receptor site. We apply the law of mass action to the above equilibrium, [LR] Ka -- [L] [R]

(6.9)

where Ka is the association constant. The reciprocal of Ka represents the dissociation constant known as Kd. Thus, Ka =

1 Kd

or Kd --

1 Ka

SECTION6.6

Mechanismsof EnzymeAction

~

Indinavir

N"~

105

OH

OH ~,,,PhHi

*HzSO4 N/:%0

0

Q 0 S. Nelfinavir

~-}

0% i

NHtBu , CH=SO=H

H OH F I G U R E 6-14 Scatchard plot of ligand-receptor interaction.

N

H

OH

Ph

H

CH=

S

Ritonavir Ph

Saquinavir

~NTNT~o H= OT ~ ~ Or" I aN~/ ~ Na""~""~= H 7C~H 0 H LL~ M [ H ? * CH=SOzH

lilO H ~ 0 '- F ' ~ NH'

~ ....O,,~,,N.,yi-,,,./N-.s

Amprenavir

~oJ

~ph)

0 @ %0

F I G U R E 6-13 Structures of HIV protease inhibitors. NHtBu denotes an amino-tertiary butyl and Ph denotes a phenyl group.

The total number of receptor binding sites, [TR], is equal to the sum of the unbound receptor sites, [R], and the bound receptor sites, [LR]. Therefore, [R] - - [ T R ] -

[LR]

(6.10)

Substituting for [R] in Equation (6.8) yields [LR] Ka = [L] ([TR] - [LR])

(6.11)

which on rearrangement gives [LR] ILl

= Ka ([TR] - [LR])

(6.12)

of the line with the abscissa yields the value for the total number of sites [TR]. Comparing this relationship to the Michaelis-Menten analysis of enzyme kinetics shows that 1/ Ka (where 50% of the sites are occupied) is equivalent to Km and the maximum concentration (or number) of bound ligand sites is equivalent to Vma• The Scatchard plot is similar to the Eadie-Hofstee plot (Figure 6-5), but the axes are reversed. These principles have been applied in the assay of ligands in human biological fluids by the use of either specific receptor proteins (radioreceptor assay) or specific binding proteins, namely, antibodies (radioimmunoassay (RIA)). In these assays, the unlabeled ligand (the quantity of which is to be determined) competes with a predetermined amount of added radioactive ligand to bind a limited amount of specific receptor protein. This step is followed by separation of protein-bound ligand from the unbound (free) ligand and measurement of radioactivity of each fraction. Separation of the protein-bound ligand from the free ligand is accomplished by protein precipitation, adsorption of the free form, chromatography, or electrophoresis. Under standard conditions of time and temperature, the amount of radioactivity found in the protein-bound ligand fraction is inversely proportional to the concentration of unlabeled ligand and the amount of radioactivity found in the unbound (free) ligand fraction is directly proportional to unlabeled ligand concentrations. This procedure has been widely used for the assay of hormones, vitamins, drugs, and other compounds. Since the concentrations of hormones in body fluids are very low (10 -8 to 10 -12 M), assay sensitivity is increased by ligands with high specific radioactivity.

where [LR] [L]

concentration of bound ligand concentration of unbound ligand

A plot of [LR]/[L] versus [LR] yields a straight line and is known as a Scatchard plot (Figure 6-14). The slope of the line gives the value (-Ka), while the intersection

6.6 Mechanisms of Enzyme Action The mechanism of a reaction catalyzed by an enzyme is a detailed description of the chemical interactions occurring among the substrates, enzymes, and cofactors.

106

For example, although the overall reaction catalyzed by acetylcholinesterase consists of hydrolysis of acetylcholine to choline and acetic acid, the detailed mechanism is a two-step displacement reaction in which an alcohol (choline) is produced first, followed by an acid (acetic acid). The same is true for the rhodanese-catalyzed reaction. The types of experiments that are carried out to arrive at the description of an enzyme mechanism are as follows.

Isotope Exchange If a particular atom of the substrate or product is labeled with an isotope other than the natural one, its location at various intermediate reaction steps can be followed, providing information on what is happening to that part of the molecule. Irreversible Inhibition Studies If an enzyme is inactivated by a reagent that reacts with a specific amino acid residue, then one (or more) of the reagent-specific amino acid residues is involved in catalysis by that enzyme. pH Dependence Usually only one form (protonated or unprotonated) of an acidic or basic residue is involved in a catalysis. Structural studies, particularly those that elucidate the three-dimensional relationships of the amino acid residues in the enzyme, permit judgments about whether the involvement of one or the other side chain is physically and sterically possible. Such techniques include nuclear magnetic resonance, electron spin resonance, single-crystal x-ray diffraction, and cross-linking studies. The important question to be answered by mechanistic studies is how an enzyme catalyzes a particular reaction so rapidly. Two general factors seem to be involved. 1. Because all the reactants are bound to a small region on the enzyme, their effective concentrations are greatly increased; i.e., the adjacent binding makes it easier for an effective collision to occur between reactants. 2. As a reaction proceeds, the conformation of the reacting molecules must change from that of the substrates to that of the products. Alterations in bonds, bond lengths, and bond angles are involved. The activation energy represents, at least in part, the work needed to bring about this transformation. The conformation that lies between those of the substrates and products and that has the highest energy is termed the transition state. Additionally, the binding energy of an enzyme distorts the substrates, so that they approach or attain the conformation of the transition state. Detailed structures and catalytic mechanisms are known for many enzymes, some of which are

CHAPTER6 Enzymes I: General Properties, Kinetics, and Inhibition

discussed elsewhere (e.g., lysozyme in Chapter 11). Serine proteinases are among the most thoroughly studied enzymes, and they share similar catalytic mechanisms (see above). However, they act on different substrates because of differences in their substrate binding sites. Chymotrypsin, trypsin, and elastase have the same amino acid sequence around the active serine residue (*), -Gly-Asp-Ser*-Gly-Gly-Proand their tertiary structures, as revealed by x-ray studies, are similar. Their amino acid sequences are 40% identical, leading to the hypothesis that these three enzymes may have evolved from a common ancestral gene. Duplication of this gene probably occurred several times during evolution and, with mutation, eventually yielded genes that now code for these three proteinases, each with its own substrate specificity. This type of evolution is known as divergent evolution. Chymotrypsin and subtilisin provide an example of independent convergent evolution. Subtilisin is a serine proteinase of bacterial origin whose mechanism of action is similar to that of chymotrypsin, but it differs from chymotrypsin in the amino acid sequence around the active serine residue: -Gly-Thr-Ser*-Met-Ala-SerSubtilisin -Gly-Asp-Ser*-Gly-Gly-ProChymotrypsin Chymotrypsin and subtilisin also differ in their amino acid sequences, number of disulfide bridges (chymotrypsin has five, whereas subtilisin has none), and overall three-dimensional structures. The striking difference in structure and common catalytic mechanism are taken as evidence of an independent but convergent evolutionary process. Coenzymes, Prosthetic Groups, and Cofactors Many enzymes require the presence of low-molecularweight nonprotein molecules. These small molecules may be bound to the enzyme by a covalent, tight, or noncovalent linkage. Prosthetic groups are usually bound by covalent or tight linkages and coenzymes by noncovalent linkages. Cofactors usually are metal ions or organic compounds (some are vitamins, e.g., ascorbic acid). Prosthetic groups and coenzymes are complex organic compounds, many of which are derived from vitamins. These compounds are recycled and are needed only in catalytic amounts to convert a large amount of reactants to products. Coenzymes function as substrates in

SECTION 6.6

Mechanisms of EnzymeAction

two-substrate reactions, being bound only momentarily to the enzyme during catalysis. They are chemically altered during the reaction and are reconverted to their original forms by the same or another enzyme (e.g., NAD +- or NADP+-dependent dehydrogenase reactions). Prosthetic groups form part of the active center and undergo cyclic chemical changes during the reaction (e.g., pyridoxal phosphate in aminotransferases). Principal coenzymes, prosthetic groups, and cofactors are listed below. Their metabolic roles are discussed elsewhere, e.g., with the reactions they catalyze and the vitamins from which they are derived. 1. Compounds involved in H-transfer reactions and derived from vitamin precursors. a. Nicotinamide adenine dinucleotide (NAD+), also known as diphosphopyridine nucleotide (DPN+). The reduced form is NADH (DPNH). b. Nicotinamide adenine dinueleotide phosphate (NADP+), also known as triphosphopyridine nucleotide (TPN +). The reduced form is NADPH (TPNH). These compounds are derived from the vitamin niacin (nicotinic acid, nicotinamide) and require it for their synthesis. Small amounts of niacin are derived from the essential amino acid tryptophan. c. Flavin mononucleotide (FMN). The reduced form is FMNH2. d. Flavin adenine dinucleotide (FAD). The reduced form is FADH2. These compounds are derived from the vitamin riboflavin. They are not true nucleotides, because the flavin ring system is attached to ribitol, a sugar alcohol, rather than ribose. 2. Compounds involved in H-transfer reactions and derived from nonvitamin precursors. a. Lipoic acid apparently can be synthesized in sufficient quantity by humans. It is also involved in acyl group transfer during oxidative decarboxylation of o~-keto acids. b. Biopterin (a pteridine-containing compound) participates in certain hydroxylase reactions (e.g., phenylalanine hydroxylase). c. Coenzyme Q (CoQ, ubiquinone) is a group of closely related compounds differing only in the length of the side chain. They can be synthesized in humans from farnesyl pyrophosphate, an intermediate in cholesterol biosynthesis. Although the structure of coenzyme Q is similar to that of vitamin K1, no interrelationship has been established. 3. Compounds involved in group transfer reactions and derived from vitamin precursors.

107

a. Coenzyme A (CoA, CoASH) takes part in acetyl and other acyl group transferase and requires the vitamin pantothenic acid for its synthesis. b. Thiamine pyrophosphate (TPE cocarboxylase) is used for oxidative decarboxylation of ot-keto acids and in the transketolase-catalyzed steps of the pentose phosphate pathway. It is derived from thiamine (vitamin B 1). c. Pyridoxalphosphate is a cofactor for a wide variety of types of reactions on amino acids, namely, racemization, decarboxylation, transamination, elimination of water or hydrogen sulfide. It is derived from pyridoxine, pyridoxal, and pyridoxamine (collectively called vitamin B6). d. Tetrahydrofolic acid (FH4) is a carrier of one-carbon fragments such as formyl, methylene, methyl, and formimino groups. It is derived from folic acid (folacin). e. Biotin is vitamin tightly bound to the apoenzyme in an amide linkage to the s-amino group of a lysyl residue and involved in carboxylation reactions. f. Cobamide coenzyme (5,6-dimethylbenzimidazole cobamide 5'-dehydroadenosyl cobalamine) contains cobalt bound in a porphyrin-like ring system and is unusual in that it contains a cobalt-carbon (organometallic) bond. It is involved in methyl-transfer reactions and participates in the 1,2 shift of a hydrogen atom accompanied by a 2,1-shift of an alkyl, carboxyl, hydroxyl, or amino group. Very small amounts, derived from cyanocobalamin (vitamin B12), are required by the body. g. Vitamin K participates in carboxylation reactions of glutamic acid residues of some proteins involved in blood coagulation and its regulation to yield v-carboxyglutamic acid residues required for Ca 2+ binding. The vitamins K are derived from naphthoquinones with isoprenoid side chains of varying lengths. h. Vitamin C participates as a cofactor in the hydroxylation reactions of proline and lysine of collagen. 4. Compounds involved in group transfer reactions and derived from nonvitamin precursors. a. Adenosine triphosphate (ATP) can be a donor of phosphate, adenosine, and adenosine monophosphate (AMP) for various purposes. The exergonic hydrolysis reaction of ATP provides the energy for driving thermodynamically unfavorable reactions.

108

b. Cytidine diphosphate (CDP) is a carrier of phosphorylcholine, diacylglycerols, and other molecules during phospholipid synthesis. c. Uridine diphosphate (UDP) is a carrier of monosaccharides and their derivatives in a variety of reactions (see bilirubin, lactose, galactose and mannose metabolism, glycogen synthesis, and other pathways). d. Phosphoadenosine phosphosulfate (PAPS, "active sulfate") is a sulfate donor in the synthesis of sulfur-containing mucopolysaccharides as well as in the detoxification of sterols, steroids, and other compounds (see metabolism of the sulfur-containing amino acids in Chapter 17). PAPS is derived from ATP and inorganic sulfate. e. S-Adenosylmethionine (SAM, "active methionine") is a methyl group donor in biosynthetic reactions. It is formed from ATP and the essential amino acid methionine. f. Heme proteins, containing the iron-protoporphyrin group, participate in oxygen transport (hemoglobin), oxygen storage (myoglobin), electron transport (cytochromes), hydrogen peroxide inactivation (catalase, peroxidase), hydroxylases, oxygenases, and other processes. 5. Metal cofactors. a. M g 2+ is required by most enzymes that use ATE The active form of ATP is a MgZ+-ATP 4complex. b. Ca 2+ is involved in a wide variety of processes, notably muscle contraction, blood clotting, nerve impulse transmission, and cAMP-mediated processes. c. FeZ+/Fe 3+ ~errous/ferric iron) is required as heme (see above) or nonheme iron in the functioning of many proteins. d. Cu+/Cu 2+ (cuprous~cupric copper). Cytochrome oxidase in mitochondrial electron transport contains Fe and Cu. Tyrosinase and lysyl oxidase also require Cu. Superoxide dismutase contains both Cu and Zn. e. Zn 2+ (zinc ion) is used by lactic acid dehydrogenases, alcohol dehydrogenases, carbonic

CHAPTER6 Enzymes I: General Properties, Kinetics, and Inhibition

anhydrase, carboxypeptidase A, DNA polymerase, superoxide dismutase, and others. f. M o 6+ (molybdenum ion). Xanthine oxidase contains Mo and Fe. g. Mn 2+ (manganous ion) is required by acetyl-CoA carboxylase, deoxyribonuclease (Mg 2+ can replace Mn 2+ in this case), arginase, and other enzymes. h. Se (selenium) is required for the functioning of glutathione peroxidase. Metal cofactors in enzymes may be bound reversibly or firmly. Reversible binding occurs in metal-activated enzymes (e.g., many phosphotransferases); firm (or tight) binding occurs in metalloenzymes (e.g., carboxypeptidase A). Metals participate in enzyme catalysis in a number of different ways. An inherent catalytic property of a metal ion may be augmented by the enzyme protein, or metal ions may form complexes with the substrate and the active center of the enzyme and promote catalysis, or metal ions may function in electron transport reactions between substrates and enzymes.

Supplemental Readings and References J. Brent, K. McMartin, S. Phillips, et al.: Fomepizole for the treatment of ethylene glycol poisoning. New England Journal of Medicine 340, 832, (1999). K. K. Burkhart and K. W. Kulig: The other alcohols. Methanol, ethylene glycol, and isopropanol. Emergency Medicine Clinics of North America 8, 913 (1990). W. D. N. Chin, R. Barnett, and G. Sheehan: Methanol poisoning. Intensive Care Medicine 18, 391 (1992). C. Flexner: HIV-protease inhibitors, New England Journal of Medicine 338, 1281 (1998). HIV-1 protease inhibitors: A review for clinicians. Journal of American Medical Association 277, 145 (1997). G. V. Sherbet and M. S. Lakshmi: The Genetics of Cancer, Academic Press, San Diego (1997). P. Houeto, J. R. Hofman, M. Imbert, P. Levillain, and E J. Baud: Relation of blood cyanide to plasma cyanocobalamin concentration after a fixed dose of hydroxycolalamin in cyanide poisoning. Lancet 346, 605 (1995). D. Jacobsen: The treatment for ethylene glycol poisoning. New England Journal of Medicine. 340, 879 (1999). H. A. James and I. Gibson: The therapeutic potential of ribozymes, Blood 91, 371 (1998). J. A. Kruse and P. Cadnapapnornchai. The serum osmole gap. Journal of Clinical Care 9, 185 (1994).

CHAPTER

7

Enzymes II: Regulation

The metabolic rate of key substances, which can proceed in multiple pathways, is regulated and integrated. A close interrelationship exists among products formed by different metabolic pathways from a common metabolite. For example, glucose can be consumed either by oxidation to CO2 or by conversion to glycogen, lipid, nonessential amino acids, or other sugar molecules. The glucose supply of the body can be derived either from the diet or from the breakdown of glycogen, a polymer of glucose (primarily from the liver and the kidney), or it can be synthesized from some amino acids or lactate (predominantly in the liver). These processes of glucose utilization and synthesis are under tight regulation. In fact, the plasma glucose level is maintained at the level at which tissues (e.g., brain, erythrocytes, kidney medulla, and the lens and cornea of the eye) that require glucose as a primary substrate are not deprived of this essential fuel. Each of these metabolic pathways is mediated by enzymes that are unique for a given pathway and that are under control. Metabolic pathways do not usually compete with each other for utilization of a substrate, and they operate only to serve a particular physiological need or function.

7.1 Types of Regulation A metabolic pathway involves many enzymes functioning in a sequential manner or in some unique arrangement to

carry out a particular metabolic process. Control of a pathway is accomplished through modulation of the activity of only one or a few key enzymes. These regulatory enzymes usually catalyze the first or an early reaction in a metabolic sequence. A regulatory enzyme catalyzes a rate-limiting (or rate-determining) chemical reaction that controls the overall pathway. It may also catalyze a chemical reaction unique to that pathway, which is known as a committed step. In the metabolic pathway for the formation of E from A, A -~ B -~-~ C -~ D -~ E the conversion of A to B, catalyzed by the enzyme El, is the rate-limiting step and also a committed step. The ratelimiting step need not be the same as the committed step. In the branched metabolic pathway, E1 A

E2 ~B

E3

E4

~ C ------* D

L

E6

*E

E7 *M

~-N

if the conversion of A to B is the rate-limiting step, the committed step in the pathway for the formation of N is the conversion of B to L (B --+ L), catalyzed by the enzyme E5. Those enzymes which catalyze the rate-limiting step or the committed step of a pathway are under regulation. When the end product exceeds the steady-state level concentration, it inhibits the regulatory enzyme in an

109

110

attempt to normalize the overall process. This type of control, known as feedback inhibition (see below), ensures a high degree of efficiency in the utilization of materials and of energy in living systems. Regulation may be achieved in other ways. The absolute amount of a regulatory enzyme may be altered through mechanisms that control gene expression (Chapter 26). This regulation at the genetic level occurs during various phases of reproduction, growth, and development, with different metabolic pathways being turned on or off in accordance with the special requirements of each phase. In eukaryotic cells, regulation at the genetic level is relatively a long-term process. Several short-term regulatory mechanisms control metabolic activity rapidly (see below). Both substrates and some hormones play significant roles in regulating the concentration of key enzymes at this level. Many drugs or other chemicals can increase levels of enzymes that affect their own metabolism. Thus, phenobarbital and polycyclic hydrocarbons increase the levels of microsomal enzyme systems involved in their metabolism. Regulation of metabolic processes can be accomplished by other methods. One is the use of a multienzyme complex (e.g., pyruvate dehydrogenase complex or fatty acid synthase complex) in which various enzymes are organized such that the product of one becomes the substrate for an adjacent enzyme. A single polypeptide chain may contain multiple catalytic centers that carry out a sequence of transformations (e.g., the mammalian fatty acid synthase; see Chapter 18). Such multifunctional polypeptides increase catalytic efficiency by abolishing the accumulation of free intermediates and by maintaining a stoichiometry of 1:1 between catalytic centers. Another type of regulation is accomplished by a series of proenzymes in which activation of the initial proenzyme by a biological signal activates the second proenzyme which, in turn, activates the third, and so on. Such an enzyme cascade process provides great amplification in terms of the amount of final product formed. Examples are blood coagulation, the dissolution of blood clots, complement activation, and glycogen breakdown. Regulation is also accomplished by compartmentalization of enzyme systems involved in anabolic and catabolic pathways into different cell organelles. For example, fatty acid synthesis occurs in the soluble fraction of the cytoplasm, whereas fatty acid oxidation takes place in mitochondria. Heme synthesis begins and is completed in mitochondria, but some of the intermediate reactions take place in the cytosol. Heme catabolism is initiated in the smooth endoplasmic reticulum. Transport of key metabolites across an organelle membrane system is also a form of regulation.

CHAPTER7 Enzymes I1: Regulation

Many enzymes occur in several molecular forms called

isoenzymes (or isozymes), and those which are genetically determined may be called primary isoenzymes. The different primary isoenzymes catalyze the same chemical reaction but may differ in their primary structure and kinetic properties. The tissue distribution of isoenzymes imparts distinctive properties and specific patterns of metabolism to organs of the body. The presence of isoenzymes may reveal differences not only between organs but also between cells that make up an organ or between organelles of a cell. During different stages of differentiation and development from embryonic life to adulthood, the isoenzyme distribution in an organ undergoes characteristic changes. When an adult organ reverts to the embryonic or fetal state (e.g., in cancer), the isoenzyme distributions change to those characteristic of that developmental state. The existence of isoenzymes in human tissues has important implications in the study of human disease. Zymogen (e.g., trypsinogen and chymotrypsinogen) synthesis, secretion, transport, and activation and the rate of inactivation of the active enzyme by inhibitors may all be considered means of enzyme regulation. Enzyme activity can be regulated by covalent modification or by noncovalent (allosteric) modification. A few enzymes can undergo both forms of modification (e.g., glycogen phosphorylase and glutamine synthetase). Some covalent chemical modifications are phosphorylation and dephosphorylation, acetylation and deacetylation, adenylylation and deadenylylation, uridylylation and deuridylylation, and methylation and demethylation. In mammalian systems, phosphorylation and dephosphorylation are most commonly used as means of metabolic control. Phosphorylation is catalyzed by protein kinases and occurs at specific seryl (or threonyl) residues and occasionally at tyrosyl residues; these amino acid residues are not usually part of the catalytic site of the enzyme. Dephosphorylation is accomplished by phosphoprotein phosphatases: ATP

ADP O

Mg2+

Enzyme-Ser-OH ~,.

f Phosphoprotein-"~ phosphatase O H20

II

Enzyme-Ser-O--P--O-

I

O-

II

HO--P--O-

I

O-

The overall process of phosphorylation and dephosphorylation is a cascade of reactions and consists of an extracellular signal, commonly referred to asfirst messenger (e.g., hormones, such as glucag0n); a specific receptor on the cell membrane of the target cell; a transducer coupled to the

SECTION 7.2

AIIostericEnzymeRegulation

receptor that produces an intracellular signal (i.e., a second messenger); one or more modifying enzymes whose activity is affected by the intracellular signal; a target enzyme, which is the substrate for the modifying enzyme and subjected to covalent modification with consequent metabolic alteration; and an enzyme that reverses the modification of the target enzyme. The transducer of the system is adenylate cyclase, and it catalyzes the cyclization of ATP to cyclic AMP (cAMP), the second messenger. The modifying enzyme is a protein kinase activated by cAME Other protein kinases are activated by non-cAMPdependent or CaZ+-dependent systems (Chapter 30). Phosphorylation of a target enzyme may be either stimulatory or inhibitory. For example, phosphorylation converts glycogen phosphorylase to an activated form and glycogen synthase to an inactivated form, thus preventing the simultaneous occurrence of glycogen breakdown and synthesis (Chapter 15).

7.2 AIIosteric Enzyme Regulation Those enzymes in metabolic pathways whose activities can be regulated by noncovalent interactions of certain compounds at sites other than the catalytic are known as allosteric enzymes. They are usually rate-determining enzymes and play a critical role in the control and integration of metabolic processes. The term "allosteric" is of Greek origin, the root word "allos" meaning "other." Thus, an allosteric site is a unique region of an enzyme other than the substrate binding site that leads to catalysis. At the allosteric site, the enzyme is regulated by noncovalent interaction with specific ligands known as effectors, modulators, or modifiers. The properties of allosteric enzymes differ significantly from those of nonregulatory enzymes. Ligands (in some instances even the substrate) can bind at such sites by a cooperative binding process. Cooperativity describes the process by which binding of a ligand to a regulatory site affects binding of the same or of another ligand to the enzyme. Allosteric enzymes have a more complex structure than nonallosteric enzymes and do not follow Michaelis-Menten kinetics. An allosteric site is specific for its ligand, just as the active site is specific for its substrate. Binding of an allosteric modulator causes a change in the conformation of the enzyme (see below) that leads to a change in the binding affinity of the enzyme for the substrate. The effect of a modulator may be positive (activatory) or negative (inhibitory). The former leads to increased affinity of the enzyme for its substrate, whereas the reverse is true for the latter. Activatory sites and inhibitory sites are separate and specific for their

111 respective modulators. Thus, if an end product of a metabolic pathway accumulates in excess of its steadystate level, it can slow down or turn off the metabolic pathway by binding to the inhibitory site of the regulatory enzyme of the pathway. As the concentration of the end product (inhibitor) decreases below the steady-state level, the number of enzymes having bound inhibitor decreases and they revert to their active form. In this instance, the substrate and the negative modulator bear no structural resemblance. An allosteric enzyme may be positively modulated by the substrate itself or by a metabolite of another pathway that depends on production of the end product in question for its eventual utilization (e.g., pathways of synthesis of purine and pyrimidine nucleotides in the formation of nucleic acids; see Chapter 27). Most allosteric enzymes are oligomers (i.e., they consist of two or more polypeptide chains or subunits). The subunits are known asprotomers. Two types of interaction occur in allosteric enzymes: homotropic and heterotropic. In a homotropic interaction, the same ligand influences positively the cooperativity between different modulator sites. An example is a regulatory enzyme modulated by its own substrate. Thus, this class of enzyme has at least two substrate binding sites which respond to situations that lead to substrate excess by increasing its rate of removal. Heterotropic interaction refers to the effect of one ligand on the binding of a different ligand. For example, a regulatory enzyme modulated by a ligand other than its substrate constitutes a heterotropic system, in which the cooperativity can be positive or negative. Some allosteric enzymes exhibit mixed homotropic and heterotropic interactions.

Kinetics of Allosteric Proteins The kinetic properties of allosteric enzymes vary significantly from those of nonallosteric enzymes, exhibiting cooperative interactions between the substrate, the activator, and the inhibitor sites. These properties are responsible for deviations from the classic Michaelis-Menten kinetics that apply to nonallosteric enzymes. Nonallosteric enzymes yield a rectangular hyperbolic curve when the initial velocity (v) is plotted against the substrate concentration [S]. For allosteric enzymes, a plot of v versus [S] yields curves of different shapes, including sigmoidshaped curves in some cases. (A sigmoidal curve can result from other mechanisms.) The v versus [S] plot for a homotropic enzyme is shown in Figure 7-1. The following features should be noted: 1. The substrate functions as a positive modulator; i.e., there is positive cooperativity between the substrate binding sites so that binding of the substrate at one

112

CHAPTER7 Enzymes I1: Regulation

FIGURE 7-1 Relationship between the initial velocity (v) and the substrate concentration [S] for an allosteric enzyme that shows a homotropic effect. The substrate functions as a positive modulator. The profile is sigmoidal, and during the steep part of the profile, small changes in [S] can cause large changes in v. K0.5 representsthe substrate concentration corresponding to half-maximalvelocity.

FIGURE 7-2 Relationship between the initial velocity (v) and the substrate concentration [S] for an allosteric enzyme that shows a heterotropiceffect with constant Vmaxbut with varying K0.5. Curve a is obtained in the absence of any modulators,curve b in the presence of a positive modulator, and curve c in the presence of a negativemodulator. Regulation is achieved by modulation of K0.5 without change in Vmax.

binding site greatly enhances binding of the substrate at the other sites. As the substrate concentration increases, there is a large increase in the velocity of the reaction. 2. Owing to the above effect, the shape of the curve is sigmoidal. 3. The value of the substrate concentration corresponding to half-maximal velocity is designated as K0.5 and not Km since the allosteric kinetics do not follow the hyperbolic M i c h a e l i s - M e n t e n relationship. 4. M a x i m u m velocity (Vmax) is attainable at a rather high substrate concentration, implying saturation of the catalytic site of the enzyme.

can be turned on or off with appropriate modulators. Figure 7-3 shows v versus [S] plots for allosteric enzymes modulated by changes in Vmax but retaining an essentially constant K0.5. This type of modulation is less c o m m o n than the two previous cases considered. The positive modulator increases gmax(curve b), whereas the negative modulator decreases Vmax (curve c).

We will consider two specific examples of allosteric p r o t e i n s m o n e an e n z y m e and the other an oxygen transport protein.

The v versus [S] plot for heterotropic enzymes is more complex. The kinetic profiles can be divided into two classes, depending upon whether the allosteric effector alters K0.5 and maintains a constant Vmax or alters Vmax and maintains a nearly constant K0.5. The v versus [S] profile of an allosteric e n z y m e that follows the former set of properties is shown in Figure 7-2. In the absence of any modulators, the profile is sigmoidal (curve a). In the presence of a positive modulator (curve b), the value for K0.5 is decreased; i.e., a lower substrate concentration is required to attain half-maximal velocity. Curve b is more hyperbolic than sigmoidal. Curve c obtained with a negative modulator is more sigmoidal than curve a, and the K0.5 value is increased, reflecting a decreased affinity for the substrate, i.e., a higher substrate concentration is required to attain half-maximal velocity. Regulation of the e n z y m e is achieved through positive and negative modulators. Thus, at a given substrate concentration (e.g., steady-state level), the activity of the e n z y m e

FIGURE 7-3 Relationship between the initial velocity (v) and the substrate concentration [S] for an allosteric enzyme that shows a heterotropiceffect with constant K0.5 but with varying Vmax.Curve a is obtained in the absence of any modulator, curve b in the presence of a positive modulator, and curve c in the presence of a negativemodulator. Regulationis achieved by modulation of Vmaxwithout change in K0.5.

E x a m p l e s of A l l o s t e r i c P r o t e i n s

SECTION7.2 AIIosteric Enzyme Regulation

113

1. Aspartate transcarbamoylase (ATCase) is an allosteric enzyme of the bacterium Escherichia coli, which has been extensively studied. This enzyme catalyzes the transfer of the carbamoyl group from carbamoyl phosphate to the u-amino group of aspartate:

CO2 +

Glutamine +

ATP

Carbamoyl phosphate Aspartate---~l N- Carbamoyl aspartate

O

O

II

II

H

+

I

3

I

E m.

H2NmC--O--P~O- + H3N~C~COO-

I

O-

CH2

I

Uridine monophosphate (UMP)

[

O

,4

COO Carbamoyl phosphate

Aspartate

Uddine triphosphate (UTP)

o --4 "o

O

II

HO~I~OOInorganic phosphate (P~ O

H

II

I

H2N--C--NmC--COO- + H + H I CH2

I

COO N-Carbamoyl aspartate

This committed reaction, catalyzed by a regulatory enzyme, is the second step (Figure 7-4) in the biosynthesis of pyrimidines. N-carbamoylaspartate ultimately is converted to the pyrimidine nucleotide cytidine triphosphate (CTP), which is the negative modulator of the enzyme. That is, as the CTP concentration increases, it inhibits the ATCase by decreasing its affinity for substrates. The preceding is an example of an allosteric feedback inhibition. ATCase has a molecular weight of 310,000 and consists of 12 polypeptide chains (subunits). The catalytic and the regulatory sites reside on two different sets of subunits: six larger subunits (M.W. 33,000) have catalytic sites, and six smaller subunits (M.W. 17,000) have regulatory sites. X-ray crystallographic studies of ATCase by Lipscomb showed that the molecule is roughly triangular. The six catalytic subunits (C) are present as two trimers (C3), one above the other (but not exactly parallel), with the dimeric regulatory subunits (R2) forming an equatorial belt around the edges of the trimers (Figure 7-5). In the center of the molecule, an aqueous cavity is accessible through several openings. ATCase can be dissociated into two unequal clusters with different subunit arrangements by treatment with mercurials (e.g., p-hydroxymercuribenzoate). The larger cluster containing three catalytic subunits (C3)

Cytidine triphosphate (CTP)

F I G U R E 7-4 Regulation of pyrimidine nucleotide biosynthesis in E. coli.

is catalytically active. The v versus [S] plot exhibits a hyperbolic profile and is insensitive to CTE The smaller cluster is a dimer of regulatory subunits (R2) and, as expected, shows no catalytic activity but does bind to CTE The native enzyme can be reconstituted with return of the allosteric activity by removal of the mercurial compound and mixing of both catalytic and regulatory clusters: 2C3 -k3R2 --+ R6C6 Catalytic Regulatory Native cluster cluster enzyme

Reconstitution also requires zinc ions (Zn2+), since the native enzyme contains six atoms of Zn 2+. C3 and R2 clusters can be dissociated further into their respective, inactive monomeric subunits by strong denaturing agents (Figure 7-5). ATCase can assume different conformations depending on whether it is active or inactive. In the presence of the substrate or positive modulator, it is in a catalytically more active conformation known as the relaxed state or R-state. In the presence of the negative modulator, the enzyme is in less active conformation known as the taut state or T-state. The allosteric kinetic effects of ATCase are shown in Figure 7-6. The interaction of substrates with the enzyme is cooperative (an example of homotropic cooperativity), as indicated by the sigmoidal shapes of the v versus [S] plots, CTP being an inhibitor and ATP an activator. These modulators compete for the same regulatory site and modulate the affinity of the enzyme for its

114

CHAPTER7 Enzymes I1: Regulation

Hg 2+

(

+3

Native enzyme

Active catalytic subunit

Active regulatory su]~unit I SDS

0 Inactive catalytic subunit

o Inactive regulatory subunit

(a)

~

Substrates

'""J

~

CTP

R state

T state

(6) F I ( ; U R E 7-5 Schematic representation of the subunit structure of aspartate transcarbamoylase and its dissociation into catalytic and regulatory subunits by mercurials, which can be further converted to inactive monomeric subunits by strong denaturing agents (e.g., sodium dodecyl sulfate). The native enzyme consists of two catalytic trimers placed one above the other, along with three dimeric regulatory subunits surrounding the catalytic trimers in an equatorial plane (a). Substrate maintains the enzyme in the catalytically more active relaxed (R) conformation, while cytidine triphosphate maintains it in the catalytically less active taut (T) conformation (b). [Reproduced, with permission from E. L. Smith, R. L. Hill, I. R. Lehman, et al: Principles of Biochemistry: General A.wects, 7th ed. McGraw-Hill, New York, 1983].

substrates, without altering Vmax. The biological significance of the activation by the purine nucleotide ATP can be appreciated, since both ATP and CTP are eventually utilized in the biosynthesis of double-helical DNA, which contains equal amounts of purines and pyrimidines. Thus, modulation of ATCase activity equalizes the rates of formation of purine and pyrimidine nucleotides. Other regulatory and metabolic aspects of purines and pyrimidines are discussed in Chapter 27. 2. Hemoglobin binding of oxygen is a classic example of the homotropic effect. Hemoglobin also shows heterotropic effects with specific molecules in its environment. These effects are intimately related to the function of hemoglobin as a carrier not only of oxygen but of H + and CO2 (Chapters 1 and 28). The heterotropic modulators are H +, CO2, and

F I G U R E 7-6 Relationship between the initial velocity (v) of aspartate transcarbamoylase and the substrate concentration. Note that ATP is a positive allosteric modulator, which causes decreased K0.5, whereas CTP is a negative allosteric modulator, which causes increased K0.5.

SECTION7.2 Allosteric Enzyme Regulation

FIGURE 7-7 Profiles of oxygenationof myoglobinand hemoglobinas a function of partial pressure of oxygen.Myoglobinshows a typicalMichaelis-Menten type of rectangularhyperbolicsaturationcurve, whereashemoglobin shows a sigmoidalsaturationcurve, consistentwith its allostericproperties. Myoglobinat any partial pressure of oxygenhas muchhigher affinityfor oxygen than does hemoglobin. [Reproducedwith permissionfrom A. Lehninger,Principles of Biochemistry. Worth,New York, 1982.]

2,3-bisphosphoglycerate (BPG or DPG). The cooperativity of oxygen binding to hemoglobin and the alterations of hemoglobin by various ligands provide the most extensively investigated molecular regulation of a biological process. Hemoglobin carries oxygen from the lungs to the tissues and carries CO2 and H + back from the tissues to the lungs (Chapter 1), whereas myoglobin functions as an oxygen store in muscle. Consistent with its function, myoglobin has a higher affinity for oxygen at any partial pressure of oxygen than does hemoglobin (Figure 7-7). Thus, oxygen can be transferred easily from hemoglobin to myoglobin. Hemoglobin is a tetramer consisting of two different subunit types (e.g., ot and/3 in hemoglobin A). Each polypeptide contains one heme group (an iron-porphyrin prosthetic group) that binds to one oxygen molecule by a cooperative process. Myoglobin, a monomeric protein with one heme group, remains monomeric under a wide range of concentrations and does not show cooperative binding with oxygen. The polypeptides of myoglobin and hemoglobin exhibit many differences with respect to their primary structures. For example, the many amino acid residues present on the surface of

115

myoglobin are polar, whereas many of those in the individual hemoglobin polypeptide chains are capable of participating in nonpolar interactions with other subunits. Despite these differences, critical regions are conserved in the polypeptide chains of myoglobin and hemoglobin, namely, the proximal and distal histidyl residues that interact with the heme iron, the hydrophobic amino acid residues that surround the heme group, and certain prolyl residues that interrupt the helical regions to allow the chain to fold back upon itself. The region of the polypeptide chain in contact with the heme group is known as the heme pocket. The amino acid residues in this pocket maintain the heme iron in the divalent state, which is the functional oxidation state of iron in both myoglobin and hemoglobin. Thus, the single polypeptide chain of myoglobin and the two different chains of hemoglobin are remarkably similar in secondary and tertiary structures (Figure 7-8). These similarities support the hypothesis that myoglobin and hemoglobin evolved by gene duplication and subsequent mutation from a common ancestral oxygen-binding heme protein. The binding of oxygen to myoglobin is not cooperative, but the binding of oxygen to hemoglobin is cooperative. This difference can be accounted for kinetically by considering the equilibrium for dissociation of oxymyoglobin (MbO2) to deoxymyoglobin (Mb) and oxygen (O2): MbO2 ~- Mb + 02

(7.1)

The equilibrium constant, Kd --

[Mb][O2] [MbO2]

(7.2)

is expressed in moles per liter, and its value depends on pH, ionic strength, and temperature. Since myoglobin has a single oxygen binding site, a single equilibrium defines the dissociation of oxymyoglobin. So that we can deal with measurable parameters, Equation (7.2) needs to be modified by the introduction of two terms, Y and Ps0. Y is defined as the fractional saturation of myoglobin, e.g., when Y = 0.3, 30% of the available sites on the myoglobin are occupied by oxygen. Thus, Y

number of binding sites occupied by 02 total number of binding sites available for binding 02

or Y=

[MbO2] [Mb] + [MbO2]

(7.3)

116

CHAPTER7 Enzymes I1 Regulation

[F$ try{

NAI d

13-Chain of hemoglobin Myoglobin FIGURE 7-8 The remarkable similarity in the conformations of myoglobin and of the/3-chain of hemoglobin. [Reproduced with permission from A. Fersht, Enzyme Structure and Mechanism. (W. H. Freeman, New York, 1977.]

Substituting Equation (7.2) into Equation (7.3), we obtain Y =

[02] [02] -']- Kd

(7.4)

Equation (7.8) is known as the Hill equation. A plot of l o g ( Y / 1 - Y) versus log Po2 yields a straight line with a slope of 1 (the Hill coefficient) (Figure 7-10). Thus, a value

Because 02 is a gas, it is convenient to express [02] in terms of its partial pressure in units of Torr (or m m Hg; multiply by 0.133 to obtain kilopascals). Therefore, y =

Po2 Po2 + Kd

(7.5)

Now we can substitute for Kd the term Ps0, which is defined as the partial pressure of oxygen at which 50% of the sites are occupied (i.e., when Y -- 0.5), because this situation is analogous to the M i c h a e l i s - M e n t e n treatment of e n z y m e kinetics. y =

Po2

(7.6)

Po: +/'50 A plot of Y versus Po2 yields an oxygen saturation profile that is a rectangular hyperbola (Figure 7-9), indicating that the binding of oxygen to myoglobin is noncooperative. Equation (7.6) can be rearranged to yield a linear plot as follows: Y 1 -

= Y

Po2 Ps0

(7.7)

Taking the logarithms of both sides of Equation (7.7) yields log

1 -Y Y ) -- log Po2 - l o g / 5 0

(7.8)

FIGURE 7-9 Profiles of fractional saturation of myoglobin and of hemoglobin with oxygen as a function of partial pressure of oxygen. Under physiological conditions,/50 for myoglobin is only 1 or 2 torr, whereas for hemoglobin it is 26 torr, indicating that oxygen is bound much more tightly to myoglobin than to hemoglobin. The loading and unloading to oxygen are cooperative in the case of hemoglobin but not cooperative in the case of myoglobin.

SECTION 7.2

AIIosteric EnzymeRegulation

117 Rearrangement of Equation (7.14) yields

Y

~.(eo2~ n

1 -Y

\-~5o/

The Hill equation is obtained by taking the logarithm of both sides: log

FIGURE 7-10 Hill plots for myoglobin and hemoglobin. A slope of 1.0 for myoglobin is consistent with noncooperative oxygen binding, whereas a slope of 2.8 for hemoglobin is indicative of cooperative oxygen binding.

of 1 for the Hill coefficient is indicative of noncooperativity; as we will see with hemoglobin, values greater than 1 indicate positive cooperativity, and values less than 1 indicate negative cooperativity. A molecule of hemoglobin (Hb) can bind four molecules of oxygen, and therefore four equilibrium expressions can be written to describe the dissociations of HbO2, Hb(O2)2, Hb(O2)3, and Hb(O2)4. For simplicity, we consider an equilibrium in which Hb(O2)4 dissociates into deoxyhemoglobin and four molecules of oxygen:

Hb(O2)4 ~- Hb + 402

(7.9)

The equilibrium constant for Reaction (7.9) is" K --

[Hb] [02] 4

[Hb (O2)4]

(7.10)

Based upon considerations similar to those for myoglobin dissociation, the value for Y is Y =

(Po2) 4 (P02)4 _Jr_(P50) 4

(7.11)

A plot of Y versus /~ yields a sigmoidal curve, indicating cooperative binding of oxygen to hemoglobin. A general expression for the dissociation of oxyhemoglobin to deoxyhemoglobin and oxygen may be written as

Hb(O2)n ~-- Hb + nO2

(7.12)

where n is the number of molecules of oxygen. The equilibrium constant for reaction (7.12) is K --

[Hb] [O21n [Hb (O2)n]

(7.13)

and Y =

(/'02)" (P02) n --]--(P50) n

(7.14)

1-Y

--nlogPo2-nlogPs0

A plot of log ( Y / 1 - Y) versus log Po2 yields a straight line with a slope of n, the Hill coefficient. For hemoglobin, n = 2.8 (Figure 7-10), which signifies that the binding of oxygen to hemoglobin exhibits positive cooperativity. From a physiological point of view, the cooperative oxygen binding characteristics of hemoglobin are eminently suited for the transport of oxygen from the lungs to the tissues. In the alveolar spaces of the lungs, the partial pressure of oxygen is about 100 Torr, and about 97% of the hemoglobin is combined with oxygen (i.e., 97% saturation with 02). As the oxygenated blood passes through the tissue capillaries, where the partial pressure of oxygen often falls below 40 Torr (in actively exercising muscle, the Poe is about 20 Torr), about 30% of the oxygen is unloaded from oxyhemoglobin to tissue cells. This process is cooperative, so that as 02 is released from oxygen-saturated hemoglobin because of a drop in Po2, the loss of a single 02 molecule causes rapid release of the remaining ones. Similarly in the lungs, the affinity of hemoglobin for binding the first 02 molecule is low; however, once this molecule is bound, the affinity increases. Myoglobin, in conformity with its storage function, has higher affinity for oxygen than does hemoglobin at any partial pressure of oxygen (Figure 7-9). X-ray studies have shown that oxyhemoglobin (R form) and deoxyhemoglobin (T form) have different threedimensional conformations. However, no changes in the tertiary structure of the individual subunits have been observed. The molecular mechanisms of cooperative binding of oxygen are known, and the details of this process along with other ligand interactions with hemoglobin are discussed in Chapter 28.

Theoretical Models for Allosteric Effect Two theoretical models for allosteric effects have been proposed to explain the mechanism for ligand-protein cooperative interactions: the concerted (or symmetry) model of Monod, Wyman, and Changeux and the sequentially inducedfit model of Koshland. The nomenclature associated with allosterism and cooperativity originated from the concerted model. Both models assume that 1. Each subunit of an oligomeric protein exists in two forms, T and R, which bind the ligand with low and high affinity, respectively; and 2. The T ~-- R transformations involve noncovalent bonds and result in changes in the quaternary structure of the enzyme.

118

CHAPTER 7

EnzymesI1 Regulation

Catalyticsiteswith low affinityf.orsubstrate

Catalytic sites with low affinity for substrate

AIIosteric

AIIosteric

site

site

eAiiosteric..___.~~ t is

~

"~

~,,_._.__ AIIosteric site

TT state TTstate Positive

modulator (or substrate in positive homotropic cooperativity)preferentially bindsto the R form and maintains the allosteric protein in the RR conformation. Catalytic sites with high affinity for substrate

modulator preferentially binds to the T form and maintains the allosteric protein in the TT conformation,

Negative

Negative

modulator

Positive

modulator

Ne0atve cooperativity

Postve coope rativity

TR state Negative

modulator

Positive

modulator

RR state FIGURE 7-11 Schematic diagram of conformational changes of concerted model for a dimeric allosteric enzyme. All subunits are either in the T-form with low affinity for the substrate or in the R-form with high affinity for the substrate.

The concerted model has the following features. The allosteric protein exists only in two states: T or R. If, for example, an allosteric protein contains two subunits, the sole permissible conformation states are TT and RR (RT is not allowed because R and T cannot form a stable pair). Thus, in this model (also called the "all or none" model), symmetry of the allosteric protein is preserved (Figure 7-11). It is assumed that the T and R forms are in equilibrium, that significant binding of ligand occurs only to the R form, and that this binding shifts the equilibrium strongly in favor of formation of the R form. Thus, a conformational change in one subunit that occurs after it binds to a ligand causes a corresponding change in all of the subunits without formation of hybrid species. The concerted model accounts for the kinetic behavior of many allosteric proteins, but it cannot account for negative cooperativity; i.e., the decrease in affinity for a ligand by the allosteric protein as the sites become occupied. The sequential model proposes that as the ligand binds to a subunit a conformational change is induced that stabilizes the ligand-allosteric protein complex such that a second ligand molecule is bound more readily than the first (Figure 7-12). This effect is of positive cooperativity. However, if the initial binding of the ligand results in a ligandprotein complex with decreased stability, the binding of additional ligand molecules, or ligand-induced substrate binding, becomes increasingly difficult. This effect is negative cooperativity. In this model, the notion of symmetry is discarded, since a conformational change induced by a ligand that binds to a subunit also induces a conformational change in an adjacent subunit. Figure 7-13 shows sequential transmission of conformational changes through contiguous subunits upon

RR state FI(;URE 7-12 Schematic diagram of conformational changes of sequentially induced-fit model for a dimeric allosteric enzyme. The TT conformation is progressively converted to the RR conformation via the intermediateTR conformation through cooperative interaction in the presence of the positive modulator. In the presence of the negative modulator, the opposite conformational changes occur. In this model, the notion of symmetryis discarded and the concept of induced fit is emphasized.

ligand binding. The behavior of some allosteric proteins is best explained by the concerted model, whereas for others the sequential model is more appropriate. For many proteins, neither model is satisfactory and a more complex model may be required.

T form

R form

m=m=FF 88---88

FIGURE 7-13 Sequential transmission of conformational changes through contiguous subunits induced by a ligand (S). In idealized form, the conformational changes proceed diagonally as the protein progressivelybinds ligand.

119

Supplemental Readings and References

Supplemental Readings and References Enzyme Regulation (General) Z. Dische: The discovery of feedback inhibition; Trends in Biochemical Sciences 1, 269 (1976). D. E. Koshland, G. Nemethy, and D. Filmer: Comparison of experimental binding data and theoretical models in proteins containing subunits; Biochemistry 5, 365 (1966). J. Monod, J.-E Changeux, and E Jacob: Allosteric proteins and cellular control systems. Journal of Molecular Biology 6, 306 (1963). J. Monod, J. Wyman, and J.-E Changeux: On the nature of allosteric transitions: A plausible model. Journal of Molecular Biology 12, 88 (1965). H. L. Segal: Enzymatic interconversions of active and inactive forms of enzymes. Science 180, 25 (1973). G. Weber, (Ed): Advances in Enzyme Regulation, Vols. 1-6. Pergamon Press, London, 1963-1979.

Allosteric Properties of Aspartate Transcarbamoylase and Hemoglobin Aspartate Transcarbamoylase E Hensley and H. K. Schachman: Communication between dissimilar subunits in aspartate transcarbamoylase: Effect of inhibitor and

activator on the conformation of the catalytic polypeptide chains. Proceedings of the National Academy of Sciences USA. 76, 3732 (1979). E. V. Kantrowitz, S. C. Pastra-Landis, and W. N. Lipscomb: E. coli aspartate transcarbamoylase. Part 2. Structure and allosteric interactions. Trends in Biochemical Sciences 5, 150 (1980). I. L. Monoco, J. L. Crawford, and W. N. Lipscomb: Three dimensional structures of aspartate transcarbamoylase from Escherichia coli and its complex with cytidine triphosphate. Proceedings of the National Academy of Sciences USA. 75, 5276 (1978).

Hemoglobin J. Baldwin and C. Chothia: Hemoglobin: The structural changes related to ligand binding and its allosteric mechanism. Journal of Molecular Biology 129, 175 (1979). M. F. Perutz: Stereochemistry of cooperative effects of hemoglobin. Nature 228, 726 (1970). M. E Perutz: Hemoglobin structure and respiratory transport. Scientific American 239(6), 92 (1978). M. E Perutz: Regulation of oxygen affinity of hemoglobin: Influence of structure of the globin on the heme iron. Annual Review of Biochemistry 48, 327 (1979).

CHAPTER

8

Enzymes lII: Clinical Applications

Enzymes are used clinically in three principal ways: 1. As indicators of enzyme activity or enzyme concentration in body fluids (e.g., serum and urine) in the diagnosis and prognosis of various diseases; 2. As analytical reagents in the measurement of activity of other enzymes or nonenzyme substances (e.g., substrates, proteins, and drugs) in body fluids; and 3. As therapeutic agents.

8.1 Diagnosis and Prognosis of Disease Enzyme assays employed in the diagnosis of diseases are one of the most frequently used clinical laboratory procedures. The most commonly used body fluid for this purpose is serum, the fluid that appears after the blood has clotted. The liquid portion of unclotted blood is calledplasma. Serum is used for many enzyme assays because the preparation of plasma requires addition of anticoagulants (e.g., chelating agents) that interfere with some assays. Enzymes in circulating plasma are either plasma-specific or nonplasma-specific. Plasma-specific enzymes are normally present in plasma, perform their primary function in blood, and have levels of activity that are usually higher in plasma than in tissue cells. Examples are those enzymes involved in blood clotting (e.g., thrombin), fibrinolysis (e.g., plasmin), and complement activation, as well as cholinesterase

(also called pseudocholinesterase or choline esterase II to distinguish it from the acetylcholinesterase of nervous tissue; see Chapter 6) and ceruloplasmin. These enzymes are synthesized mainly in the liver and released into the circulation at a rate that maintains optimal steady-state concentrations. Hereditary enzyme defects or impaired liver function can cause suboptimal levels. For example, deficiency of thrombin or plasmin can cause defective blood coagulation or fibrinolysis, respectively. Cholinesterase deficiency can cause prolonged muscle paralysis when succinylcholine is administered as an adjunct to anesthesia to produce skeletal muscle relaxation. Succinylcholine, a structural analogue of acetylcholine, competes with acetylcholine receptors at the motor end-plate of the neuromuscular junction and brings about depolarization that leads to muscle relaxation. The effect of succinylcholine is terminated through the enzymatic action of plasma cholinesterase but is not significantly altered by acetylcholinesterase at the neuromuscular junction. Measurement of plasma cholinesterase before surgery may prevent this adverse effect. Non-plasma-specific enzymes are intracellular enzymes normally present in plasma at minimal levels or at concentrations well below those in tissue cells. Their presence in plasma is normally due to turnover of tissue cells, but they are released into the body fluids in excessive concentrations as a result of cellular damage or impairment of membrane function. Tissue injury and impairment of

121

1~

CHAPTER8 Enzymes II1: Clinical Applications

membrane function can be caused by diminished oxygen supply (e.g., myocardial infarction), infection (e.g., hepatitis), and toxic chemicals. Proliferation of cells, with consequent increased turnover, can also raise levels in plasma of enzymes characteristic of those cells (e.g., elevation of serum acid phosphatase in prostatic carcinoma). Table 8-1 lists clinically useful enzymes and their tissue of origin. Intracellular enzymes are essentially confined to their cells of origin. A few are secretory enzymes that are secreted by some selected tissue (salivary gland, gastric mucosa, or pancreas) into the gastrointestinal tract, where they participate in digestion of food constituents (Chapter 12). Plasma levels of secretory enzymes increase when their cells of origin undergo damage or membrane impairment or when the usual pathways of enzyme secretion are obstructed. For example, large amounts of pancreatic amylase and triacylglycerol lipase (commonly known as lipase) enter the blood circulation in patients suffering from pancreatitis. These enzymes can digest the pancreas itself and surrounding adipose tissue in a process known as enzymatic necrosis (death of tissue cells). The diagnosis of organ disease is aided by measurement of a number of enzymes characteristic of that tissue or organ. Most tissues have characteristic enzyme patterns (Table 8-2) that may be reflected in the relative serum concentrations of the respective enzymes in disease. The diseased tissue can be further identified by determination of the isoenzyme pattern of one of these enzymes (e.g., lactate dehydrogenase, creatine kinase) in the serum, since many tissues have characteristic isoenzyme distribution patterns for a given enzyme. For example, creatine kinase (CK) is a dimer composed of two subunits, M (for muscle) and B (for brain), that occur in three isoenzyme forms, BB(CK1), MB(CK2) and MM(CK3), which catalyze the reversible phosphorylation of creatine with adenosine triphosphate (ATP) as the phosphate donor: CK

Creatine + ATP ~- phosphocreatine +

ADP

(adenosine diphosphate)

This reaction provides ATP for muscle contraction (Chapter 21). Skeletal muscle contains predominantly CK3, whereas heart muscle (myocardium) contains CK3 and CK2. Serum normally contains a small amount of CK3 derived predominantly from skeletal muscle. Detection of CK2 in serum (in an appropriate clinical setting) is strongly suggestive of myocardial damage. Since an abnormal isoenzyme level may occur with apparently normal total activity of the enzyme, determination of the isoenzyme concentration is essential in the diagnostic enzymology.

T A B L E 8-1

Tissues of Origin of Some Diagnostically Important Serum Enzymes Enzyme

Principal Tissue Source

Acid phosphatase Alanine aminotransferase (glutamate pyruvate transaminase) Alcohol dehydrogenase Alkaline phosphatase

Prostate Liver

Amylase* Arginase Aspartate aminotransferase (glutamate oxaloacetate transaminase) Ceruloplasmin Cholinesterase Chymotrypsin(-ogen)* Creatine kinase Fructose-bisphosphate aldolase -Glutamyltransferase

Glutamate dehydrogenase Isocitrate dehydrogenase Lactate dehydrogenase

Leucine aminopeptidase Ornithine carbamoyltransferase Pepsin(-ogen)* Prostatic specific antigen (a serine proteinase) Sorbitol dehydrogenase Triacylglycerol lipase* (lipase) Trypsin(-ogen)*

Liver Bone, intestinal mucosa, hepatobiliary system, placenta, kidney Pancreas, salivary glands Liver Heart and skeletal muscle, liver kidney, brain Liver Liver Pancreas Skeletal and heart muscle, brain Skeletal and heart muscle Kidney, hepatobiliary system, prostate, pancreas Liver Liver Skeletal and heart muscle, liver, kidney, erythrocytes, pancreas, lungs Hepatobiliary system, intestine, pancreas, kidney Liver Gastric mucosa Prostate Liver Pancreas Pancreas

*Secretory enzymes.

Factors Affecting Presence and Removal of Intracellular Enzymes from Plasma Many factors are taken into consideration in the clinical interpretation of serum enzyme levels. Membrane permeability changes and cell destruction affect the release of

SECTION 8.1

123

Diagnosisand Prognosis of Disease

TABLE 8-2

Properties of Cardiac Markers Used in the Evaluation of Acute Myocardial Infarction (AM1)

Marker

Molecular Weight (Daltons)

Elevation after Peak AMI Elevation (Hours) (Hours)

Return to Normal Levels (Hours or Days)

Myoglobin Total CK CKMB

17,200 86,000 86,000

1-3 4-8 4-6

4-6 12-24 12-24

20-24 hrs 36-48 hrs 24-48 hrs

Cardiac Troponin T

39,700

4-6

18-36

5-14 days

Cardiac Troponin I LDH

24,000

4-6

12-24

5-9 days

8-12

48-72

10-15 days

135,000

intracellular enzymes. These changes can result from a decrease in intracellular ATP concentration due to any of the followingconditions: deficiency of one or more of the enzymes needed in ATP synthesis (e.g., pyruvate kinase in red blood cells); glucose deprivation; localized hypoxia; and high extracellular K + (ATP depletion results from increased activity of the Na +, K+-ATPase in the cell plasma membrane required to maintain the proper K+/Na + ratio between the intracellular and extracellular environments). Localized hypoxia can result from poor blood flow, the result of obstruction of blood vessels responsible for the territorial distribution of the blood (a condition known as ischemia). Ischemia may result from narrowing of the lumen of the blood vessels (e.g., deposition of lipids in the vessel wall, which leads to atherosclerosis; see Chapter 20) or from formation of blood clots within the vessels. Ischemic necrosis leads to formation of an infarct (the process is called infarction). When cells of the myocardium die as a result of severe ischemia, the lesion is known as a

myocardial infarct. The amount of enzymes released depends on the degree of cellular damage, the intracellular concentrations of the enzymes, and the mass of affected tissue. The nature of the insult (viral infection, hypoxia, or surgical, chemical, or mechanical trauma) has no bearing on the enzymes released into the circulation. The nature of the enzymes released also reflects the severity of the damage. Mild inflammatory conditions are likely to release cytoplasmic enzymes, whereas necrotic conditions yield mitochondrial enzymes as well. Thus, in severe liver damage (e.g., hepatitis), the serum as-

Clinical Usefulness Earliest marker; not cardiac specific. A relatively early marker; not cardiac specific. A relatively early marker; has higher cardiac specificity over total CK. A relatively early marker; cardiac specific; however elevated in diseases of regeneration of skeletal muscle and chronic renal disease. A relatively early marker; cardiac specific. Late marker; not cardiac specific; LD 1/LD2 determination increases cardiac specificity.

partate aminotransferase (AST) level is extremely high (much greater than that of alanine aminotransferase) because the mitochondrial isoenzyme of AST is released in addition to the corresponding cytoplasmic isoenzyme. The amount of enzyme released into the plasma from an injured tissue is usually much greater than can be accounted for on the basis of tissue enzyme concentration and magnitude of injury. Loss of enzymes from cells may stimulate further synthesis of enzymes. In experimental animals, biliary occlusion leads to increased synthesis of hepatic alkaline phosphatase. Many drugs cause an increase in drug-metabolizing enzymes as a result of an increase in de novo synthesis (i.e., enzyme induction). These drug-metabolizing enzymes, located in the smooth endoplasmic reticulum (microsomal fraction; see Chapter 2) of liver and other tissues, catalyze the following chemical reactions: hydroxylation, demethylation, deethylation, acetylation, epoxidation, deamination, glucuronidation, and dehalogenation. Thus, serum levels of some of these enzymes may be elevated following exposure to enzyme-inducing agents, such as ethanol, barbiturates, phenytoin, and polycyclic hydrocarbons. While plasma levels of enzymes can become elevated because of tissue injury, the levels may drop (in spite of continued progress of the injury) to normal (or below normal) levels when the blood circulation is compromised or when the functional part of the tissue is replaced by repair or nonfunctional tissue (e.g., connective tissue, as in extensive fibrosis of the liver in the disease known as

cirrhosis).

124

CHAPTER 8

Presence

Removal

Normal turnover of tissues Leakage through .

]

cell membranes - ~ ~ Tissue

necrosis/S

Increased enzyme synthesis ~

~___ Non-plasma- ~ specific enzymes

~....

Intravascular inactivation (dilution, lack of substrates and coenzymes, inhibitors, and proteinases) Uptake by tissues, with

subsequentinactivation "" Removal by the

~,,N mticuloendothelialsystem Excretion in urine (of lowmolecular-weightenzymes)

F I G U R E 8-1 Factors affecting the presence and removal of non-plasma-specific enzymes from plasma.

Inactivation or removal of plasma enzymes may be accomplished by several processes: denaturation of the enzyme due to dilution in plasma or separation from its natural substrate or coenzyme; presence of enzyme inhibitors (e.g., falsely decreased activity of amylase in acute pancreatitis with hyperlipemia); removal by the reticuloendothelial system; digestion by circulating proteinases; uptake by tissues and subsequent degradation by tissue proteinases; and clearance by the kidneys of enzymes of low molecular mass (amylase and lysozyme). A schematic representation of the causes for the appearance and disappearance of non-plasma-specific enzymes is shown in Figure 8-1. Since enzymes differ in the rates of their disappearance from plasma, it is important to know when the blood specimen was obtained relative to the time of injury. It is also important to know how soon after the occurrence of injury various enzyme levels begin to rise. The biological half-lives for enzymes and their various isoenzymes are different. We can illustrate this by examining the disappearance rates of the lactate dehydrogenase (LDH) isoenzymes. LDH consists of four subunits of two different types: H (heart) and M (muscle). The subunits combine to yield a series of five tetramers: LDHI (H4), LDH2(H3M), LDH3(H2M2), LDH4(HM3), and LDHs(M4). The overall rate of disappearance of LDH activity from plasma is biphasic because of the more rapid disappearance of LDH5 (the predominant isoenzyme of liver and skeletal muscle), followed by the relatively slower removal of LDH1 (the predominant isoenzyme of heart, kidney cortex, and erythrocytes). The remaining LDH isoenzymes have intermediate rates of disappearance. In chronic disease states, the plasma enzyme levels continue to be elevated. The use of appropriate normal ranges is important in evaluating abnormal levels of plasma enzymes. However, an abnormal isoenzyme pattern may occur despite normal total activity (see above). The standard unit for enzyme activity was discussed in Chapter 6. The normal range is affected by a variety of factors: age, sex, race, degree of obesity, pregnancy, alcohol or other drug consumption,

Enzymes II1: Clinical Applications

and malnutrition. Drugs can alter enzyme level in vivo and interfere with their measurement in vitro. Enzyme activities may also be measured in urine, cerebrospinal fluid, bone marrow cells or fluid, amniotic cells or fluid, red blood cells, leukocytes, and tissue cells. Cytochemical localization is possible in leukocytes and biopsy specimens (e.g., from liver and muscle). Under ideal conditions, both the concentration of the enzyme and its activity would be measured. Radioimmunoassay (RIA) and its alternative modes such as fluorescence immunoassay (FIA), fluorescence polarization immunoassay (FPIA), and chemiluminescence immunoassay (CLIA) (discussed later), can be used to measure enzyme concentration as well as other clinically important parameters. M e a s u r e m e n t of Enzyme

Activity

In the assessment of enzyme levels in the clinical laboratory, the most frequently used procedure consists of measuring the rate of the enzyme-catalyzed chemical reaction. The initial rate of an enzymatic reaction is directly proportional to the amount of enzyme present when the substrate concentrations are maintained at saturating levels (i.e., zero-order kinetics with respect to substrate concentration) and other factors (e.g., pH, temperature, and cofactors) are maintained at optimal and constant levels (see Chapter 6). Under these conditions, the rate of substrate removal or product formation in a given time interval is directly related to enzyme activity. The reaction rate is determined by measuring product formation (or substrate removal) between two fixed times or by continuously monitoring the reaction with time. For accuracy, it is preferable to measure product formation rather than substrate removal because the former involves measurement of a change in concentration from an initial zero or low level to higher levels, which is analytically more reliable than the reverse. Both methods, fixed-time and continuous monitoring, are kinetic procedures. A schematic diagram of an enzymatic reaction is shown in Figure 8-2. The true rate of enzyme activity is obtained only when the reaction rate is measured in the linear region (a period of constant reaction rate and of maximum velocity). Inaccurate results are obtained if the rate is measured during nonlinear or nonmaximum phases. In fixed-time procedures, in which a single given interval of time is chosen to measure enzyme activity, it is essential to select reaction conditions, such as concentration of substrate, pH, temperature, and cofactors, that will provide linearity with maximum slope during the time period in question. False negative values for enzyme rates may arise from substrate depletion, inhibition of the enzyme by product, increase in the reverse reaction due to product accumulation, denaturation of the

SECTION 8.1

Diagnosis and Prognosis of Disease

F I G U R E 8-2 Diagrammatic representation of the measurement of the activity of an enzyme, showing product formation as a function of time. The true activity of the enzyme is calculated from data obtained when the reaction rate is linear with maximum velocity. The reaction rate is directly proportional to the amount of active enzyme only when the substrate concentrations are maintained at saturating levels and when other variables (e.g., pH, and cofactors) are held constant at optimal conditions.

enzyme, presence of certain endogenous metabolites, and drugs (or their metabolites). In the design of an enzyme assay, a key element is selection of a suitable physical or chemical technique for following the appearance of a product or the disappearance of a substrate. The rate of enzymatic reactions can be monitored continuously by photometric procedures (such as light absorption, fluorescence, or optical rotation) if the reaction is accompanied by a change in optical properties. For example, serum alkaline phosphatase, used in the diagnosis of hepatobiliary and bone diseases, is measured by employing its ability to catalyze the hydrolysis at alkaline pH of the colorless synthetic substrate p-nitrophenylphosphate to the yellow-colored product p-nitrophenol. The rate of reaction can be measured from changes in absorption at 405 nm. Many enzymatic reactions can be followed by a change in absorbance at 340 nm if they are linked (either directly or indirectly) to a dehydrogenase reaction requiring a nicotinamide adeninedinucleotide (NAD + or NADP +). The coenzyme absorbs ultraviolet light at 340 nm in the reduced state but not in the oxidized state (Figure 8-3). Hence, changes in reaction rate in either direction can be followed by measurement of the absorbance at 340 nm. An example of one such enzyme assay is given below. Red cell glucose-6-phosphate dehydrogenase (Chapter 15) can be specifically assayed in a red cell hemolysate. The enzyme catalyzes the reaction D-Glucose 6-phosphate + NADP + D-glucono-6-1actone 6-phosphate + NADPH + H +

125

F I G U R E 8-3 Absorption spectra of NAD + (NADP +) and NADH (NADPH). At 340 nm, the reduced coenzymes (NADH or NADPH) show significant absorbance, whereas the oxidized forms (NAD + or NADP +) show negligible absorbance. Thus, many enzymatic reactions can be monitored at 340 nm if the reaction is directly or indirectly dependent upon a dehydrogenase reaction involving a nicotinamide adenine dinucleotide as a coenzyme.

Since NADPH absorbs light at 340 nm and NADP + does not (Figure 8-3), this reaction can be followed by measurement of the change in absorbance at this wavelength with time. A similar assay (with lactate and NAD +) can be used for serum lactate dehydrogenase. The difference in absorbance is measured so readily that a number of assays make use of it directly or indirectly. In the latter case, the reaction to be measured is linked by one or more steps to a reaction in which a nicotinamide adenine dinucleotide is oxidized or reduced. Thus, in the measurement of aminotransferases (Chapter 17), one of the products of the primary reaction serves as a substrate for a secondary reaction in which NADH is required. For example, in the assay of aspartate aminotransferase, L-Aspartate +

c~-ketoglutarate

~ oxaloacetate

(also known as 2-oxoglutarate)

§ L-glutamate (primary reaction) the oxaloacetate formed in the primary reaction is reduced to L-malate by NADH in the presence of malate dehydrogenase: Oxaloacetate § NADH § H +

malate

>L-malate

dehydrogenase

§ NAD + (indicator reaction) This secondary reaction is an indicator reaction, i.e., the one whose rate is followed. The primary, not the secondary,

126

CHAPTER 8

reaction must be the rate-limiting step. The indicator reaction should be capable of instantaneous removal of the product formed in the primary reaction to prevent or minimize reversal of the primary reaction. Maintenance of a large excess of the second enzyme (malate dehydrogenase) in the reaction mixture fulfills this requirement. The indicator reaction must function at the concentration of the substrate (oxaloacetate) of the indicator enzyme, so that the rate of the indicator reaction is directly proportional to substrate concentration. Under these conditions, the rate of reaction catalyzed by the indicator enzyme is directly proportional to the rate of product formation in the primary reaction. Thus, during the initial phase of the reaction, the indicator reaction must wait for a period of time known as the lag phase for the primary reaction to generate adequate quantities of oxaloacetate to permit the maximum rate of conversion of NADH to NAD +. In nicotinamide adenine dinucleotide-linked assays, endogenous metabolites and enzymes in the serum may cause oxidation or reduction of the coenzyme, thereby introducing errors. For example, in the AST assay, the NADH of the indicator reaction may be oxidized by the presence of pyruvate and of LDH in the serum: Pyruvate + NADH + H + ~ L-lactate + NAD + This reaction falsely raises the measured activity of AST. A way to prevent this is to include a preincubation step in the assay procedure. This step consists of a time period during which all components of the reaction mixture except one of the substrates of the primary reaction (in this case, c~-ketoglutarate) are allowed to incubate. This period permits all undesirable reactions to reach completion before the primary reaction is initiated by addition of o~-ketoglutarate. Because of the need for rapid and accurate measurement, automated procedures have been introduced.

8.2 Serum Markers in the Diagnosis of Tissue Damage

Myocardium Coronary artery occlusion causes heart tissue damage due to ischemia and can lead to myocardial infarction (MI). The immediate and common cause of artery obstruction is formation of a thrombus. Antithrombolytic therapy (Chapter 36), with streptokinase or recombinant tissue plasminogen activator, protects the myocardium from permanent damage by restoring of blood flow. An early diagnosis of acute MI (AMI) is crucial for proper

Enzymes ii1: Clinical Applications

management. A patient's history, presence of chest pain, and electrocardiograms are problematic in the diagnosis of AMI. Therefore, measurements of circulatory proteins (enzymes and nonenzyme proteins) released from the necrotic myocardial tissue are useful in the diagnosis of AMI. Characteristics of an ideal myocardial injury marker are cardiac specificity, rapid appearance in the serum, substantial elevation for a clinically useful period of time and ease and rapidity of the analytical essay. At present, no serum marker fulfills all of these criteria. Some cardiac markers that appear in plasma are myoglobin, LDH, CK, and troponins. Measurements are made at the appropriate time intervals based upon the appearance of the marker in the plasma (Table 8-1). Frequently utilized markers are CKMB and cardiac troponin I (cTn I). Troponins consist of three different proteins I, C, and T, and are expressed in both cardiac and skeletal muscle. The tripartite troponin complex regulates the calcium-dependent interaction of myosin with actin (Chapter 21). Troponins are encoded by different genes. Cardiac I and T isoforms have unique structural differences from their skeletal muscle counterparts. However, cTn T like CKMB undergoes ontogenic recapitulation and is reexpressed in regenerating skeletal muscle and in patients with chronic renal failure. CKMB is also present in the skeletal muscle, albeit in small concentration. Compared to the myocardium (about 360- 400 g), skeletal muscle consists of a much larger mass (about 40% of total body mass). In rhabdomyolysis (disintegration or breakdown of muscle) CKMB and myoglobin appear in plasma in significant quantities. Myoglobin, although an early marker (Table 8-1), lacks specificity, and CKMB is elevated in many circumstances other than cardiac injury, e.g., rhabdomyolysis, chronic renal disease, and degenerative diseases of skeletal muscle. CKMB has a subform in the plasma due to the fact the M subunit undergoes cleavage of a lysine residue from the carboxy terminus by the plasma enzyme carboxypeptidase N. Tissue and the plasma subforms of CKMB have been designated as CKMB2 and CKMBI, respectively. The ratio of the serum levels, CKMB2/CKMB1 can also yield information useful in the early diagnosis of MI. Measurement of LDH isoenzymes (LDHI and LDH2) also lacks specificity; they appear significantly later after the myocardia! injury and show ontogenic recapitulation. Cardiac troponin I, a highly specific marker for AMI, does not suffer from these disadvantages and appears in the plasma as early as CKMB and persists as long as LDH isozymes. For these reasons, serial serum cTn I measurements may be superior in the detection of MI even in patients with chronic renal disease, rhabdomyolysis, or diseases of skeletal muscle regeneration (e.g., muscle dystrophies).

SECTION8.2 Serum Markers in the Diagnosis of Tissue Damage

Gallbladder

Common bile duct

127

f

tomac

i

_

~

~

~

-

-

~-

Pancreas

Accessory pancreatic duct (duct of Santorini)

Gallstone in Ampulla of Vater Main pancreatic duct (duct of Wirsung)

Duodenum

F I G U R E 8-4 Pancreatic duct obstruction by a gallstone at the ampulla of Vater. Obstruction can lead to pancreatitis, from induction of bile reflux, which eventually damages acinar cells of the pancreas.

Pancreas

The pancreas is both an exocrine and an endocrine gland. The exocrine function is digestion of food substances (Chapter 12); the endocrine function involves glucose homeostasis (Chapter 22). Acute pancreatitis is characterized by epigastric pain; it is an inflammatory process and potentially fatal. Obstruction of the pancreatic duct (Figure 8-4), which delivers pancreatic juice to the small intestine, by gallstones (Chapter 12) or alcohol abuse is the most common cause of acute pancreatitis, representing 80% of all causes. The pathophysiology is due to inappropriate release of pancreatic enzymes and their premature activation. The principal pancreatic enzyme is trypsinogen, which after activation to trypsin converts many other enzymes to their active forms. Some of these are kallikrein, phospholipase A2, elastase, enzymes of blood coagulation and fibrinolysis, and complement. Effects of these abnormal processes are autodigestion of the pancreas, vasodilation, increased capillary permeability, and disseminated intravascular coagulation. These can result in circulatory collapse, renal insufficiency, and respiratory failure. Laboratory diagnosis of acute pancreatitis involves the measurement of the pancreatic digestive enzymes: amylase and lipase (Chapter 12). Elevated serum amylase level is a sensitive diagnostic indicator in the assessment of acute pancreatitis, but it has low specificity because there are many nonpancreatic causes of hyperamylasemia. Furthermore, amylase (M.W. 55,000) is rapidly cleared by the kidneys and returns to normal levels by the third or fourth day after onset of the abdominal pain. Amylase activity in the serum appears within 2-12 hours after the onset of

pain. Serum lipase also is used to assess pancreatic disorders and has a higher specificity than serum amylase. It appears in the plasma within 4-8 hours, peaks at about 24 hours and remains elevated for 8-14 days. Measurements of amylase and lipase provide 90-95 % accuracy in the diagnosis of acute pancreatitis with abdominal pain. Pancreatic isoamylase level has not proven useful in comparison with measurements of both amylase and lipase. The catalytic activity of lipase requires the presence of both bile salts and colipase (Chapter 12) and must be incorporated in assays of serum lipase activity. Since exocrine cells of the pancreas contain many enzymes, attempts have been made to use markers other than amylase and lipase to diagnose acute pancreatitis. One such enzyme is trypsinogen. It is a 25,000-Da protein that exists in two isoenzyme forms: trypsinogen-1 (cationic) and trypsinogen-2 (anionic). Both forms are readily filtered through the kidney glomeruli. However, the tubular reabsorption of trypsinogen-2 is less than for trypsinogen-1; a dipstick method has been developed to detect trypsinogen-2 in the urine of patients suspected of having acute pancreatitis. The test strip contains monoclonal antibodies specific for trypsinogen-2.

Liver The liver is the largest glandular organ and its parenchymal cells are called hepatocytes. Liver has numerous functions, including metabolism, detoxification, formation and excretion of bile, storage, and synthesis. Liver diseases include alcohol abuse, medication, chronic hepatitis B and C infections, steatosis and steatohepatitis, autoimmune

128

hepatitis, hemochromatosis, Wilson's disease, otlantitrypsin deficiency, malignancy, and poisons and infectious agents. These disorders require specific laboratory testing procedures and are discussed at the appropriate places in the text. The serum enzymes used in assessment of liver function are divided into two categories: (1) markers used in hepatocellular necrosis and (2) markers that reflect cholestasis. Serum enzymes used as markers of cholestasis include alkaline phosphatase, 5'-nucleotidase, and y-glutamyl transferase. Alanine aminotransferase and aspartate aminotransferases are markers for hepatocellular necrosis. Other tests used in the assessment of liver disorders include measurement of bilirubin, albumin, and ot-fetoprotein.

8.3 Enzymes as Analytical Reagents The use of enzymes as analytical reagents in the clinical laboratory has found widespread application in the measurement of substrates, drugs, and activity of other enzymes. Above we discussed the use of enzymes as reagents in the coupled assay of aspartate aminotransferase. Enzyme-dependent procedures, used in the assay of substrates such as glucose, urea, uric acid, and cholesterol, provide many advantages over classical chemical procedures. These advantages include specificity of the substrate that is being measured and direct measurement of the substrate in a complex mixture that avoids preliminary separation and purification steps such as serum. A reagent enzyme can be used in substrate measurements by two methods: end-point and rate assays. In an end-point assay, the substrate is completely converted to product before it is measured; in the rate assay, a change in substrate concentration produced during a fixed-time interval is measured. This second method, also referred to as the two-point kinetic method, when carried out under constant conditions of pH, temperature, amount of enzyme, and time interval, yields very accurate values for substrate concentration. This procedure is calibrated with standard solutions of the substrate. Of course, the first-order reaction condition with respect to substrate must be maintained by keeping the reagent enzyme concentration high. Under these conditions, when average velocities during the chosen time interval are plotted against various standard substrate concentrations, the profile obtained should exhibit a straight line from which unknown values of substrate can be calculated. Optical properties can be used in monitoring assay procedures as they are in measuring enzyme activity. If the primary substrate or product does not have a suitable optical property (e.g., absorption in visible or ultravio-

CHAPTER 8

Enzymes II1: Clinical Applications

let light), then a coupled assay is constructed in which one or more auxiliary enzymes are employed to form an ultimate product that possesses an easily measured optical property. For example, glucose in a biological specimen can be measured by the following reactions under optimal conditions: D-Glucose + ATP

hexokinase

>D-glucose 6-phosphate + ADP glucose-6-phosphatase dehydrogenase

D-Glucose 6-phosphate + NADP +


D-glucono-8-1actone 6-phosphate + NADPH + H + The second reaction (in which NADPH is formed) functions as the indicator reaction. Although the hexokinase reaction is relatively nonspecific and acts on hexoses other than glucose (Chapter 13) to form 6-phosphate esters, the indicator reaction is specific for glucose 6-phosphate making the overall assay procedure highly specific for glucose. Enzymes immobilized on an insoluble surface can be used repeatedly in substrate assays. This type of assay is feasible if the reaction product can be measured directly. Immobilization can be accomplished through chemical linkages involving diazo, triazine, and azide groups with many types of insoluble matrices, such as diethylaminoethylcellulose, carboxymethylcellulose, agarose, microcrystalline cellulose, and inner walls of plastic tubings. Enzymes immobilized on the inner surface of plastic tubings are particularly useful in continuous-flow analyzers. Reagent enzymes that have been immobilized include glucose oxidase, urease, c~-amylase, trypsin, and leucine aminopeptidase. Reagent enzymes and antibodies formed against specific molecules can be combined to determine the concentration of a variety of molecules to which antibodies (Chapter 35) can be formed. Such analytical procedures are known as enzyme immunoassays (EIAs). The reagent enzyme can be linked to antibodies or antigens such that the complexes possess immunological or enzymatic activity. Antibodies can be raised in vertebrate animals when injected with specific proteins (antigens) foreign to them. Macromolecules other than proteins can also be antigenic. Low-molecular-weight compounds (e.g., substrates, drugs) by themselves do not elicit antibodies but do so if covalently linked to a carrier protein (e.g., albumin) before injection. The term hapten designates the low-molecular-weight substance that can combine with the antibody produced against the cartier protein complex. EIAs are either heterogeneous or homogeneous. A heterogeneous assay consists of at least one separation step in which the bound enzyme-labeled

129

SECTION8.3 Enzymes as Analytical Reagents

Specific antibody against the antigen Adsorption of antibody onto a solid surface, followed by washing procedure. An inert solid surface (e.g., a bead) Ant~en

1111111111111111 llllll llF

Incubation with the test specimen containing antigen, which binds to the specific antibody, followed by aspiration and washing procedure.

Incubation with enzyme-linked specific

antibody, which binds wherever the antigen is affixed, followed by washing

and aspiration procedure.

Incubation with specific substrate, and quantitation

•'•./Substrates

of products formed. Under standardized and optimal conditions, the concentrations of products are directly proportional to the concentration of antigen.

F I G U R E 8-5 Diagrammatic representation of an ELISA (solid-phase, sandwich technique) for an antigen.

reagent is separated from the unbound enzyme, enabling the measurement of either bound or free activity. A procedure based upon this principle is the enzyme-linked immunosorbent assay (ELISA), which is analogous to an RIA. An example of a heterogeneous immunoassay that uses a solid-phase system is shown in Figure 8-5. This method consists of coating or adsorbing antibodies generated in an animal (e.g., guinea pig) against a human antigen, whose concentration is to be determined in a test serum sample, onto a solid surface (e.g., beads or inner surface of a test tube) and incubating with the serum specimen. During the first incubation step, the antigen combines with its specific antibody and is fixed to a solid surface. Subsequently, the unreacted serum components are removed by aspiration of the fluid, and the solid surface (containing antibody-antigen complexes) is washed with suitable buffers. Incubation with antibodies covalently linked to enzyme molecules follows. During this step, the enzymelabeled antibody (E-Ab) will combine wherever the antigens are affixed; a washing procedure then removes the unbound E-Ab conjugates. Finally, incubation with the substrate specific for the enzyme yields a colored product that is quantitated by suitable spectrophotometric methods. The time interval for the substrate incubation step and

other reaction conditions must be kept constant for both test sera and the standards. The enzyme reaction is stopped by altering the pH of the reaction mixture by adding an acid solution (e.g., 1 N hydrochloric acid). The change in absorbance of color is proportional to the concentration of antigen in the test sera. A standard curve is obtained by plotting known antigen concentrations against their absorbances. Various modifications of this technique are in use. For example, if the concentration of an antibody is to be determined, the process can begin with immobilization of the antigen onto the solid phase. The enzymes used as labels should have a high turnover number, act on substrates that are stable and soluble, yield easily measurable products, be obtainable in a highly purified form, and be relatively inexpensive. Enzymes that fit these criteria are horseradish peroxidase, glucose oxidase, E. coli or calf-intestine alkaline phosphatase, catalase, and/3-D-galactosidase. The covalent linkage of the enzyme to the protein (antigen or antibody) through free amino groups can be accomplished with the bifunctional reagent glutaraldehyde. The periodate oxidation method (Chapter 9), involving the carbohydrate side-chains of proteins, also yields reactive groups useful in the preparation of enzyme conjugates. The ELISA exhibits high levels

130 of both sensitivity and specificity. The sensitivity, which is due to the amplification factor introduced by way of the catalytic activity of the labeled enzyme, can equal (with comparable precision) that obtainable with RIA. The specificity is attributable to specific molecular recognition properties of antigens and antibodies. The ELISA may be more advantageous than RIA because ELISA reagents have longer shelf lives than do many radioisotopes (e.g., 125I-labeled reagents) and present no health hazards. The ELISA also has been adapted to identify antigens or antibodies in tissue slices by the use of protein-peroxidase conjugates. Homogeneous EIA procedures have been especially useful for rapid determination of low-molecular-weight substances (haptens). Thus, these procedures have found application in therapeutic drug monitoring and toxicology. A commercial reagent for the assay of many drugs is available under the trade name of EMIT (EnzymeMultiplied Immunoassay Technique). Figure 8-6 diagrams the steps involved in this procedure. The principle is based on competition between the enzyme-labeled hapten and free hapten in the test serum for binding with a limited amount of a specific antibody. This assay requires a hapten covalently linked to an enzyme (e.g., glucose-6-phosphate dehydrogenase, lysozyme, or malate dehydrogenase), with retention of enzymatic activity, and antibodies raised against the hapten (through injection into an animal of the hapten covalently linked to a carrier protein) that inhibit enzyme activity when they combine with the hapten-linked enzyme reagents. The inhibition by the antibody of catalytic activity of the enzyme-linked hapten complex may be due to steric hindrance or to conformational constraints affecting access of substrate of the active center. This reaction system consists of the test specimen, the enzyme-labeled hapten, a limited amount of the antibody specific to the hapten, and the substrate. Any haptens present in the test serum compete with the hapten linked to the enzyme for binding with the antibody. The unbound hapten-linked enzyme acts on the substrate to convert it to an easily measurable product. Thus, the enzyme activity is directly correlated with the free hapten concentration in the test specimen. The method is calibrated with known standard concentrations of the hapten. The ELISA and EIA receptor systems measure substances at concentrations as low as a few nanograms (10 -9 g). This sensitivity is not sufficient for detecting many substances and alternate methods have been devised. One is chemiluminescence immunoassay (CLIA), which can measure concentrations in femtogram (10-15 g) quantities. CLIA depends on the detection of emitted light associated with the dissipation of energy from a substance

CHAPTER8 Enzymes II1: Clinical Applications

F i ( ; U R E 8-6 Diagrammatic representation of a homogeneous enzyme immunoassay procedure for the determination of low-molecular-weight compounds (haptens).

excited electronically as a result of a chemical reaction. An example of a chemiluminescent tracer is acridinium ester conjugated to a desired protein, polypeptide, or other organic molecule (Figure 8-7). CLIA is similar to EIA and ELISA techniques except that the final receptor enzyme assay is replaced with a chemiluminescent tracer followed by measurement of light released as a result of the chemical reaction. The principles of a chemiluminescence competitive binding assay are shown in Figure 8-8.

8.4 Enzymesas Therapeutic Agents Enzymes have found a few applications as therapeutic agents. Some examples are transfusion of fresh blood or its active components in bleeding disorders, oral administration of digestive enzymes in digestive diseases

SECTION8.4 Enzymes as Therapeutic Agents CH3

131 C !~

H202, Alkaline pH (H02")

~ I ~0 0 H = C .O, o

~C=O O"

(.)

R=protein or peptide or an organic

R

molecule

Acridinium Ester

C !~

OH3 !

-- f

0

Light, C02 10-methylacridinium (Electronically excited)

F I G U R E 8-7 Acridinium ester used as a chemiluminescent tracer. Labeled acridinium ester in the presence of alkaline hydrogen peroxide undergoes chemical changes producing light that is detected at 430 nm by a photomultiplier detector.

~ +

~

+

~

0 Solid Phase Antibody

Antigen in Serum Sample

cc cc cc

Antigen Labeled with Chemiluminescent Compound (CC)

CC

CC CC F I G U R E 8-8 Competitive binding chemiluminescence assay. The chemiluminescent competitve binding assay is similar to a RIA. The acridinium ester-labeled substance and the endogenous substance compete with the specific antibody that is available in limited concentration. The concentration is determined by the quantity of tracer that binds to the antibody. The use of solid-phase antibody simplifies separation of free from bound tracer.

132 (e.g., cystic fibrosis),administration of fibrinolytic enzymes (e.g., streptokinase) to recanalize blood vessels occluded by blood clots (thrombi) in thromboembolic disorders (e.g., pulmonary embolism, acute myocardial infarction), treatment of selected disorders of inborn errors of metabolism (e.g., Gaucher's disease), and cancer therapy (e.g., L-asparaginase in acute lymphocytic leukemia). For enzymes to be therapeutically useful, they should be derived from human sources to prevent immunological problems. Although enzymes derived from human blood are readily obtainable, enzymes derived from tissues, which would be particularly useful in the treatment of inborn errors of metabolism, are difficult to obtain in adequate quantities. Transport of specific enzymes to target tissues is also a problem, but some recent advances and commercial applications (e.g., propagation of human tissue culture cell lines, isolation and cloning of specific genes) have the potential of overcoming these difficulties. Such techniques have been used in the production of peptide hormones such as somatostatin and insulin, interferon, and tissue plasminogen activator. In the treatment with enzymes or proteins, covalent attachment of an inert polymer polyethylene glycol (PEG) provides many therapeutic benefits. These include slowing the clearance, diminished immunogeneity, prevention of degradation, and binding to antibody. PEG-enzyme therapy is used in the treatment of immunodeficiency disease caused by adenosine deaminase deficiency (Chapter 27)

CHAPTER8 Enzymes II1: Clinical Applications

and PEG-interferon alfa complex (peginterferon alfa-2a) is used in treatment of chronic hepatitis C infection. Ultimately, however, when a gene is cloned, techniques will be developed to incorporate it into the genome of persons lacking the gene or having a mutated gene.

Supplemental Readings and References N. V. Bhagavan, A. R Goldstein, S. A. A. Honda, et al.: Role of cardiac troponin I in the evaluation of myocardial injury. Journal of Clinical Laboratory Analysis 12, 276 (1998). L. Coudrey: The troponins. Archives oflnternal Medicine 158, 1173 (1998). R. E Dudley: Chemiluminescence immunoassay: An alternative to RIA. Laboratory Medicine 21, 216 (1990). A. H. Gershlick and R. S. More: Treatment of myocardial infarction. British Medical Journal 316, 280 (1998). E. A. Kemppainen, J. I. Hedotrom, E A. Puolakkainen, et al.: Rapid Measurement of Urinary Trypsinogen-2 as a screening test for acute pancreatitis. New England Journal of Medicine 336, 1788 (1997). K. Mergener and J. Baillie: Acute Pancreatitis. British Medical Journal 316, 44 (1998). D. S. Pratt and M. M. Kaplan: Evaluation of abnormal liver-enzyme results in asymptomatic patients. New England Journal of Medicine 342, 1266

(2OOO). J. J. Reichling and M. M. Kaplan: Clinical use of serum enzymes in liver disease. Digestive Diseases and Sciences 33, 1601 (1988). W. Steinberg and S. Tenner: Acute pancreatitis. New England Journal of Medicine 330, 1198 (1994). S. Zeuzem, S. V. Feinman, J. Rasenack, et al: Peginterferon alfa-2a in patients with chronic hepatitis C. New England Journal of Medicine 343, 1666

(2OOO).

CHAPTER

9

Simple Carbohydrates

Carbohydrates are major functional constituents of living systems. The primary source of energy in animal cells, carbohydrates are synthesized in green plants from carbon dioxide, water, and solar energy. They provide the skeletal framework for tissues and organs of the human body and serve as lubricants and support elements of connective tissue. Major energy requirements of the human body are met by dietary carbohydrates. They confer biological specificity and provide recognition elements on cell membranes. In addition, they are components of nucleic acids and are covalently linked with lipids and proteins.

9.1 Classification Carbohydrates consist of polyhydroxyketone or polyhydroxyaldehyde compounds and their condensation products. The term "carbohydrate" literally means hydrate of carbon, a compound with an empirical formula (CHzO)n. This formula applies to many carbohydrates, such as glucose, which is C6H1206, or (CH20)6. However, a large number of compounds are classified as carbohydrates even though they do not have this empirical formula; these compounds are derivatives of simple sugars (e.g., deoxyribose, C5H1004). Carbohydrates may be classified as monosaccharides, oligosaccharides, and polysaccharides; the term saccharide is derived from the Greek word for sugar. Monosaccharides are single polyhydroxyaldehyde

(e.g., glucose) or polyhydroxyketone units (e.g., fructose), whereas oligosaccharides consist of two to ten monosaccharide units joined together by glycosidic linkages. Sucrose and lactose are disaccharides, since they are each made up of two monosaccharide units. Names of common monosaccharides and disaccharides take the suffix-ose (e.g., glucose, sucrose). Polysaccharides, also known as glycans, are polymers that may contain many hundreds of monosaccharide units. They are further divided into homopolysaccharides and heteropolysaccharides. The former contain only a single type of polysaccharide unit (e.g., starch and cellulose, both of which are polymers of glucose), whereas the latter contain two or more different monosaccharide units. Monosaccharides Monosaccharides are identified by their carbonyl functional group (aldehyde or ketone) and by the number of carbon atoms they contain. The simplest monosaccharides are the two trioses: glyceraldehyde (an aldotriose); and dihydroxyacetone (a ketotriose). Four-, five-, six-, and seven-carbon-containing monosaccharides are called tetroses, pentoses, hexoses, and heptoses, respectively. Structures of some monosaccharides are shown in Figure 9-1. All monosaccharides, with the exception of dihydroxyacetone, contain at least one asymmetrical or chiral carbon atom, and therefore two or more stereoisomers are

133

134

CHAPTER9 Simple Carbohydrates CHO

I I

HmC~OH

CH,OH D-Glycerose (D-glyceraldehyde) CHO

I I HmC--OH I H--C~OH

CH2OH D-En/throse

CHO

CHO

HO~?~H HO~C--H

I I

HmC~OH CH2OH D-Lyxose

I HO~CnH I H--C~OH I H--C~OH I

CH2OH D-Xylose

CHO

CH,OH

CHO

I I HO--C n H I HBCmOH I HmCmOH I

I

H~C~OH

I I

H~C~OH

H--C--OH

CH,OH D-Galactose

D-Mannose

D-Ribose

H--CmOH

HO~?mH

I

I I H--C--OH I H~C--OH I HnC~OH

CH2OH D-Arabinose

CHO

I H--C~OH I HO--C~H I HO--? ~ H

CHO

CHO

I H~C~OH I HO~C--H I H~C~OH I

I

CH,OH

CH2OH D-Glucose

Aldoses

CH2OH H2OH H2OH

H2OH

C=O

~H2OH C--O

I

CH2OH Dihydroxyacetone

I Ho--cBH I H--C~OH I

CH2OH D-Xylulose

C=O

I

H--?--OH H--C--OH

I

CH2OH o-Ribulose

C=O HO~C~H

C--O

I HO~C~H I H~C~OH I H~C~OH I

CH2OH D-Fructose

H~C~OH H~C~OH H~C~OH CH2OH D-Sedoheptulose

Ketoses

FIGURE 9-1 Structures of some monosaccharides.

possible for each monosaccharide depending on the number of asymmetrical centers it contains. Glyceraldehyde, with one asymmetrical center, has two possible stereoisomers, designated D and L forms (Figure 9-2). A method for representing the O and L forms is the Fischer projection formula. The D and L representations are used for all monosaccharides. The designation of D or L, given to a monosaccharide with two or more asymmetrical

centers, is based on the configuration of the asymmetrical carbon atom located farthest from the carbonyl functional group. Thus, if the configuration at that carbon is the same as that of D-glyceraldehyde (with the hydroxyl group on the right-hand side), it belongs to the D series. A similar relationship exists between L-glyceraldehyde (with the hydroxyl group on the left-hand side) and the L series of monosaccharides. The optical rotation of a

SECTION9.1 Classification 1CHO

135 ICH0

HD,.-2~..~ OH a~

a~1 CH,OH

CH2OH

(a) CHO

CliO H

OH

HO'

H CH2OH

CH2OH (b) CHO

CHO

I HCOH I

I I

HOCH

CH2OH D-Glyceraldehyde

CH2OH L-Glyceraldehyde

are physiologically important: D-glucose, D-galactose, and D-mannose. Structurally, D-glucose and D-galactose differ only in the configuration around C4; D-glucose and D-mannose differ only in the configuration around C2. Pairs of sugars (e.g., glucose and galactose, glucose and mannose), which differ only in the configuration around a single carbon, are known as epimers. D-Galactose and D-mannose are not epimers, since they differ in configurations around both C2 and C4 (Figure 9-1). D-Fructose, one of eight 2-ketohexoses, is the physiologically important ketohexose. Monosaccharides with five or more carbons occur predominantly in cyclic (ring) forms owing to a reaction between the carbonyl group (aldehyde or ketone) and an alcohol group:

(c) F I G U R E 9-2 Stereoisomers of glyceraldehyde. (a) Perspective formulas showing tetrahedral arrangement of the chiral carbon 2 with four different substituents. (b) Projection formulas in which the horizontal substituents project forward and the vertical substituents project backward from the plane of the page. (c) A common method of representation.

H

RI_ C ~~ O H + HO--R2 ~

OH Aldehyde

Alcohol

O

Hemiacetal OR3

II

RI--C--R2 + HO--R3 ~

monosaccharide with multiple asymmetrical centers is the net result of contributions from the rotations of each optically active center. Thus, the prefix D o r L provides no information with regard to optical rotation; it indicates only the configuration around the asymmetrical carbon atom located farthest from the carbonyl carbon. The numbering system for monosaccharides depends on the location of the carbonyl carbon (or potential carbonyl carbon), which is assigned the lowest possible number. For glucose (an aldohexose), carbon C1 bears the carbonyl group and the farthest asymmetrical carbon atom is C5 (the penultimate carbon), the configuration around which determines the O and L series. For fructose (a ketohexose), C2, bears the carbonyl group and C5 is the highest numbered asymmetrical carbon atom. Glucose and fructose have identical configurations around C3 to C6. The structure of glucose, written in straight-chain form (Figure 9-1), shows four asymmetrical centers. In general, the total number of possible isomers with a compound of n asymmetrical centers is 2". Thus, for aldohexoses having four asymmetrical centers, 16 isomers are possible, 8 of which are mirror images of the other 8 (enantiomers). These two groups constitute members of the D and L series of aldohexoses. Most of the physiologically important isomers belong to the D series, although a few L-isomers are also found. In later discussions, the designation of D and L is omitted, and it is assumed that a monosaccharide belongs to the D series unless it is specifically designated an L-isomer. Of the D-series of aldohexoses, three

I RI--I~--O--R 2

I

R1--C--R 2

I

OH Ketone

Alcohol

Hemiketal

Formation of the cyclic forms of monosaccharides is favored because these structures have lower energies than the straight-chain forms. Cyclic forms of D-glucose are formed by the hemiacetal linkage between the C1 aldehyde group and the C4 or C5 alcohol group. If the ring structure is formed between C1 and C4, the resulting five-membered ring structure is named D-glucofuranose because it resembles the compound furan: Hc/O~cH %//

or

H H

If the ring structure is formed between C1 and C5, the resulting six-membered ring is named D-glucopyranose because it resembles the compound pyran"

HC--O

HCII\ I~CH

or

C--C H2 H Aldohexoses exist in solutions mainly in six-membered pyranose ring forms, since these forms are thermodynamically more stable than furanose ring forms. Cyclization of a monosaccharide results in the formation of an additional asymmetrical center, known as

136

CHAPTER9 Simple Carbohydrates 6CH2OH

H 11 C--O

H--~2~~OH

4

HO---3(~--H

OHo H

1

H

H~C~OH

H H

H---~? ~ O H

OH

or,-D -Glucopyranose

CH,OH

6CH2OH

D-Glucose A linear representation of D-glucose 6

H'~ OH

r, OH H

H

,_..

~OH

OH

-D-Glucopyranose

~/I~ 0

F I G U R E 9-4 Haworth projection formulas of anomers of D-glucopyranose. The thick line of the structure projects out toward the observer, and the upper edge (thin line) projects behind the plane of the paper.

H OH A modified representation of the

D-glucose molecule, showingthe

formation of the hemiacetal linkage

Anomedc carbon 6CH=OH ~1

.... o 4

H

OH

a- D-Glucopyranose

H COH

H

\ . r/c" OH

13"O-Glucopyranose

0t- and B-Anomers of D-glucopyranose F I G U R E 9-3 Formation of or- and/~-anomers from D-glucose.

the anomeric carbon, when the carbon of the carbonyl group reacts with the C5 hydroxyl group. The two possible stereoisomers resulting from the cyclization are called or- and fl-anomers (Figure 9-3). Aldohexoses in their cyclic forms have five asymmetrical centers and therefore 32 stereoisomers. In other words, each of the 16 isomers that belong to the D o r L series has two anomeric forms. The systematic names for these two anomers are ot-D-glucopyranose and/~-D-glucopyranose. Three-dimensional representations of ring structures are frequently shown as Haworth projection formulas (Figure 9-4), in which the lower edge of the ring is presented as a thick line, to indicate that this part of the structure projects out toward the observer, and the upper edge as a thin line that projects behind the plane of the paper. Carbon atoms of the ring are not explicitly shown but

occur at junctions of lines representing bonds. Sometimes the hydrogen atoms are also omitted and are presumed to exist wherever a bond line ends without a specified group. The pyranose ring is not planar, being similar to that of cyclohexane. The bond angles in cyclohexane are similar to those between the bonds of a tetrahedral carbon (i.e., 109~ In the pyranose ring, all bond angles are similar to those of cyclohexane, including the hemiacetal C - O - C bond angle, which is 111 ~ Most pyranoses occur in the chair conformation, in which most of the substituents can assume equatorial positions (i.e., lie approximately in the same plane as the ring) instead of axial positions (i.e., lie approximately vertically above or below the plane of the ring). In the equatorial positions, the bulky substituent groups ( - O H , - C H 2 O H ) can more easily be accommodated than in the axial positions, and the preferred conformation is usually the chair conformation (Figure 9-5).

O

orial

Axial Boat

O

Equatorial

Axial Chair F I G U R E 9-5 Conformational formulas for the boat and chair forms of pyranose.

137

SECTION9.1 Classification q..

.......,1_

C~H,~

HO/H~,,..O.

H C,

'c,

,CH2OH

OH C,

O ~'"~176176

6H

The e~-anomer: The axial hydroxyl group is in a favorable dipoledipole interaction with the ring oxygen and results in a more stable conformation.

Z

..

c~ OH

I

OH

'c, F I G U R E 9-6 The two chair conformational formulas for/~-D-glucopyranose.

However, two chair forms can be drawn for each of the two anomers of D-glucopyranose, as shown in Figure 9-6 for/~-D-glucopyranose. The structure on top is designated the 4C~ form because it contains carbon 4 and carbon 1 above and below the plane of the molecule, respectively. The designation of ~C4 for the structure on the bottom conveys the opposite sense. In the 4C1 conformation, all the large substituent groups are in equatorial positions, whereas in the 1C4 conformation, they are in axial positions. 4C1 is the more stable and therefore the preferred chair conformation. The conformational formulas for ol- and /~-Dglucopyranose are shown in Figure 9-7. In aqueous solutions, the/3-anomer of glucose is better solvated, more stable, and therefore the predominant form. However, for other aldohexoses (e.g., mannose and galactose), the o~-anomer is more stable than the/~-anomer because of the dipole effect. The dipole effect involves the C 1-hydroxyl group and the ring oxygen. In the o~-anomer, the dipoles of the axial C 1-hydroxyl group and the ring oxygen are

C~ 2OH _sO

L

H( f H N

ON

(z- D-Glucopyranose

I_ CxI"I2OH_...O H

O

/

~

o

H

H H I~-o-Glucopyranose F I G U R E 9-7 Conformational formulas for c~- and/~-D-glucopyranose.

The 13-anomer:The equatorial hydroxyl group is in an unfavorable dipole-dipole repulsive interaction with the ring oxygen and yields a less stable conformation. F I C U R E 9-8 Dipole-dipole interaction between the C 1-hydroxyl group and the ring oxygen in aldohexoses. The arrows indicate the direction of the dipole moment.

nearly antiparallel, whereas in the/~-anomer, the dipoles of the equatorial C 1-hydroxyl group and the ring oxygen are nearly parallel (Figure 9-8). The dipoles impart stability to a structure when they are antiparallel. Although the dipole effect is also applicable to c~- and/~-anomers of glucose, the solvation effect overrides the dipole effect. Interconversion of or- and/?-forms can be followed in a polarimeter by measuring the optical rotation. Crystallization of D-glucose from water yields o~-D-glucopyranose, a form which is least soluble and has a specific rotation of § ~. Ordinary crystalline glucose is i n t h e o~-form, but if the crystallization of D-glucose takes place in pyridine,/~-D-glucopyranose, which has a specific rotation of + 18.7 ~ is obtained. Freshly prepared solutions of the o~- and/~-forms show specific rotations of .1.112.2 ~ and -t-18.7 ~ respectively. However, over a period of a few hours at room temperature, the specific rotation of both forms in aqueous solution changes and attains a stable value of .1.52.7 ~. This change in optical rotation, mutarotation, is characteristic of sugars that form cyclic structures. It represents the interconversion of the o~- and /~-forms, yielding an equilibrium mixture consisting of about two thirds r one third ~-form, and a very small amount of the noncyclic form (Figure 9-9). Thus, the change in structure can occur in solution and attain equilibrium, which favors the formation of more stable (lowest energy) forms. However, the less stable forms may occur under certain conditions, such as during the formation of

138

CHAPTER9 Simple Carbohydrates

' H OH

'~

6CH2OH

6CH2OH

\1 v, 6

H

OH

H

(z-o-Glucopyranose

OH o-Glucose

H

OH

13-o-Glucopyranose

(a)

Equilibrium mixture containingor, J3-,and ~ open-chain mrms

~- D-Glucose ~ Specific

rotation

[a]~. 913

+112.2"

+52.7"

[

13-o-Glucose +18.7"

~ l

t~-form: 36% open form:

,.o .,..~

o,.,~

.,.~

o~

9

~

,4--~

~d 9

r ;>

Supplemental Readings and References native form rather than in a processed state may provide an adequate amount of fiber. Fiber should be derived from a variety of plant foods such as high- and low-fiber breads and cereals, fruits, vegetables, legumes, nuts, and seeds, and eaten as part of normal meals. The recommended dietary fiber intake for adults is 20-25 g/day or 10-13 g per 1000 kcal (4184 kJ) of energy intake. A suggested intake of dietary fiber for children older than 2 years is the age of the child plus 5-10 g/day. The consumption of dietary fiber in childhood is also associated with health benefits, such as normal laxation, and may reduce the future risk of cardiovascular disease, some cancers, and diabetes mellitus type 2.

151

Supplemental Readings and References L. B. Ferzoco, V. Raptopouoos, and W. S. Len: Acute diverticulitis. New England Journal of Medicine 338, 1521 (1998). J. S. Hampl, N. M. Betts, and B. A. Benes: The "age +5" rule: Comparison of dietary fiber intake among 4- to 10-years old children. Journal of the American Dietetic Association 98, 1418 (1998). J. L. Listinsky, G. P. Siegal, and C. L. Listinsky: ot-L-Fucose: A potentially critical molecule in pathologic processes including neoplasia. American Journal of Clinical Pathology 110, 425 (1998). Position of the American Dietetic Association: Health implications of dietary fiber. Journal of the American Dietetic Association 97, 1157 (1997). S. M. Riordan and R. Williams: Treatment of hepatic encephalopathy. New England Journal of Medicine 337, 473 (1997). C. L. Williams (Ed.): The role of dietary fiber in childhood. Pediatrics 96, 985 (1995).

CHAPTER

10

Heteropolysaccharides 1: Glycoproteins and Glycolipids

10.1 Glycoproteins Glycoproteins, a wide range of compounds of diverse structure and function, are components of cell membranes, intercellular matrices, and extracellular fluids, such as plasma. They occur in soluble and membrane-bound forms (Table 10-1). The proportion of carbohydrate varies considerably in glycoproteins derived from different tissues or from various sources. Collagen, for example, contains an amount of carbohydrate that varies with the source: skin tissue collagen, about 0.5%; cartilage collagen, about 4%; and basement membrane collagen, more than 10%. Glycoproteins containing high amounts of carbohydrate include glycophorin, a membrane constituent of human erythrocytes, about 60%, and soluble blood group substances, as much as 85%. In glycoproteins, the protein and the carbohydrate residues are bound in covalent linkage. There are five common types of carbohydrate, four of which are common to human glycoproteins (Figure 10-1). All the linkages are either N- or O-glycosidic bonds. The amino acid residue that participates in the N-glycosidic linkage is asparagine, and the amino acid residues that participate in O-glycosidic linkage are serine, threonine, hydroxylysine, and hydroxyproline. The glycoproteins exhibit microheterogeneity, i.e., glycoproteins with an identical polypeptide sequence

may vary in the structure of theire oligosaccharide chains. Microheterogeneity arises from incomplete synthesis or partial degradation and poses problems in the purification and characterization of glycoproteins. For example, human serum o~1-acid glycoprotein has five linkage sites for carbohydrates and occurs in at least 19 different forms as a result of differences in oligosaccharide structures. The oligosaccharide side chains of glycoproteins consist of only a limited number (about 11) of different monosaccharides. These monosaccharides are hexoses and their derivatives (N-acetylhexosamine, uronic acid, and deoxyhexose), pentoses, and sialic acids derived from neuraminic acid, a nine-carbon sugar. The most common of the many different types of sialic acids is N-acetylneuraminic acid. (All of these sugars are in the D-configuration, unless indicated otherwise.) The sugar residues of the oligosaccharide chains are not present in serial repeat units (unlike the proteoglycans, Chapter 11), but they do exhibit common structural features. For example, the oligosaccharide chains bound in N-glycosidic linkages may be envisaged as consisting of two domains. The inner domain, common to all glycoproteins, is attached to the protein via an N-fl-glycosidic linkage between an asparaginyl residue and N-acetylglucosamine. The inner domain consists of a branched polysaccharide, which is trimannosyl-di-N-acetylglucosamine (Figure 10-2). The peripheral mannose residue of the inner domain is linked

153

154

CHAPTER10 Heteropolysaccharides I: Glycoproteins and Glycolipids

A 13-N-glycosidiclinkage between N-acetylglucosamine and the amide nitrogen of an asparagine residue of the protein (GIcNAc-Asn). Linkage of wide occurrence. ?H20_~HO,,

HOC HO H~I~

O

~

NH2

NH--ICI--CHF --IC-cOOH HI

An o~-O-glycosidic linkage between N-acetylglucosamine and the hydroxyl group of serine (R=H) or threonine (R=CH3) residue of the protein (GalNAc-Ser/Thr). Linkage found in glycoproteins of mucus secretions and blood group substances. HO /

CH2OH

_

H

Some Functions of Glycoproteins in the Body

Function

Examples

Structure Lubrication and protection Transport Endocrine regulation Catalysis Defense against infection Membrane receptors

I I HOOO--?--?--. H,N

T A B L E 10-1

H

A 13-O-glycosidiclinkage between galactose and a hydroxylysine residue of the protein (GaI-Hyl). Linkage found in collagen.

Antigens Cell-cell recognition and adhesion Miscellaneous

COOH HO

I

CH2OH

NH~--Cf--H /

H

CH2

I

NHa

A 13-O-glycosidic linkage between xylopyranose and the hydroxyl group of a serine residue of the protein (XyI-Ser). Linkage found in thyroglobulin and proteoglycans.

-

CH--COOH I NH2 F I G U R E 10-1 Carbohydrate-peptide linkages in glycoproteins.

to an oligosaccharide chain, known as the outer domain. The outer domain is made up of either oligosaccharides consisting of mannose residues (oligomannosidic types)or N-acetyllactosamine units (complex types, Figure 10-3). The latter are disaccharides consisting of galactosyl/3-(1 --+ 4)-N-acetylglucosamine units. Glycoproteins with O-glycosidic linkages (Figure 10-4) do not show the common features of glycoproteins with N-glycosidic linkages. The number of sugar residues may vary from one (e.g., collagen) to many (e.g., blood group substances). Many glycoproteins, however, do show the presence of a common disaccharide constituent, namely, galactosyl-fl-(1 -+ 3)-N-acetylgalactosamine, which is

Collagen Epithelial mucins, synovial fluid glycoproteins Ceruloplasmin (copper carrier), transferrin (iron carrier) Thyrotropin, chorionic gonadotropin, erythropoietin Proteases, nucleases, glycosidases, hydrolases Immunoglobulins, complement proteins, interferon Receptors for hormones (e.g., insulin), acetylcholine, cholera toxin, electromagnetic radiation (e.g., rhodopsin) Blood group substances Fibronectin, laminin, chondronectin Glycophorin (an intrinsic red blood cell membrane constituent), intrinsic factor (essential for absorption of dietary vitamin B I 2 ) , clotting factors (e.g., fibrinogen)

linked to either serine or threonine. In collagens, the O-glycosidic linkages occur via hydroxyproline or hydroxylysine residues, which are unique to collagens. A given glycoprotein may contain oligosaccharide chains of both N- and O-glycosidic types. If more than one N-glycosidic carbohydrate linkage site are present in a glycoprotein, they are usually separated by several amino acid residues. In contrast, O-glycosidic carbohydrate linkages

Man or Man o~(1--,,3)/

Man 13(1"4) GIcNAc l](1--,,4)GIcNAc 13*Asn

or schematically,

F I G U R E 10-2 Structure of the common branched inner domain of oligosaccharides linked in N-glycosidic linkages with asparagine. Man = Mannose T, GlcNAc = N-acetylglucosamine O, A s n = Asparagine. [Adapted with permission from N. Sharon and H. Lis, Chem. Eng. News, p. 28 (March 30, 1981) (~)1981 by the American Chemical Society.]

SECTION10.1 Glycoproteins

155

Oligomannosidic V. . V ~ O m l t ~ /

Humanimmunoglobulin M (IgM), bovine rhodopsin

i V--V~

hickenovalbumin, Sindbis virus

~~V~O~O~ I.

,

Complex

I

~

Inner domain

r'

i

m--A--o--v~ m_A_o_v/v-o-oHumanand rabbit transferrin, rat liver plasma membrane

m-A-o\ m--A--o--v~

I

m~/k~O~V~V--OmO

m--~--o-!\

m_~__o_~

~

V-O-ore

I

Vesicularstomatitus virus

(IgG)Humanimmunoglobulin G

A

m--A--o--v\

,~V~O~O~

I

Bovineimmunoglobulin G (IgG)

F I G U R E 10-3 N-Glycosidically linked oligosaccharides. The oligomannosidic type is rich in mannose residues, whereas the complex type contains carbohydrate residues in the outer domain. Both types have a common core.

f'-

]

,j 9 A = galactose, 9 = mannose, ~ = L-fucose, 9 = N-acetylglucosamine, 9 = N-acetylneuramine acid. [Adapted, with permission, from N. Sharon and H. Lis, Special Report, Chem. Eng. News, p. 28 (March 30, 1981) @ 1981 by the American Chemical Society.]

may be found in adjacent hydroxyamino acid residues, or they may occur in close proximity, e.g., in glycophorin, human chorionic gonadotropin, and antifreeze glycoprotein. This last is found in the blood of Arctic and Antarctic fish species and other species on the eastern coast of North

ZX--

Collagen Ovine submaxillary mucin

A--F-l-m-A-~-

I m

Antifreeze glycoprotein, human immunoglobulin A, (IgA,), rat and rabbit brain glycoproteins Calf fetuin, glycophorin, human chorionic gonadotropin

dmh

I

F-1--AmFlm

A+ porcine submaxillary mucin

F I G U R E 10-4 O-Glycosidically linked oligosaccharides. The sugar residues vary from one to many and are not arranged in any particular pattern. A = Galactose; 8 = L-Fucose; [] = GalNAc; 9 -- N-acetylneuraminic acid. [Adapted with permission from N. Sharon and H. Lis, Special report, Chem. Eng. News, p. 28 (March 30, 1981). 9 1981 by the American Chemical Society.]

America. It contains a very high amount of carbohydrate, since every threonine residue of the glycoprotein is linked with a galactosyl-/3-(1 --+ 3)-N-acetylgalactosamine unit. The protein consists of the repeating tripeptide sequence of alanyl-alanyl-threonine. The presence of antifreeze glycoprotein, along with high concentrations of NaC1 in the blood, prevents water from freezing in blood vessels and permits survival at the low temperatures of polar seawater. This freezing-point depression by antifreeze glycoproteins has been attributed to their highly hydrated and expanded structure, which interferes with the formation of ice crystals. In all glycoproteins, the polypeptide component is synthesized first on the membrane-bound ribosomes of the rough endoplasmic reticulum; carbohydrate side chains are added during passage through the endoplasmic reticulum and Golgi apparatus. The carbohydrate additions involve specific glycosyltransferases and their substrates (uridine diphosphate sugars) and, in some glycoproteins, an oligosaccharide carrier known as dolichol (a lipid). (See also Chapters 16 and 25.) Glycoproteins can also be formed by addition of carbohydrate residues without any of the complex enzymatic pathways of carbohydrate addition. This process, known as nonenzymatic glycation, proceeds by the condensation of a monosaccharide, usually glucose, with certain reactive amino groups on the protein. The initial, labile Schiff base adduct slowly rearranges to the stable ketoamine or fructosamine form (Chapter 2). For example, a small fraction of hemoglobin A, the major hemoglobin of adult humans, is present in the red blood cells as glycated hemoglobin (HbAlc). The glycation of hemoglobin is a continuous process occurring throughout the 120-day life span of the red cell. In HbAac, glucose is incorporated via an N-glycosidic linkage into the N-terminal amino group of valine of each/3-chain. Enhanced levels of HbAlc occur in individuals with diabetes mellitus (Chapter 22), and measurement of glycated hemoglobin has been useful in monitoring the effects of therapy. Human serum albumin, which has a half-life of 19 days, is also subjected to nonenzymatic glycation, producing a stable condensation product known as fructosamine. Fructosamine is a generic term applied to the stable condensation product of glucose with serum proteins, of which albumin is quantitatively the largest fraction. Measurement of fructosamine concentration provides a means by which short-term (1-3 weeks) plasma glucose levels can be estimated, whereas measurement of HbAlc concentration reflects integrated plasma glucose levels over a longer period (2-3 months). Human lens proteins, or-,/3-, and ?'-crystallins, which have much longer life spans than other proteins in the body, also undergo agedependent, nonenzymatic glycation at the s-amino groups

156

CHAPTER 10

of their lysine residues. In diabetics, this process occurs twice as often as in normal individuals of comparable age. Lens cells, like red blood cells, do not require insulin for the inward transport of glucose. However, the extent of nonenzymatic glycation of crystallins is much lower than that of hemoglobin (about 2.4% versus 7.5% at age 50) because

tissues, cells, and intracellular domains. They function as protective barriers and as transducers of extracellular messages carried by the chemical agents because they have recognition sites that interact with metabolites, ions, hormones, antibodies, or other cells in a specific manner. This characteristic selectivity of membranes to interact with specific molecules confers unique properties on a given cell type. Within the cell, the membranes of organelles are highly differentiated and have properties consistent with metabolic function. Examples include electron transport and energy conservation systems in the mitochondrial membrane, protein biosynthesis in the rough endoplasmic reticulum, modification and packaging of proteins for export in the membranes of the Golgi complex, drug detoxification in the smooth endoplasmic reticulum, and light reception and transduction in the disk membranes or retinal cells. The membrane constituents are lipids (phospholipids, glycosphingolipids, and cholesterol; Figure 10-5), carbohydrates, and proteins. The ratio of protein : lipid : carbohydrate on a weight basis varies considerably from membrane to membrane. For example, the human erythrocyte membrane has a ratio of about 49:43:8, whereas myelin has a ratio of 18:79:3. The composition of the normal human erythrocyte membrane is shown in Table 10-2. All membrane lipids are amphipathic (i.e., polar lipids). The polar heads of the phospholipids may be neutral, anionic, or dipolar. The surface of the membrane bears a net negative charge. The distribution of lipid constituents in the bilayer is asymmetrical. For example, in the erythrocyte membrane, phosphatidylethanolamine and phosphatidylserine are located primarily in the internal monolayer, whereas phosphatidylcholine and sphingomyelin are located in the external monolayer. Lipids are organized in bilayers that account for most of the membrane barrier properties. Membrane proteins may be peripheral (extrinsic) or integral (intrinsic). Peripheral proteins are located on either side of the bilayer and are easily removed by ionic solutions, whereas integral proteins are embedded in the bilayer to varying degrees (Figure 10-6). Some integral proteins penetrate the bilayer and are exposed to both external and internal environments. By spanning both external and internal environments of the cell, these proteins may provide a means of communication across the bilayer that may be useful in the transport of metabolites, ions, and water, or in the transmission of signals in response to external stimuli provided by hormones, antibodies, or other cells. Because of hydrophobic interactions, isolation of integral proteins requires harsh methods, such as physical disruption of the bilayer and chemical extraction procedures using

1. The lens cells contain about one sixth the glucose of red cells. 2. The content of lysine is lower in crystallin compared to hemoglobin 3. The lysine residues are inaccessible in crystallins owing to the high content of/3-pleated sheet structures packed in a structured array oriented orthogonally to the lens optic axis, restricting rotational and translational movement. Crystallins contain almost no a-helix, whereas hemoglobin, a globular protein, possesses a high content of a-helical structure and can freely rotate in the fluid media, exposing the lysine residues. Crystallins constitute 90% of the soluble proteins of the lens cells (also called fiber cells). The human lens, a transparent, biconvex, elliptical, semisolid, avascular structure, is responsible for focusing the visual image onto the retina. The lens grows throughout life at a slowly decreasing rate, building layer upon layer of fiber cells around a central core and never shedding the cells. Crystallin turnover is very slow or nonexistent. In addition to nonenzymatic glycation, crystallins undergo other age-dependent, posttranslational modifications in vivo: formation of disulfide bonds and other covalent cross-links, accumulation of high-molecular-weight aggregates, deamidation of asparagine and glutamine residues, partial proteolysis at characteristic sites, racemization of aspartic acid residues, and photo-oxidation of tryptophan. Some of these processes contribute to the increasing amount of insoluble crystallins during aging. Nonenzymatic incorporation of carbohydrates into proteins in vivo can be extensive and may contribute to the pathophysiology of diabetes mellitus and galactosemia.

10.2 Cell Membrane Constituents Various aspects of the cell membrane are discussed throughout this text, and a brief introduction is presented here. The living system's ability to segregate from and protect itself against--and interact with and against---changes in the external environment is accomplished by membranes. In the body, membranes function at the level of

Heteropolysaccharides I: Glycoproteins and Glycolipids

p...

c~

0

.=.

o

o oo

ooo

cq cq oo ~

cSc;c;ocSc;c~

0

9

~-~ ~ "~

oo

v ' ~ 0 0

g

g

c~ oo c~

c~

~

.,..a

.,..~

9

c~

~

9

c~

c~ c~

9

o

*

157

158

CHAPTER10 Heteropolysaccharides I" 61ycoproteins and 61ycolipids

(a) A (D

Phosphoglycerides _

CH=----O~C

r Q.

=8 Glycerol moiety

CH - - O - - C . I

.

.

.

.

,,Fatty add ,/residues CH3

I

CH= CHs o~11 I+ -O ~ P'~, O ~ CH~-- CH=----N ~CH=

.m

o/.

' I,

=1 . . . . . .

Phosphatidylcholine (lecithin) Choline moiety

O

li

oH, CH - - O - - C CH=

CH=

o%_,io --

I

.../ P.,,< u , O--CH=----CH=-----NH= ~ Ethanolaminem o i e t y r "

"l"

C H - - NH+ ""•O--CHi--J

Phosphatidylethanolamine

Serine moiety

, !

Phosphatidylserine

: . . . . . . . . . .c. .o.o. .-. 8

II i~C~

CH3

CH ~ C

I

OH3

CH=

I

O~..,~O ', ,

i

~ OH

~

O

H

Phosphatidylinositol Inositol moiety

F I G U R E 11)-5 Structures of some membrane lipids.

synthetic detergents or bile salts to disrupt the lipid-protein interactions. Carbohydrate residues are covalently linked (exclusively on the external side of the bilayer) to proteins or lipids to form glycoproteins or glycolipids, respectively, both of which are asymmetrically distributed in the lipid bilayer (Figure 10-7). Fluidity of the membrane structure is determined by the degree of unsaturation of the hydrocarbon chains of the phospholipids and by the amount of cholesterol in the membrane. Hydrocarbon chains

with cis-double bonds produce kinks and allow a greater degree of freedom of movement for the neighboring alkyl side chains; hence, these unsaturated chains give rise to more fluidity than do saturated alkyl chains, which associate in ordered arrays. Cholesterol, an inflexible polycyclic molecule, is packed between fatty alkyl chains, the ring bearing the polar hydroxyl group interacting with the polar groups of phospho- and glycolipids. The presence of cholesterol disrupts the orderly stacking of alkyl side chains, restricts their mobility, and causes increased

159

SECTION 10.2 Cell Membrane Constituents

(b)

Phosphosphingolipids These lipids contain a long unsaturated hydrocarbon chain amino alchohol known as sphingosine: oH

C

I

CH3

HOH,C/I ~C " I

H

C,,I-~NO=

A derivative of sphingosine, in which a fatty acid is linked by an amide linkage, is ceramide: ~

Fatty acid residue

Amide,,,.--------~/C linkage H.N H

I

o

H~./?~cJ Primar~ I alchohol group~'''~" HO

H

~.

CH,

I

H

The product obtained when the alchohol hydroxyl group of sphingosine is esterifled with phosphorylcholine is sphingomyelln .The conformationsof phosphatidylcholine and sphingomyelin are similiar. O IJcI CH3 HN"

I OH ,C.... I ~,

OH,

%_//0 .~CH= H3C/!H"~3CH3

(c)

Glycosphingolipids (cerebrosides)

A derivative of ceramide that contains a monosaccharide unit (either glucose or galactose) linked in a 13-glycosidiclinkage is a cerebroside. These neutral lipids occur most abundantly in the brain and myelin sheath of nerves. The specific galactocerebroside, phrenosine, contains a 2-hydroxy-24-carbon fatty acid residue. Following is a structure of a glucocerebroside. O HN m

CH3

I

"o2"2

o"jCH2

C

c.,

3-Glycosidic linkage

FIGURE 10-5 (Continued) membrane viscosity. Thus, the lipid composition of the membrane at physiological temperatures can have significant effects on fluidity and permeability. Some correlation appears to exist between the concentrations of sphingomyelin and cholesterol in different membranes. Plasma membranes (e.g., red blood cells and myelin sheaths) are rich in both; the inner membrane of mitochondria contains neither.

Membrane proteins show considerable mobility in the plane of the bilayer (lateral motion). There is no evidence that proteins migrate from one side of the bilayer to the other. The frequency of reorientation of lipid components (flip-flop migration) is extremely slow or nonexistent, for thermodynamic reasons. Artificial membrane systems (liposomes) have increased our understanding of the dynamic nature of lipid

160

CHAPTER10 Heteropolysaccharides I: Glycoproteins and Glycolipids

(d) Cholesterol Cholesterol (3-hydroxy-5,6-cholestene) belongs to a family of compounds derived from a fused, reduced, nonlinear four-ring system of cycIopenta[ (x]-phenanthrene. Bile acids, steroid hormones, and vitamin D metabolites are derived from cholesterol.

H~ ~

Angular methyl

~

orou,:,s~--_____.~''-r ~ 1

\

lg

'F ~ 1T D "]" I

,..-x,, il-/{'-, 13-Hydroxylgroup

~H

21"" . ~ I ~

"

I

|Structural /

l,.,

/4.1

]

~'~ 2Gall31 "N 3GalNAc]31__> 3Gal/31----> 4Glc-Cer Fucc~ 1,7

The Lewis antigens are not components of developng red blood cells. They are acquired as glycosphingolipidsduring circulation by adsorption from plasma low- and high-density lipoproteins (Chapter 20).

P Locus P antigen pK antigen

GalNAc/31--4 3Galc~l---> 4Gal/31 ~ 4Glci31-Cer Galo~l---> 4Gal/31----> 4Glc]31-Cer

I Locus I antigen

Gall31 ___>4GlcNAc/31"N 6Gal/31---> 4-X* Gall31---> 4GlcNAc/31//"

i antigen

Gal/31 ~ 4GlcNAc]31---> 3Gall31---> 4-X* X* = GlcNAc]31----> 3Gal/31--> 4Glc/31-Cer

Gal=galactose, GalNAc=N-acetylgalactosamine, Glu=glucose, GluNAc=N-acetylglucosamine, Fuc=fucose (6-deoxy-L-galactose), Cer=ceramide (a sphingolipid).

precursor oligosaccharide. The classification of individuals into ABO types is based upon whether A or B antigens are present or not; type A individuals have the A antigen, type B, the B antigen, and type O, the H antigen, respectively. Normally, an antigen and its specific antibody are not present simultaneously. For example, individuals having type A red blood cells possess anti-B antibodies; those of type B possess anti-A antibodies; those of type AB have neither of these antibodies; and those of type O have both anti-A and anti-B antibodies (Table 10-5).

The antigenic variation of blood group substances is due to specific glycosyltransferases (the primary gene products) responsible for synthesis of the oligosaccharide determinants (the secondary gene products). Under appropriate experimental conditions, the blood type of red blood cells can be changed by addition or removal of specific carbohydrate residues. For example, when type 0 red blood cells are incubated with the galactose donor substrate (uridine diphosphate galactose) and the specific ot-galactosyltransferase enzyme, they are

168

CHAPTER10 Heteropolysaccharides I: Glycoproteins and Glycolipids

TABLE 10-5

ABO Blood Group System and the Agglutination Reaction* Blood Type

Type of Oligosaccharide Antigens Present on Red Blood Cells

Antibody in Serum

Types of Serum That Cause Agglutination When Mixed with Red Blood Cells

O A B AB

H A B A and B

Anti-A, anti-B Anti-B Anti-A None

None O, B O, A O, A, B

*Red blood cells from type O individuals can be donated to individuals with any other cells from type within the ABO system without causing agglutination. Individuals with type AB can accept red blood cells from all other types but cannot donate to individuals with other types except AB. Thus, type O individuals are known as universal donors and AB as universal acceptors.

converted to type B red blood cells. The reverse conversion can be accomplished by incubation of cells in the presence of ot-galactosidase, which removes the terminal galactose residue. Glycophorins A and B possess antigenic determinants for the M and N blood group antigens, respectively. Rh blood group antigens are clinically important due to their involvement in hemolytic disease of the newborn, transfusion medicine, and autoimmune hemolytic anemia. The designation of Rh stands for Rhesus because the antibody specificity was identical to that of antibodies generated in rabbits injected with red blood cells from Rhesus monkeys. The Rh blood group system consists of more than 50 nonglycosylated protein antigens; however, only five are commonly identified; these are encoded by two genes termed RHD and RHCED. Marked racial differences are observed for Rh alleles in different populations. Typically, 85% of all Caucasians are Rho(D)-positive. Anti-Rho(D) immunoglobulin G (IgG) prepared from human plasma is administered intramuscularly as means of passive immunization to Rho(D)-negative individuals exposed to Rho(D)-positive cells. Rh proteins have a molecular weight of about 32,000 and span the red blood cell membrane. Rh proteins are incorporated into the erythrocyte membrane after palmitolyation via thioester linkages involving free sulfhydryl groups of cysteine residues. The exact function of Rh proteins is not known, however, individuals lacking all Rh protein expression exhibit multiple red blood cell abnormalities including abnormal morphology and survival. This condition is known as Rhnull syndrome. A number of protein blood group antigens possessing a variety of functions are shown in Table 10-6. Blood group antigens including both oligosaccharides and proteins are expressed on other cell surface struc-

tures as well as on erythrocytes. These antigens function as receptors for a variety of infectious agents. Examples of oligosaccharide antigens include P antigen, which is the receptor for B 19 parvovirus; individuals who do not express this antigen show no serological evidence of past infections with this common childhood pathogen. Le b antigen is the epithelial receptor for Helicobacter pylori, which is the causative agent of peptic ulcer disease (Chapter 12). Lewis antigens function as receptors for a wide variety of pathogens including S. aureus, N. meningitidis, H. influenzae, N. gonorrhoeae, and

C. albicans. Blood group antigen-bearing proteins also serve as receptors for infection agents (Table 10-7). A notable example is the presence of receptors in the Duffy blood group antigens to Plasmodium vivax and Plasmodium knowlesi. Individuals who do not express Duffy antigens are resistant to infection by these malarial parasites.

10.4 Serum Glycoproteins Almost all serum proteins, with the notable exception of albumin, are glycoproteins. The sugar residues found most commonly in the outer domain of the oligosaccharides of these glycoproteins are galactose, Nacetylhexosamine, and sialic acid. L-Fucose is a minor constituent. The structure of a typical oligosaccharide chain of a serum glycoprotein is shown in Figure 10-14. The liver plays a major role in the synthesis and catabolism of these proteins. Glycoproteins lose their terminal sialic acid residues through the action of neuraminidase (sialidase) during circulation in the blood, which exposes the galactose residues (Figure 10-14). The resulting galactose-terminated glycoproteins, known as

SECTION 10.4

Serum61ycoproteins

169

TABLE 10-6 Functions of Blood Group Protein Antigens of Erythrocytes

Blood Group (abbreviation)

Locus Name

Protein

a. Proteins that contribute to the structural integrity Rhesus (Rh) Gerbich (Ge) Diego (Di), Wright (Wr) XK

RH GYPC AE1 Kx

Rh acylproteins Glycophorins C and D Anion channel protein Kx protein

DAF

Decay accelerating factor (CD55)

Fy

Chemokine receptor

AE1 JK AQP1

Anion channel protein Urea transporter Aquaporin 1 water channel protein

KEL YT

93kD protein, metalloproteinase (?) Acetylcholinesterase

b. Proteins with complement-related functions Cromer (Cr) c. Proteins with receptor functions Duffy (Fy) d. Proteins with transport functions Diego (Di), Wright (Wr) Kidd (Jk) Colton (Co) e. Proteins with enzymatic activity Kell (K,k,Kp,Js) Cartwright (Yt)

asialoglycoproteins, are taken up after binding to receptors on hepatocytes. The bound asialoglycoprotein is internalized by a process known as receptor-mediated endocytosis (Chapter 11) and subjected to lysosomal degradation. In TABLE 10-7 Blood Group Protein Antigens as Microbial Receptors*

Oligosaccharides

P Lewis (Leb) H, A

Escherichia coli, Parvovirus B 19 Helicobacter pylori Candida albicans

Proteins Glycophorin A (MN) Decay accelerating factor (Cromer) CD44 (Indian) Duffy AnWj

Plasmodium falciparum Echovirus, E. coli Poliovirus Plasmodium vivax, Plasmodium knewlesi H. influenzae

*Modified and reproduced with permission from M. J. Telen. Erythrocyte Blood Group Antigens: Polymorphisms of functionally Important Molecules. Seminars in Hematology33, 302 (1996).

liver disease, the plasma levels of asialoglycoproteins are elevated. The internalized glycoproteins are catabolized to their monomeric units by the lysosomal enzymes. The oligosaccharides are degraded sequentially by specified hydrolases, starting from the nonreducing termini. Hereditary deficiency of some of these enzymes has been reported: ot-o-mannosidase in mannosidosis, o~-L-fucosidase in fucosidosis, glycoprotein-specific ot-neuraminidase in sialidosis, and aspartylglycosaminidase in aspartylglycosaminuria. In these disorders, undigested or partially digested oligosaccharides derived from glycoproteins accumulate within the lysosomes. Keratin sulfate (a glycosaminoglycan, Chapter 11) also accumulates, presumably because the above enzymes are required for its degradation. These disorders are inherited as autosomal recessive traits and can be diagnosed prenatally. Clinically, they resemble mild forms of mucopolysaccharidosis (Chapter 11). There is no definitive treatment. Removal of senescent red blood cells from the circulation has been attributed to desialylation of the membrane glycoproteins. In vitro removal of sialic acid from human red blood cells and introduction of the modified cells into the circulatory system result in drastic shortening of their life span. However, aging and removal of red blood

170

CHAPTER10 Heteropolysaccharides I: Glycoproteins and Glycolipids

Outer domain ,

Inner domain

i

II--~O-~-y~

I__A__I+I/I--I--I~I

Protein backbone

,,

Asparagine--

i

Typical serum glycoprotein 1

+ l~Neuraminidase

/ X - - O - - Y ~ V - - O m O m Asparagine-AmO myJ Asialog lyco protei n F I G U R E 10-14 Oligosaccharide side chain of a serum glycoprotein. The removal of the terminal sialic acid residue by neuraminidase exposes the penultimate galactose residue. The resulting asialoglycoprotein is cleared from the circulation by a galactose receptor-mediated process in the hepatocytes. I = Sialic acid, 9 = N-acetylglucosamine, A = galactose, IIt = mannose.

cells depend on many other factors (e.g., inactivation of enzymes and oxidation of sulfhydryl proteins). If desialylation occurs normally as part of the physiological mechanism in clearing glycoproteins and cells of the circulatory system, the anatomical location of this process is not known. Other clearance systems for glycoproteins have been identified, e.g., one in the reticuloendothelial system that terminates with mannose and N-acetylglucosamine. Information of this type has potential application in targeting biologically active molecules to a specific tissue site. For example, mannose-terminated catalytically active lysosomal enzymes can be targeted to reticuloendothelial cells. Such targeting of lysosomal enzymes could be useful in the treatment of lysosomal enzymedeficiency diseases. The same principle can be applied to targeting drugs to specific tissue sites by coupling the drugs to glycoproteins with the appropriate terminal sugar residues.

10.5 Molecular Mimicry of Oligosaccharides and Host Susceptibility In the previous section we discussed the presence of oligosaccharide and protein blood group substances that cause susceptibility to certain pathogens. In this section we discuss an infectious agent that contains a common antigenic epitope with the host and that elicits antibodies that cause a disease. Guillain-Barr~ syndrome (GBS) is one such disease. GBS is an acute inflammatory neuropathy with progressive weakness in both arms

and legs leading to paralysis. It is a self-limited autoimmune disease. Most GBS cases have resulted from an antecedent, acute infection of bacterial or viral origin; in children GBS has also been identified following vaccination. One major cause of bacterial gastroenteritis is Campylobacter jejuni and this infection also may be followed by GBS. Some serotypes of C. jejuni possess liposaccharides that contain terminal tetrasaccharides identical to ganglioside GM 1. Antibodies made in response to the bacterial infection also attack GM1 which is widely present in the nervous system. The antibodies directed against the GM1 epitope cause an immune-mediated destruction of nerve fibers. Thus, the sharing of homologous epitopes between bacterial liposaccharides and gangliosides is an example of molecular mimicry, which causes disease. Host factors may also influence a person's susceptibility to GBS. Plasma exchange and intravenous immunoglobulin administration are used for immunomodulation in the therapy of GBS.

Supplemental Readings and References Extracellular Matrix M. H. Ascarelli and J. C. Morrison: Use of fetal fibronectin in clinical practice. Obstetrical and Gynecological Survey 52(Suppl.), S I (1997). R. O. Hynes: Integrins: Versatility, Modulation and Signalling in Cell Adhesion. Cell 69, I1 (1992). K. A. Piez: History ofextracellular matrix: A personal review. Matrix Biology 16, 85 (1997). J. Labat-Robert, M. Bihari-Varga, and L. Robert: Extracellular Matrix. FEBS Letters 268, 386 (1990). W. K. Stadelmann, A. G. Digens, and G. R. Tobin: Physiology and healing dynamics of chronic cutaneous wounds. American Journal of Surgery 176(Suppl. 2A), 265 (1998). W. K. Stadelmann, A. G. Digens, and G. R. Tobin: Impediments to wound healing. American Journal of Surgery 176(Suppl. 2A), 395 (1998).

Blood G r o u p Antigens E Agre and J-E Cartron: Molecular biology of Rh antigens. Blood 78, 551 (1991). C-H. Huang, E Z. Liu, and J. G. Cheng: Molecular biology and genetics of the Rh blood group system. Seminars in Hematology 37, 150 (2000). A. O. Pogo and A. Chaudhuri: The Duffy protein: a malarial and chemokine receptor. Seminars in Hematology 37, 122 (2000). M. Rios and C. Bianco: The role of blood group antigens in infectious disease. Seminars in Hematology 37, 177 (2000). M. J. Telen: Erythrocyte blood group antigens: Polymorphisms of functionally important molecules. Seminars in Hematology 33, 302 (1996). M. J. Telen: Red blood cell surface adhesion molecules: Their possible role in normal human physiology and disease. Seminars in Hematology 37, 130 (2000).

Supplemental Readings and References

Molecular Mimicry T. E. Feasby and R. A. C. Hughes: Campylobacter jejuni, antiganglioside antibodies, and Guillain-Barr6 syndrome. Neurology 51, 340 (1998). A. E Hahn: Guillain-Barr6 syndrome. The Lancet 352, 635 (1998). T. Lasky, G. J. Terraccians, L. Magder, et al.: The Guillain-Barr6 syndromes and the 1992-1993 and 1993-1994 influenza vaccines. New England Journal of Medicine 339, 1797 (1998). J. J. Ma, M. Nishimura, H. Mine, et al.: HLA and T-cell receptor gene polymorphisms in Guillain-Barr6 syndrome. Neurology 51, 379 (1998). A. H. Ropper and M. Victer: Influenza vaccination and Guillain-Barr6 syndrome. New England Journal of Medicine 339, 1845 (1998).

171 K. A. Sheikn, I. Nachamkin, T. W. Ho, et al.: Campylobacter jejuni lipopolysaccharides in Guillain-Barr6 syndrome. Neurology 51, 371 (1998).

Disorders of Red Blood Cell Membrane Skeleton E S. Becker and S. E. Lux: Hereditary Spherocytosis and Hereditary Elliptocytosis. In the Metabolic Bases of Inherited Disease, 7th ed., C. R. Scriver, A. L. Beaudet, W. S. Sly, D. Valle., (Eds.) McGraw-Hill, New York, 1995, p. 3513. S. C. Liu and L. H. Derick: Molecular anatomy of the red cell membrane skeleton: Structure-function relationships. Seminars in Hematology 29, 231 (1992).

CHAPTER

11

Heteropolysaccharides II: Proteoglycans and Peptidoglycans

11.1 Protein Fibers and Proteoglycans Connective tissues are composed of insoluble protein fibers (the glycoprotein collagen and the nonglycoprotein elastin) embedded in a matrix of proteoglycans (ground substance). The connective tissues bind tissues together and provide support for the organs and other structures of the body. Their properties depend on the proportion of different components present. A tissue of very high tensile strength, the Achilles tendon, is composed of about 32% collagen and 2.6% elastin, whereas an elastic tissue, the ligamentum nuchae, is composed of about 32% elastin and 7% collagen. The proteins and proteoglycans are synthesized by connective tissue cells: fibroblasts (generalized connective tissue), chondroblasts (cartilage), and osteoblasts (bone). Connective tissue also contains blood and lymphs vessels and various transient cells including macrophages and mast cells. Adipose tissue is a specialized form of connective tissue consisting of a collection of adipocytes (stores of triacylglycerol) that cluster between the protein fibers.

Collagen Collagens are extracellular proteins of connective tissue and they make up about one third of all body protein. They are a family of related glycoproteins in which hydroxylysyl residues provide the sites for attachment

of glucose, galactose, or an or(1 --+ 2) glucosylgalactose residue via a /3-O-glycosidic linkage (Figure 11-1). In brief, the synthesis of collagen can be considered to occur in two stages: intracellular and extracellular. The intracellular stage consists of the production of procollagen from precursor polypeptide chains that undergo, in sequence, hydroxylation, glycosylation, formation of a triple helix, and secretion. The extracellular stage consists of the conversion of procollagen to tropocollagen by limited proteolysis from the amino and carboxyl termini, self-assembly of tropocollagen molecules into fibrils, and finally cross-linking of the fibrils to form collagen fibers (Chapter 25). Collagen exists predominantly as fibrous protein; however, in the basement membrane of many tissues, including kidney glomeruli and the lens capsule, it is present in a nonfibrous form. The unique property of each connective tissue (e.g., the flexibility of skin, rigidity of bone, elasticity of large arteries, and strength of tendons) depends on the composition and organization of collagen and other matrix components.

Collagen Types More than 16 different types of collagen have been reported. They constitute the most abundant family of proteins in the human body. The collagens are encoded by 28 genes dispersed in at least 12 different chromosomes.

173

174

CHAPTER11 Heteropolysaccharides I1: Proteoglycans and Peptidoglycans HO

"\

H

NH3

OJ

example, human fibroblasts and smooth muscle cells synthesize both types I and III collagens; epithelial and endothelial cells synthesize type IV collagen; and chondroblasts synthesize type II collagen. Structure and Function

II

0 [

I

Polypeptide chain 13-o-Glucosylhydroxylysylresidue

HO

HO

0 HO 0

o~

HO~~

~H'~C/

H

OHo~C I

II o I

Polypeptide chain OH D-Glucosyl ( o~1-4"2)-O-13- D-galactosylhydroxylysylresidue F I G U R E 11-1 Glycosides of the collagen polypeptide chain.

The individual genes are identified by both collagen type and the polypeptide chain. For example, COL1A2 gene codes for type I collagen (COL l) and or2 (A2) polypeptide chain. The tissue distribution of collagens also varies (Table 11-1 ). Type I collagen consists of two identical chains of ot 1(I) and one chain of or2; it is the major connective tissue protein of skin, bone, tendon, dentin, and some other tissues. Type II collagen consists of three identical chains ofc~ 1(II); it is found in cartilage, cornea, vitreous humor, and neural retinal tissue. Type III collagen consists of three identical chains of ot 1(III); it is present, along with type I collagen, in skin, arteries, and uterine tissue. Type IV collagens are composed of ot 1(IV) to ol5(IV) and are found in the basement membranes of various tissues. Unlike collagen types I, II, III, V, and XI which are fibrillar, type IV collagen is not fibrillar in structure. The synthesis of collagens in cultured cells has aided the understanding of collagen biochemistry. The tissue specificity of various types of collagen is also reflected in cultured cells obtained from appropriate tissues. For

Each collagen molecule contains three polypeptide chains coiled around each other in a triple helix. These chains are called or-chains and are designated by Roman numerals according to the chronological order of their discovery. The three polypeptide chains may be identical or may consist of two identical chains and one dissimilar chain. Each ot-polypeptide chain consists of about 1000 amino acid residues, of which every third following a glycine is also a glycine. Thus, the molecular formula of an u-chain may be written as (Gly-X-Y)333, where X and Y represent amino acid residues other than glycine. In mammalian collagens, about 100 of the X-positions are occupied by proline residues, and 100 of the Y-positions are occupied by 4-hydroxyproline residues. At a few X-positions, 3-hydroxyproline residues are present; however, they only occur next to 4-hydroxyproline. Most hydroxyproline residues are present as the trans isomers. Although all collagen polypeptides have the general structure (Gly-X-Y),, differences between the various collagen types are associated with the particular sequences of amino acid residues in the X and Y positions. Hydroxyproline residues are not common in proteins; other than the collagens, hydroxyproline residues are found in elastin, acetylcholinesterase, and the C lq subcomponent of the complement system (Chapter 35). Another unique amino acid residue found in collagen is hydroxylysine, which occurs at the Y-position. The number of hydroxylysine residues per polypeptide chain lies in the range of 5 to 50. The hydroxylysine residues provide the sites for/~-O-glycosidic linkage with galactose or glucose or ct(1 --+ 2) glucosylgalactose. The collagens differ in their ratio of monosaccharide to disaccharide residues, as well as in their total carbohydrate content. For example, the carbohydrate contents of collagen from skin, cartilage, and basement membrane are about 0.5%, 4%, and more than 10%, respectively. Because collagen biosynthesis involves intracellular and extracellular posttranslational modifications (e.g., hydroxylation, glycosylation, fibril formation, and formation of cross-links), a given genetically determined collagen may show a great deal of heterogeneity. This is particularly true for type I collagen. Collagen also contains alanine residues in relatively high quantities. The only amino acid not found in collagen

~ 9

~'.~

~

~

~

~~

o=

~

"=

o

~

~

o= ~

~

~

~~

-

:.~

c~

9

9

.~-.~

~

9,...~

"~

;;>

c.~

~

~

~

~

.,..~

o

o

c.~

~

E

.~

~

..

~

~

"a

~

o

"~

~=

~

c.~

"~

O

~ o

E~ ~

""

.~

"-==

~

~=

~.

R,

~o

~ r~

,.0 c.=

9 ~,,,i

r~

x

C',l

C~O

%

9

.;>

;>

;>

;>

X.

X

X

%

175

1~6

is tryptophan (an essential amino acid). Collagen consists essentially of four amino acids in abundant quantities and negligible amounts of almost all other amino acids. For this reason, collagen is an inferior protein nutritionally. It is very insoluble in water and, for the most part, indigestible. However, collagen can be converted to soluble and digestible products by hydrolysis of some covalent bonds (by the use of heat or by certain plant proteinases) to yield gelatin. In the human body, collagen is resistant to most proteinases. Polymerized fibrillar collagen is a stable component of the extracellular matrix and its turnover rate is insignificant except in areas where tissue remodeling and repair occur. However, nonfibrillar collagens (e.g., type IV) are susceptible to proteolytic attack. Specific proteases (collagenases) cleave collagen at specific sites. The amino acid sequence of collagen produces its unique secondary and tertiary structures. A tropocollagen molecule contains three polypeptide chains, each coiled into a left-handed helix having about three amino acid residues per turn (Figure 11-2). This type of helix is unique to collagen and differs significantly from the c~-helix in its periodicity and dimensions (Chapter 4). The three helical polypeptides twist tightly around each other, except for the two short nonhelical regions at the C and N termini to form a right handed triple-stranded superhelix. Tropocollagen has a molecular weight of about 300,000, a length of 300 nm, and a diameter of 1.5 nm. The glycine residue, which occurs at every third position, has the smallest R-group and is thus able to fit into the restricted space where the three polypeptide chains are closest to each other. Since the proline and hydroxyproline residues hinder free rotation around the N-C bond (Chapter 4), the polypeptide chain has a rigid and kinked conformation. These stereochemical properties are responsible for the superhelix. Hydrogen bond formation between the NH group of a glycyl residue in one chain and the CO group of a prolyl or other amino acid residue in the X-position of an adjacent chain stabilizes the triple helix. The hydrogen bond between the glycyl residue of one chain and the prolyl residue of another chain is direct. If the prolyl residue is replaced by any other amino acid, the interchain hydrogen bond occurs through a water-bridged structure. These bonds are further stabilized by hydrogen bonding with the hydroxyl group of the trans-4-hydroxyprolyl residue, which occurs at the Y-position. Additionally, the chains are held together by covalent linkages involving lysine residues (Figure 11-3). Polypeptide chains consisting only of glycine, proline, and hydroxyproline residues (in that order) form an extremely stable triple helix. Furthermore, the stability (thermal) of the triple helix decreases in the following order for repeating sequences of the chain: Gly-Pro-Hyp >

CHAPTER11 Heteropolysaccharides I1: Proteoglycans and Peptidoglycans

Gly

~---Onl y Gly

fo) Only Gly ~ --

0.5 nm

(a) F I G U R E 11-2 Structure of tropocollagen. (a) The coiling of three left-handed helices of collagen polypeptides. The dotted lines indicate hydrogen bonds. (b) The right-handed triple-stranded superhelix. The curved arrow indicates the covalent linkage between two chains. [Reproduced with permission from I. Geis and R. E. Dickerson.]

Gly-Pro-Y > Gly-X-Pro > Gly-X-Y. In a given polypeptide chain of native tropocollagen, about one third of the molecule contains the Gly-Pro-Hyp sequence and two thirds involve Gly-X-Y, which decreases the stability of the triple helix. Amino acid residues other than proline and hydroxyproline that occupy the X- and Y-positions decrease helix stability but are essential for the next level of organization of collagen--the formation of microfibrils.

SECTION11.1 Protein Fibers and Proteoglycans Polypeptide chain

f

O

Polypeptidechain (helical region) 0

H

0

II I ~C~N~

I

Lysyl ~C~H oxidase t" ~, ; H2 2H20 2NH4 H.,,,. //.O

II

I

O

II

I

,

I

\

~N /

O

H

II

I

II

O

I

I

H

Dehydrohydroxylysinonorleucine

i

Polypeptide chain (nonhelical region) (b)

Formation ofdehydrohydroxylysinonorleucine linkage.

Polypeptidechains(nonhelicalregions)

Polypeptidechain (helicalregion) O H

II

~C /

N

i

Polypeptide (a) chain Formation of aldol condensation product--linkage involving nonhelical regions of the collagen molecule.

/C...~.,..,./N ~ /~H

c./C~N /

II

I

C--H

H~

O

H

N II

-H~O

Allysine residue

N.../

i

,

HO

H

~N /

O

H

i

HO

I~#snd~e

H--N

O H Aldol condensation product

~C

~H

Hydroxy-

/

II

I

~H

~H C / ~C=O ~kk J H

~c/C~

~N ~

c~ H

H

II I /%/N~

H

II

I

HO-~/

Aldol condensation (-H20)

H

O

H

II

/C,,,._/N~

"H

H~

~G /

O

H

II I ~C...c~N~

~Al~"~y~ine ~ residue

Lysine residue

H2N

O

,

,

H

II

177

O

H

II

/C~_/N~ /C~H

I

O

H

II

I

/C~c/N ~ ~H

O

\

/

Hydroxy-

'r S n,eue

H2N

-H20 "

~1 ii

H~c~.O

H-- C

~c'/C'H'" N/

II

I

O

H

, , Polypeptide chain (nonhelicalregion)

Amadori rearrangement

/,/

O

NH2

0

I

H/C~cH2 ~ L HQ ~

o~O

//o__~

H--N I H--C--H

OH

OH Hydroxyallysine H //residue

II

~o.

Condensation.~

(severalsteps)" L.~..~/

I

OH H~

CH2 OH

H~

~C"/II ~ NI/

o

H

I

(an aldimine)

H~ / : N - - H CH2~C C__ O

~c/C~ II

N/ I

H~

O

H

~N/C~c

II

(a ketamine)

(c)

Formation of dehydrohydroxylysinohydroxynorleucine(I), a n aldimine, and hydroxylysino-5-keto-norleucine(II), a ketamine. The linkage in II is more stable.

I

~

H~

~c/C~ N/

II

H O Polypeptidechain (helicalregion)

J H

II O

(d) Formation of pyridinoline cross-linkage, a polyfunctional cross-linkage.

F I G U R E 11-3 Formation of cross-links in collagen. All cross-links are derived from lysyl and hydroxylysyl amino acid residues. The initial step is the oxidative deamination of the e-NH2 group of two amino acid residues located at certain strategic positions (e.g., short nonhelical segments at both ends of the collagen molecule), with the formation of corresponding aldehyde-containing residues, allysine and hydroxyallysine. This reaction occurs extracellularly and is catalyzed by lysyl oxidase, a copper-dependent enzyme. The aldehyde groups react spontaneously with other aldehyde groups located in the adjacent chain of the same molecule or adjacent molecule. The structures do not show carbohydrate moieties for the sake of clarity.

1~8

Hydroxylysine glycosides occur at the Y-position and may play a role in determining fibril diameter. The side chains of these amino acids project outward from the center of the triple helix, permitting hydrophobic and ionic interactions between tropocollagen molecules. These interactions determine the manner in which individual tropocollagen molecules aggregate to form microfibrils initially, then larger fibrils, and eventually fibers. The microfibril, about 4 nm wide, consists of four to eight tropocollagen molecules that aggregate in a highly ordered and specific manner owing to interactions of amino acid residues at the X- and Y-positions. In this ordered arrangement, each molecule is displaced longitudinally by about one-fourth of its length from its nearest neighbors. The longitudinally displaced tropocollagen molecules are not linked, and there is a gap of about 40 nm between the end of one triple helix and the beginning of the next (Figure 11-4). These holes may provide sites for deposition of hydroxyapatite [Cal0(PO4)6(OH)2] crystals in the formation of bone (Chapter 37). Electron microscopic studies of negatively stained collagen fibrils reveal alternating light and dark regions. The light region, where the stain does not deposit, represents the overlapping of

F I G U R E 11-4 Packing of collagen polypeptides into a fibril. The individual tropocollagen molecules are quarter-staggered with respect to their nearest neighbors by a distance of 68 nm (D). A gap of about 40 nm (0.6 D) known as the hole region comes between the end of one tropocollagen molecule and the beginning of the next. The overlap zones are about 27 nm (0.4 D) in length and do not contain any hole zones. [Modified and reproduced, with permission, from A. Cohen, Ed.: Rheumatology and Immunology (W.B. Saunders, 1979).]

CHAPTER11 Heteropolysaccharides I1: Proteoglycans and Peptidoglycans

tropocollagen molecules; the dark region corresponds to a hole where the stain is deposited. The tensile strength of collagen fibrils is determined by covalent cross-links involving lysyl and hydroxylysyl side chains. The packing arrangement of tropocollagen molecules provides tensile strength and also prevents sliding of molecules over one another. This organization does not allow for stretching, which does occur in elastin. The nature and extent of crosslinking depend on the physiological function and age of the tissue. With age, the density of cross-linkages increases, rendering connective tissue rigid and brittle. The arrangement of fiber bundles varies with the tissue; it is random in bone and skin, sheet-like in blood vessels, crossed in the cornea, and parallel in tendons.

Turnover of Collagen and Tissue Repair The catabolism of collagen in the connective tissue matrix is carried out by enzymes known as collagenases. Part of the degradation may also involve neutral proteinases. A metalloenzyme specific for collagen catalyzes the cleavage of the triple helix at a single peptide bond, located at a distance from its N terminus corresponding to about three fourths the length of the tropocollagen molecule. The cleavage sites in type I collagen are the peptide bonds of Gly-Ile of the ot 1-chain and of Gly-Leu of the c~2-chain. The role and regulation of collagenase activity in vivo are unclear. The activity of the enzyme may be regulated via the formation of an enzyme-inhibitor complex or by the activation of a proenzyme, or both. A substantial amount (20-40%) of newly synthesized polypeptide chains of collagen undergo intracellular degradation. This degradation, which appears to occur in lysosomes, may be important in regulating the amount of collagen synthesized and in removing any defective or abnormal polypeptide chains that may be synthesized. Turnover of collagen in humans has been estimated by measurement of urinary hydroxyproline, which occurs mostly in the form of a peptide. Hydroxyproline makes up about 9-13 % of collagen residues and is not reutilized. Two aspects of hydroxyproline metabolism affect its use in the assessment of the true rate of collagen turnover: 1. Hydroxyproline is rapidly metabolized in a pathway initiated by the enzyme hydroxyproline oxidase (Chapter 17). 2. Hydroxyproline is present in C lq, which is synthesized at a rate estimated to be about 4.5 mg/kg per day. The first aspect has been overcome by measurement of urinary hydroxyproline in individuals with an inherited deficiency of hydroxyproline oxidase. Such persons excrete

SECTION 11.1

Protein Fibers and Proteoglycans

0.3 g of hydroxyproline per day in urine, which corresponds to about 2.25 g of hydroxyproline-containing proteins, most of which is collagen. Total protein catabolism in a normal well-fed adult is about 300 g; thus, collagen catabolism constitutes only about 0.8% of total protein catabolism. Collagen turnover has important clinical implications. The location, amount, type, and form of collagen depend on the coordinated control of its synthesis and degradation. In tissue injury (physical, chemical, infectious, or radiation), repair process comprises regeneration and fibrous connective tissue formation. Regeneration is the most desirable form of repair: a cut surface of an epidermis is replaced with new epidermis; scattered dead liver cells are replaced with new liver cells. Degradation products of type I collagen, namely N-telopeptides, C-telopeptides, hydroxyproline, and the collagen cross-links pyridinolone and deoxypyridinolone, have been used as markers of osteoclast activity in bone resorption. Collagen metabolite markers used for bone formation reflecting osteoblast activity are procollagen type I carboxyterminal propeptide and procollagen type I N-terminal propeptide. Both of these propeptides are cleaved byproducts generated from the conversion of procollagen to tropocollagen. Osteocalcin and bone-specific alkaline phosphatase are also used as bone formation markers. Biochemical markers of bone turnover are used for assessing and monitoring therapy in a number of diseases of bone including osteoporosis (Chapter 37). The cells of the body fall into three groups according to regenerative capacity: labile, stable, or permanent. The labile cells multiply throughout life and are maintained at an optimal level by continual proliferation of reserve cells. Labile cells are found in all epithelial surfaces, the spleen, and lymphoid and hematopoietic tissues. Stable cells have the potential to regenerate but do not normally undergo replication. However, under appropriate stimuli, they can proliferate rapidly. Stable cells include parenchymal cells of all glandular organs of the body and mesenchymal cells (e.g., fibroblasts, smooth muscle cells, osteoblasts, chondroblasts, and vascular endothelial cells). Permanent cells do not undergo significant replication postnatally. Nerve cells, skeletal muscle, and cardiac muscle cells are permanent cells. In fibrous tissue scar formation (a tough mass of collagen), normal cells are permanently lost. Many tissue injuries are repaired partly by regeneration and partly by scar formation (e.g., healing of dermal cut, improperly united bone fracture, and damaged liver). In tissues with permanent cells, injury and repair result in the formation of scar tissue only (e.g., myocardium). Although the liver normally undergoes repair by regeneration, chronic liver disease

119

resulting from ethanol abuse, some forms of malnutrition, viral infections, or exposure to chemicals can lead to hepatic fibrosis (cirrhosis) with distortion of the liver architecture by newly synthesized collagen fibers. In hepatic fibrosis, decreased collagenolytic activity and increased collagen synthesis may determine the net collagen deposition. A defect in the turnover of collagen may be responsible for the pathogenesis of idiopathic pulmonary fibrosis. This usually fatal disorder of the lungs is characterized by chronic inflammation of alveolar structures (alveolitis) and progressive interstitial fibrosis. An active collagenase in the lower respiratory tract may be responsible for sustained collagen lysis followed by disordered resynthesis. In rheumatoid arthritis, collagenous tissues of the joint (including articular cartilage) are eroded by collagenases and neutral proteinases derived from proliferating cells of the synovial membrane and leukocytes. Therapeutic approaches may depend on regulation of collagen synthesis, breakdown, or both. In a large family with osteoarthritis, which is a disorder of progressive degeneration of joint cartilage, in all affected members the ot(II) collagen contained a single-base mutation converting the codon for arginine at position 519 to a codon of cysteine, an amino acid not found in normal ot(II). In unaffected members of the family, this mutation does not occur. Other disorders of collagen metabolism, heritable or acquired, are discussed in Chapter 25. Elastin

Structure and Function

Elastin is a fibrous, insoluble protein that is not a glycoprotein but is present with collagen in the connective tissues. Connective tissues rich in elastic fibers exhibit a characteristic yellow color. Elastic fibers are highly branched structures responsible for physiological elasticity. They are capable of stretching in two dimensions and are found most notably in tissues subjected to continual high-pressure differentials, tension, or physical deformation. Elastin imparts to these tissues the properties of stretchability and subsequent recoil that depend only on the application of some physical force. Tissues rich in elastic fibers include the aorta and other vascular connective tissues, various ligaments (e.g., ligamentum nuchae), and the lungs. Microscopically, elastic fibers are thinner than collagen fibers and lack longitudinal striations. Elastin fibers can be separated into amorphous and fibrillar components. The amorphous component consists of elastin, which is characterized by having 95% nonpolar amino acid residues and two unique lysine-derived amino acid residues, desmosine and isodesmosine.

181]

CHAPTER11 HeteropolysaccharidesI1"Proteoglycansand Peptidoglycans

A major fibrillar component is a glycoprotein called The structure of fibrillin consists of several cysteine-rich motifs and exhibits a multidomain organization similar to epidermal growth factor. The structure is stabilized by disulfide linkages. Fibrillin monomers undergo aggregation extracellularly into supramolecular structures. The fibrillar structures have a diameter of 10-12 nm and surround the amorphous component elastin. During fetal development, the fibrillar component appears first in the extracellular matrix. With increasing fetal age and with maturation of the fibers the amorphous component is deposited within the framework of the microfibrillar bundles producing mature, elastic microfibrils. Fibrillin is encoded by a gene located in the long arm of chromosome 15. Mutations in the fibrillin gene lead to an autosomal dominant trait known as Marfan's syndrome. The incidence of this disorder is 1:10,000, and 15-30% of cases are caused by new mutations in the fibrillin gene. Consistent with the function of fibrillin in the elastic connective tissues, the clinical manifestations present as disorders of cardiovascular, musculoskeletal, and opthalmic systems. For example, dissecting aneurysm of

fibrillin.

the aorta, preceded by a dilatation, is a potentially fatal cardiac manifestation. Mature elastin is a linear polypeptide, tropoelastin, which has a molecular weight of about 72,000 and contains about 850 amino acid residues. Although glycine accounts for one third of the residues, the repeat sequence GlyX - Y characteristic of collagen is not present in elastin. Instead, glycine residues are present in the repeat units Gly-Gly-Val-Pro, Pro-Gly-Val-Gly-Val, and Pro-GlyVal-Gly-Val-Ala. Elastin is relatively rich in the nonpolar amino acids; alanine, valine, and proline. In contrast to collagen, only a few hydroxyproline residues are present in elastin. Elastin contains no hydroxylysine or sugar residues. A feature of mature elastin is the presence of covalent cross-links that connect elastin polypeptide chains into a fiber network. The major cross-linkages involve desmosine and isodesmosine, both of which are derived from lysine residues. Several regions rich in lysine residues can provide cross-links. Two such regions that contain peptide sequences that are repeated several times in tropoelastin have the primary structure-Lys-Ala-Ala-Ala-Lys- and

R3

H I

O II

--N,,~.C--

I 1=14

I O---C~ H--N H ' ~ C ~ 8 I

-

I

H/C'~'-O

/N--H 8r " ~ ' ' ~

1 c e

CH

Xc=o

I

e c :

o

NH, ye 8

XkX~ndensation ?

__cZCH'LN,

II O

I

H

R,

,

Desmosine cross-link

F I G U R E 11-5 Formation of desmosine and isodesmosine covalent cross-links in elastin. Three allysine residues (R2, R3, and R4) and one lysyl residue (R1) condense to give a desmosine cross-link. The allysine residues (e-aldehydes) are derived from the oxidative deamination of lysyl residues. The isodesmosine cross-link is formed similarly, except that it contains a substitution at position 2 rather than at position 4, along with substitutions at 1, 3, and 5 on the pyridinium ring.

SECTION 11.1

Protein Fibers and Proteoglycans

-Lys-Ala-Ala-Lys-. The clustering of lysine residues with alanine residues provides the appropriate geometry for the formation of cross-links. Cross-linking elastin occurs extracellularly. The polypeptides are synthesized intracellularly by connective tissue cells (such as smooth muscle cells), according to the same principles for the formation of other export proteins (e.g., collagen; see Chapter 25). After transport into the extracellular space, the polypeptides undergo crosslinking. A key step is the oxidative deamination of certain lysyl residues, catalyzed by lysyl oxidase (a Cu 2+ protein). These lysyl residues are converted to very reactive aldehydes (ot-aminoadipic acid 6-semialdehyde) known as allysine residues. Through an unknown mechanism, three allysine residues and one unmodified lysine residue react to form a pyridinium ring, alkylated in four positions, which is the basis for the cross-links (Figure 11-5). The cross-links may occur within the same polypeptide or involve two to four different polypeptide chains. Current models of elastin structure suggest that only two polypeptide chains are required to form the desmosine cross-link, one chain donating a lysine and an allysine residue, the other contributing two allysine residues. Elastin polypeptides are also cross-linked by condensation of a lysine residue with an allysine residue, followed by reduction of the aldimine to yield a lysinonorleucine residue (Figure 11-6). These linkages are present to a much lesser extent than in collagen. Formation of cross-linkages in elastin can be prevented by inhibition of lysyl oxidase. Animals maintained on a copper-deficient diet and those administered lathyrogens such as #-aminopropionitrile or o~-aminoacetonitrile develop connective tissue abnormalities. Lathyrogens are so-called because of their presence in certain peas of the genus Lathyrus. They are noncompetitive inhibitors of lysyl oxidase, both in vitro and in vivo. Consumption of certain types of sweet pea (e.g., Lathyrus odoratus) can lead to lathyrism. In osteolathyrism, the abnormalities involve bone and other connective tissues, the responsible compounds being the above-mentioned nitrites. The toxic agent responsible for neurolathyrism, #-N-oxalyl-Loe,#-diaminopropionic acid, has been isolated from Lathyrus sativus but its mode of action is not known. Thus, copper deficiency or administration of lathyrogens in animals prevents the formation of the highly insoluble cross-linked elastin but results in the accumulation of soluble elastin. Soluble elastin obtained in this manner is particularly useful in structural studies. Purification of the insoluble elastin without disruption of the integrity of the polypeptide chains is a formidable task. Several models of the macromolecular structure of elastin have been suggested to account for its elasticity: cross-linked globular elastin subunits, cross-linked

181

I

o=cx

I

c."~

HC ~ , . / n--,~ I

Y 13~

~.C He I I C/ H/' t ~ - - N - - H

e

N 6

..u n,

Lysine residue

~

~

fellYi~ue

1

I O--C_

/c=o

L. I N

I

-

I

C

I"-) H

I

I

,,-,,OH

._N ~ ~

c=o

~

~

__

I]1 H

(Schiff base) -H20

I

O--C~ HC~ ~ H--N/ ~

H ~

I

~

,-,/ ~ N ~ t ~ ~ Oehydrolysinonorleucine cross-link

I

sC---O CH ~N--H

I

/

[ Reduction

I O--C~

HC~

H__N /

V

A

A V

H

H

~,',/ ~N/~'~""L'LN__

HI

I

I .C=O

C~

H

I

Lysino no rleucine cross-link

FIGURE 11-6 Formationof the lysinonorleucinecross-linkbetweenan allysineand a lysine residuein elastin. random elastin chains, the oiled-coil model, and the fibrillar model. The fibrillar model incorporates a unique conformation due to the presence o f a glycine residue followed by an amino acid residue with a bulky R-group (e.g., Pro-Gly-Gly-Val). This type of sequence gives rise to near right angle turns (#-turns; Chapter 4) in the polypeptide backbone. The oiled-coil model assumes that each polypeptide chain is fibrillar and consists of alternating segments of cross-linked regions and "oiled coils," each chain being linked to many others to form a threedimensional network similar to a mattress spring. It is postulated that these coils can stretch up to 2-2.5 times their original length. The driving force for recoil of elastic fibers is due to a change in entropy.

Turnover of Elastin The turnover rate of mature elastin in healthy persons is relatively low. Insoluble elastin in healthy elastic tissue is usually stable and subjected to minimal proteolytic degradation. In several clinical conditions (e.g., emphysema, advanced atherosclerosis, pancreatitis), increased degradation of fragmentation of elastic fibers may play a significant role. The interaction between insoluble elastin and soluble elastolytic enzymes, and the regulation of these enzymes, may shed light on certain cardiovascular diseases, in view of the role of elastin in arterial dynamics.

1~

CHAPTER11 Heteropolysaccharides I1: Proteoglycans and Peptidoglycans

Elastolytic proteinases (elastases) are found in pancreatic tissue, polymorphonuclear leukocytes, macrophages, and platelets. These enzymes exhibit broad specificity. They catalyze preferential cleavage of peptide bonds adjacent to aliphatic amino acids, namely, glycine, alanine, and valine, which are present in high amounts in elastin. Elastases degrade elastin at neutral or slightly alkaline pH. The pancreas secretes elastase as an inactive precursor (zymogen) known as proelastase, which is converted to its active form by trypsin in the duodenum; elastase takes part in the digestion of dietary proteins (Chapter 12). This enzyme has a structure homologous to that of other pancreatic serine proteinases, including the amino acid sequence at the active site (Chapter 6). Polymorphonuclear leukocyte elastase is also a serine proteinase. Elastases are inactivated by serum otl-proteinase inhibitor and otz-macroglobulin; thus, their proteolytic action may be checked to prevent indiscrimination digestion of elastin-containing tissues (Chapter 6). An enzyme that degrades only soluble forms of elastin and exhibits trypsin-link activity has been described. Its biological significance is unknown. Several

heritable and acquired disorders of elastin are known (see Chapter 25).

Proteoglycans Proteoglycans are high-molecular-weight, complex molecules with diverse structures and functions. They are polyanionic substances containing a core protein to which at least one glycosaminoglycan (also known as mucopolysaccharide) chain is covalently attached. Proteoglycans are major components of connective tissue and participate with other structural protein constituents, namely, collagen and elastin, in the organization of the extracellular matrix.

Types, Structures, and Functions of Glycosaminoglycans Six classes of glycosaminoglycans have been described. All are heteropolysaccharides and contain repeating disaccharide units. The compositions and structures of the repeating disaccharide units are shown in Table 11-2 and Figure 11-7. In five glycosaminoglycans, the

TABI~E 1I-2

Compos#ion, Properties, and Distribution of Glycosaminoglycans Glycosaminoglycan (and range of molecular weight)

Amino Sugar

Uronic Acid

Type of Sulfate Linkage (extent of sulfation is variable)

Hyaluronate (4 to 80 x 106)

D-Glucosamine

D-Glucuronate

None

Chondroitin sulfate (5,000 to 50,000)

D-Galactosamine

D-Glucuronate

4-0- and/or 6-0-sulfate on galactosamine

Dermatan sulfate (15,000 to 40,000)

D-Galactosamine

L-Iduronate, D-Glucuronate

Keratan sulfate (4,000 to 19,000)

D-Glucosamine

None (but contains D-Galactose)

Heparan sulfate (104 to 105 )

D-Glucosamine

Heparin (103 to 106)

D-Glucosamine

D-Glucuronate (major), L-Iduronate (minor) L-Iduronate (major) D-Glucuronate (minor)

4-0-sulfate on galactosamine; 2-0-sulfate on iduronate 6-0-sulfate on both carbohydrate residues 6-0-sulfate and N-sulfate (or N-acetyl) on glucosamine; 2-0-sulfate on iduronate 2-0-sulfate on iduronate; 6-0-sulfate and N-sulfate (or N-acetyl) on glucosamine

Tissue Distribution Connective tissues, cartilage, synovial uid, vitreous humor, umbilical cord Cartilage, bone, skin, cornea, blood vessel walls Skin, heart valve, tendon, blood vessel walls Cartilage, cornea, intervertebral disks Lung, blood vessel walls, many cell surfaces Lung, liver, skin, intestinal mucosa (mast cells)

SECTION11.1 Protein Fibers and Proteoglycans

183

Hyaluronic Acid

~~

co~ \ "xZ-----~O\

HO ----~ O

CH20 H O I Ac

[glucuronate O 1, 3-NacetylglucosamineO 1, 41-

Chondronitin Sulfate / Dermatan Sulfate ~o~ --0

0

no i-

,~,o

o

~u2 \ ~ 0

OR_

oH

~ . SO~

L

"'J'x) / CH

~fi-.x~o~

I Ac [glucuronate-13 1, 3-Nacetylglucosamine-13 1, 4][ 4 or 6 sulfate

Dermatan Sulfate [iduronate-o~ 1, 3 ...]

Keratan Sulfate

OH(S030) dH

/0...~

\'~.s \

-

OH

\

HO

OSOp

/

I

~

I Ac

CH 2

~

\ XO'~'-'O~

[Nacetylglucosamine-[31, 3-galactose-[~ 1, 4]-

I

I

6-sulfate

6-sulfate

Heparan Sulfate/Heparin

IO~

CO ~

b../LLo

OR /

/

~ .o

HO

OH

. . O ~

"o~

CH 2

~2----..L~o

(o, .) . # - Rn /

so~ (orAc) -[glucuronate-~ 1, 3-...]-[iduronate-c~ 1, 4-Nsulfate glucosamine-~ 1, 4]I (acetate) I 2-sulfate 6-sulfate

F I G U R E 11-7 Basic repeating disaccharide units of six classes of glycosaminoglycans. Chondroitin sulfate and dermatan sulfate differ in type of principal uronic acid residue; the former contains glucuronate and the latter iduronate. Heparin and heparan sulfate differ in that heparin contains more iduronate 2-sulfate and N-sulfated glucosamine residues than heparan sulfate. In the sulfate-containing glycosaminoglycans, the extent of sulfation is variable. All of these molecules have a high negative charge density and are extremely hydrophilic. [Reproduced with permission from V. C. Hascall and J. H. Kimura; Proteoglycans: Isolation and characterization. In Meth. Enzymol. 82(A), L. W. Cunningham and D. W. Frederiksen. Eds. Academic Press, San Diego (1982).1

disaccharide units consist of amino sugars (usually D-galactosamine) alternating with uronic acids. In one type, keratan sulfate, the uronic acid is replaced by galactose. The amino sugars are usually present as N-acetyl derivatives. In heparin and heparan sulfate, however, most amino sugars are found as N-sulfates in sulfamide linkage, with a small number of glucosamine residues as N-acetyl derivatives. With the exception of hyaluronate, all glycosaminoglycans contain sulfate groups in ester linkages with the hydroxyl groups of the amino sugar residues.

The presence of carboxylate and sulfate groups provides the glycosaminoglycans with a high negative charge density and makes them extremely hydrophilic. Almost all water in the intercellular matrix is bound to glycosaminoglycans, which assume a random conformation in solution, occupying as much solvent space as is available by entrapping the surrounding solvent molecules. In solution, glycosaminoglycans also undergo aggregation and impart a high viscosity. In order to maintain electrical neutrality, negatively charged (anionic) groups of glycosaminoglycans, fixed to the matrix of the connective tissue, are neutralized by positively charged (cationic) groups (e.g., Na+). Osmotic pressure within the matrix is thereby elevated, contributing turgor to the tissue. Thus, proteoglycans, because they contain glycosaminoglycans, are polyanionic and usually occupy large hydrodynamic volumes relative to glycoproteins or globular proteins of equivalent molecular mass. Usually, they have high buoyant densities, and this property has been utilized in their isolation and characterization. The physical properties of connective tissue also depend on collagen or elastin fibers. Collagen fibers resist stretching and provide shape and tensile strength; the hydrated network of proteoglycans resists compression and provides both swelling and pressure to maintain volume (Figure 11-8). Thus, the properties of these different macromolecules are complementary, and their relative compositions vary according to tissue function. For example, sulfated glycosaminoglycans tend to provide a solid consistency, whereas hyaluronate provides a softer, more fluid consistency. The vitreous space behind the lens of the eye is filled with a viscous, gelatinous solution of hyaluronate free of fibrous proteins and transparent to light. Hyaluronate is not sulfated, and there is no evidence that it is linked to a protein molecule, as are the other glycosaminoglycans. Hyaluronate is also present in synovial fluid in joint cavities that unite long bones, bursae, and tendon sheaths. The viscous and elastic properties of hyaluronate contribute to the functioning of synovial fluid as a lubricant and shock absorber. Hyaluronate is depolymerized by hyaluronidase, which hydrolyzes the fl(1--+ 4) glycosidic linkages between N-acetylglucosamine and glucuronate. Some pathogenic bacteria secrete hyaluronidase, which breaks down the protective barrier of hyaluronate and renders the tissue more susceptible to infection. Hyaluronidase, found in 9 spermatozoa, may facilitate fertilization by hydrolyzing the outer mucopolysaccharide layer of the ovum, thereby enabling the sperm to penetrate it. Hyaluronidase (prepared from mammalian testes) is used therapeutically to enhance dispersion of drugs administrated in various parts of the body.

18~

CHAPTER11 Heteropolysaccharides I1 Proteoglycans and Peptidoglycans

HA

Collagen (type II)

Proteoglycan Collagen (type II)

F I G U R E 11-8 Schematic representation of the molecular organization of structural elements in cartilage matrix. Collagen fibrils provide tensile forces, and proteoglycans, because of their large solvent domains, accommodate reversible compressible forces. LP -- link protein; HA = hyaluronate; KS -- keratan sulfate; CS -- chondroitin sulfate; PC = core protein. [Reproduced with permission from L. C. Junqueira, J. Carneiro, and J. A. Long, Basic Histology, 5th ed. Appleton & Lange, Norwalk, CT, 1986. @ 1986 Appleton-Century-Crofts.]

Heparin is a heterogeneous glycosaminoglycan found in tissues that contain mast cells (e.g., lungs and perivascular connective tissue). Its primary physiological function is unknown, but it is a powerful inhibitor of blood clotting and is used therapeutically for that purpose. The therapeutic anticoagulant action of heparin is due to its ability to produce conformational changes in the proteinase inhibitor antithrombin. Both activated factor X (factor Xa) and thrombin are inactivated by a heparinantithrombin complex (Chapter 36). The antithrombin is homologous in structure with the ot~-antitrypsin family of proteinase inhibitors and is a suicide substrate for factor Xa and thrombin. The proteinase binds to a specific Arg-Ser peptide bond present at the reactive site of antithrombin. This 1:1 antithrombin-thrombin is a stable inactive complex. The function of heparin is catalytic. After the formation of the heparin-mediated antithrombinthrombin complex, heparin is released to initiate another cycle. Factor Xa is inhibited by a specific heparin pentasaccharide bound to antithrombin. However, the inhibition of thrombin requires an antithrombin bound to the heparin consisting of the specific pentasaccharide that consists of chains of at least 18 monosaccharide units.

Heparin therapy has a serious risk. In about 3% of patients on heparin therapy, thrombocytopenia (low platelet levels) develops. This can be life threatening if it leads to thromboembolic complications such as disseminated intravascular coagulation. Thrombocytopenia is caused by a heparin-induced immune-mediated process. The antibody is directed against a cryptic epitope present on a platelet factor (PF4) that appears only after the protein binds to heparin. The multimolecular complex consisting of antibody-heparin-PF4 causes platelet activation by binding to specific receptors (FcTRIIA) on the platelet membrane. A genetic predisposition to this disorder is due to a specific mutation in the FcTRIIA gene changes arginine to histidine at position 131. This amino acid change causes a strong affinity for the Fc region of immunoglobulin IgGl and IgG2. Thus, identification of patients with homozygosity for the Arg 131 --+ His 131 mutation in FcTRIIA and who are on heparin therapy has important therapeutic implications. Heparin-induced thrombocytopenia occurs less frequently with the use of low-molecular-weight heparin (M.W. 5,000) compared to unfractionated heparin (M.W. 3,000-30,000). The treatment of life threatening complications of heparin-induced thrombocytopenia requires cessation of heparin therapy. Heparin's anticoagulant action can be reversed rapidly by intravenous administration of prolamine sulfate. Protamines are basic proteins, isolated from fish sperm and bind tightly to heparin. One of the laboratory tests used in the identification of patients with heparin-induced thrombocytopenia, employs a micro-ELISA plate coated with purified PF4-heparin complex. Upon incubation of patient's plasma, if antibody is present, it binds to the PF4-heparin complex. The bound antibody is detected with goat antihuman antibody coupled to peroxidase, followed by incubation with substrates o-phenylenediamine/H202 (Chapter 8). Tendons have a high content of collagen and sulfated glycosaminoglycans (chondroitin and dermatan sulfates). Tendons are fibrous cords that fuse with skeletal muscle at each end and penetrate bones at the two sides of a joint. Thus, tendons are aligned along their long axis, providing flexible strength in the direction of the muscle pull. Remarkable progress has been achieved in understanding the organization of the macromolecular components of cartilage. Cartilage is one type of dense connective tissue; bone is the other (Chapter 37). Cartilage is less resistant to pressure than is bone, and its weightbearing capacity is exceeded by that of bone. It has a smooth, resilient surface. During growth and development of an embryo, cartilage provides the temporary framework for the formation and development of bone. In postnatal life, it permits the long bones of the extremities to grow until

SECTION11.1 ProteinFibers and Proteoglycans

~

........

~e

4-----------'--- Hyaluronicacid

k '~-~~-~ ~ & ! ' ---

Link protein Keratansulfate Chondroitinsulfate

~ "lw'Ll'~V'~l'~t'~'J;l"l~ Ig ,,l,l,l,lA,~,t,hl

Core protein

,11,11,1i, LI, 11,11,,a_.~ , " - ' - " ' I' I' I ' 1' I ' I ' I ' t ....... e tjl,l,l,l.l,li 1 I I l,l,~-.~el -'-'''~'I'l']'1'1'1 t.I,I,I,L

~,t',l"t"t't" ~

|

t

,,ljI,I,I,I,I,L,I,IA

7~T'T'l'l'l'r';'l'[ '' -|~ ~,lJl,ll~,l,l,L,J,l J,l I I I I I 1 1 ol .... ltl'l't'l'l'l'l'l

Subunits

~'-T*T' l ' T ' l ' l ' r ' l ' 1 " - P L , , I , I , J , I , I , I , I t l l,l,l,l,l, l,l,l,, L_el-~"l'l'l' t,[,[qq , r['~'tl'/'/',r',r'r" _IL],I,I,I,I,I,I,I,~,j ~ 1 - , , , 7 , i,l,I, r,i,l,i,FrT ' o -~'~'-~" ' , ,l,i,l,j,j,]

F I G U R E 11-9 Schematic representation of cartilage proteoglycan. The monomers, consisting of glycosaminoglycan chains linked to a core protein by covalent linkage, extend laterally at intervals from opposite sides of a very long filament of hyaluronate. The interaction between the core protein and hyaluronate is noncovalent and is aided by the link protein. The entire structure is highly hydrated and occupies a large volume. [Reproduced with permission from W. J. Lennarz, The Biochemistry of Glycoproteins and Proteoglycans, Plenum Press, New York, 1980].

their longitudinal growth ceases. Its smooth surface provides for freely movable joints (e.g., knee and elbow). The properties of cartilage depend on its content of collagen (or collagen and elastin) and proteoglycans. Cells responsible for the synthesis and maintenance of cartilage are known as chondrocytes. The proteoglycan monomer of cartilage consists of glycosaminoglycan chains of chondroitin sulfate and keratan sulfate covalently linked to a core protein (Figure 11-9) about 300 nm long with molecular weight of 250,000. Each molecule of core protein contains about 80 chondroitin sulfate and 100 keratan sulfate chains. The linkage between the oligosaccharides and the protein can be O-glycosidic between xylose and serine residues, O-glycosidic between N-acetylgalactosamine and serine or threonine residues, or N-glycosidic between N-acetylglucosamine and the amide nitrogen of asparagine. The glycosaminoglycan chains with their negatively charged groups extend out of the core protein to occupy a large volume and to entrap surrounding solvent. One end of the core protein has relatively few or no attached glycosaminoglycan chains and resembles a

185 glycoprotein; at this end, the core protein possesses an active site that binds to a very long filament of hyaluronate by a noncovalent interaction at intervals of 30 nm. The interaction between the core protein and the hyaluronate is aided by a protein (M.W. 50,000) known as link protein. The macromolecular complex is negatively charged and highly hydrated; it interacts electrostatically with basic charges on collagen fibrils, which define tissue space and provide tensile strength. The interspersed proteoglycan aggregates provide a hydrated, viscous gel for resistance to compressive loads. Proteoglycans originating in different tissues and exhibiting differences in molecular structure are presumed to be products of different genes. Synthesis and secretion of proteoglycans consist of several different phases of the internal membrane systems of the cell. Synthesis of the protein component occurs on ribosomes attached to the rough endoplasmic reticulum, with translocation of the newly synthesized polypeptide chains to the cisternal side of the membrane. The polypeptide chains travel through the cisternae of the endoplasmic reticulum to the Golgi apparatus, where the oligosaccharide side chains are synthesized by the addition of one sugar residue at a time. Further modification of the sugar residues (the conversion of selected residues of D-glucuronate to Liduronate, and sulfation) takes place after completion of the oligosaccharide (Chapter 16). The fully synthesized proteoglycans are then transferred from the Golgi apparatus, in the form of vesicles, to the cell membrane for the purpose of secretion into the extracellular spaces. The proteoglycan content in animal tissues appears to change with aging. Tissues rich in keratan sulfate show a continual increase in amount of this proteoglycan throughout life. The amounts of chondroitin sulfate in cartilage and in nucleus pulposus (of intervertebral disk) and of hyaluronic acid in skin decrease with age. Administration of growth hormone to an older animal appears to reverse the pattern of proteoglycan synthesis, making it similar to that of a younger animal. Some of the effects of growth hormone are mediated indirectly by a group of peptides known as somatomedins, which are secreted by the liver (and possibly by the kidney) under the influence of growth hormone (Chapter 31). Somatomedin A (formerly known as sulfation factor) exhibits cartilagestimulating activity, in part by affecting the incorporation of sulfate into proteoglycans. Testosterone, a steroid hormone (Chapter 34), increases the rate of hyaluronate synthesis. Insulin deficiency causes diminished synthesis of proteoglycans in rats that are rendered diabetic experimentally; synthesis is restored to normal following insulin administration. Some chronic complications of diabetes mellitus, namely, accelerated vascular degeneration, poor wound healing, and increased susceptibility to

186 infection, may be partly attributable to altered proteoglycan metabolism.

Turnover of Proteoglycans and Role of Lysosomes Proteoglycans undergo continuous turnover at rates dependent upon the nature of the proteoglycan and tissue location. Their half-life is between one and several days. Degradation of proteoglycans is initiated by proteolytic enzymes that release glycosaminoglycans; the latter are subsequently degraded by lysosomal enzymes. Some of the products of hydrolysis (e.g., dermatan sulfate and heparan sulfate) are excreted in the urine. Lysosomes are subcellular organelles in which a wide range of catabolic enzymes are stored in a closed, protective membrane system. They are major sites of intracellular digestion of complex macromolecules derived from both intracellular (autophagic) and extracellular (heterophagic) sources. Lysosomal enzymes show optimal activity at acidic pH. The pattern of enzymes in lysosomes may depend upon the tissue of origin, as well as upon the physiological or developmental state of the cells. Lysosomal enzymes are synthesized on the ribosomes of the rough endoplasmic reticulum, passed through the Golgi apparatus, and packaged into vesicles. The hydrolyases are glycoproteins, some of which contain mannose 6-phosphate markers necessary for the normal uptake of glycoproteins into lysosomes. Thus, carbohydrates may also serve as determinants of recognition in the intracellular localization of the glycoproteins following their synthesis. In two biochemically related disorders of lysosomal function, I-cell disease (mucolipidosis II) and pseudoHurler's polydystrophy (mucolipidosis III), the lesion is in the posttranslational modification step for acid hydrolases destined to be packaged into lysosomes. In normal cells, these acid hydrolyases are glycoproteins carrying mannose 6-phosphate markers that direct them to lysosomes through a receptor-mediated process. In other words, the presence of phosphomannose residues on the newly synthesized acid hydrolases and of phosphomannose receptors on selected membranes leads to the segregation of these enzymes in the Golgi apparatus, with subsequent translocation into lysosomes. In mucolipidosis II, the acid hydrolases are not phosphorylated; in mucolipidosis III, the enzymes either are not phosphorylated or have significantly diminished phosphate content. The lack of mannose 6-phosphate leads to defective localization of acid hydrolases that, instead of being packaged in lysosomes, are exported outside the cell; thus, the enzyme activity in plasma reaches high levels. These enzymes could cause indiscriminate damage. At least eight acid hydrolases (glycosidases, sulfatases, and cathepsins) appear to be affected

CHAPTER11 Heteropolysaccharides I1: Proteoglycans and Peptidoglycans

in this manner. However, not all lysosomal enzymes and tissue cells are affected. For example, lysosomal acid phosphatase and/~-glucosidase, and hepatocytes and neurons, appear to be spared from this defect. In mucolipidosis II and III, large inclusions of undigested glycosaminoglycans and glycolipids occupy almost all of the cytoplasmic space in cultured skin fibroblasts. In addition to the phosphorylation defect, the acid hydrolases are much larger than their normal counterparts, presumably owing to lack of the limited proteolysis of the hydrolases that occurs in normal lysosomes. Both disorders are inherited as autosomal recessive traits, affect primarily connective tissue, and are characterized by psychomotor retardation, skeletal deformities, and early death. The segregation of lysosomal enzymes into lysosomes requires carbohydrate recognition markers (phosphomannose in some) and also the formation of coated vesicles into which the enzymes are sequestered. Coated vesicles shuttle macromolecules between organelles and may be responsible for selectivity in intercompartmental transport (e.g., from endoplasmic reticulum to Golgi, from Golgi to lysosomes). The major component of coated vesicles is clathrin, a nonglycosylated protein (M.W. 180,000). It is located on the outer surface (cytoplasmic side) of the coated vesicles. Clathrin and its tightly bound light chains (M.W. 33,000 and 36,000) form flexible lattices that function as structural scaffolds surrounding the vesicles. In addition to their role in intracellular transport, coated vesicles are involved in receptor-mediated endocytosis. This process accomplishes internalization of macromolecules (ligands) by binding them to receptors on the cell membranes located in specialized regions of clathrincontaining coated pits, which invaginate into the cell to form coated vesicles. Inside the cell, the vesicles lose their clathrin coat (which may be reutilized) and fuse with one another to form endosomes, whose contents are acidified by proton pumps driven by free energy of hydrolysis derived from ATE In the endosome, the ligand and receptor undergo dissociation. The endosome fuses with the primary lysosome to form a secondary lysosome. Receptors may also be recycled or degraded. Thus, there are several pathways of receptor-mediated endocytosis. In some cells, the receptors migrate continuously to coated pits and undergo internalization whether or not ligands are bound to them (e.g., receptors for low-density lipoproteins, transferrin, and asialoglycoproteins). In other cells, the receptors are diffusely distributed and do not migrate to coated pits unless they are bound with ligands (e.g., epidermal growth factor). Following internalization, the receptor-ligand complex may be disposed of by one of four routes:

SECTION11.1 Protein Fibers and Proteoglycans

1. Ligand degradation and receptor reutilization (e.g., low-density lipoproteins, Chapter 20; asialoglycoproteins, Chapter 10; transcobalamin II, Chapter 38; some peptide hormones; 2. Both ligand and receptor reutilization (e.g., transferrin, Chapter 29; class I major histocompatibility complex molecules on T cells and class II molecules on macrophages, Chapter 35); 3. Both receptor and ligand degradation (e.g., epidermal growth factor, immune complexes); 4. Both ligand and receptor transportation out of the cell (e.g., maternal IgG, secretory IgA Chapter 35). Receptors for different ligands have been characterized and show multiple functional domains (see low-density lipoproteins, Chapter 20; insulin, Chapter 22). Phagocytosis is a transport system that does not involve receptor-mediated endocytosis coupled to the formation of coated vesicles. This type of internalization consists of forming phagosomes, which are membrane-bound vesicles containing ingested substances (e.g., bacteria or their products, extracellular material). Secondary lysosomes are formed when primary lysosomes fuse with phagosomes; they can also be formed when primary lysosomes fuse with autophagosomes, which contain internally derived defective organelles (e.g., mitochondria or rough endoplasmic reticulum). Within the secondary lysosomes, the ingested macromolecules are broken down by a wide variety of hydrolases into their respective monomeric units (e.g., sugars, amino acids, purines, pyrimidines, fatty acids, and cholesterol). Eventually these molecules appear in the cytoplasm and are usable for metabolic purposes. Undigested material is retained within the lysosomes and forms a residual body. A schematic representation of the formation and some functions of lysosomes is shown in Figure 11-10. In some circumstances, lysosomal enzymes are extruded into the extracellular milieu and cause destruction of the surrounding matrix. This extracellular catabolic process releases materials that are taken up in phagosomes for further breakdown by the cells. This process occurs in the remodeling of bone and cartilage. In the thyroid gland, lysosomes participate in the regulation of hormone release from the cells to the blood circulation. The thyroid hormones, thyroxine and triiodothyronine, are synthesized on a polypeptide, thyroglobulin, and stored within the thyroid follicles. Under appropriate stimuli, the thyroglobulin is internalized (endocytosed) and hydrolyzed by lysosomal hydrolases to release the free hormones, which are then transferred to the blood (Chapter 33). In vitro, estradiol, testosterone, and vitamins A and D have disruptive

187

effects on the lysosomal membrane, causing the release of lysosomal enzymes. The anti-inflammatory steroid hormone cortisol has an opposite effect. The destructive potential of leukocytic lysosomal proteinases is checked by O~l-proteinase inhibitor and otz-macroglobulin in plasma and synovial fluids. Enhanced catabolism of proteoglycans of articular cartilage occurs in rheumatoid arthritis as a result of the action of lysosomal enzymes on articular cartilage. Many pathological conditions occur as a result of a deficiency of lysosomal enzymes (e.g., storage disorders of glycoproteins and glycosphingolipids). At least 41 distinct lysosomal storage diseases are known. Most of these diseases are due to deficiency of a particular enzyme and a few are due to defects in the nonlysosomal proteins that are necessary for lysosomal biogenesis. Lysosomal storage disorders are predominantly autosomal recessive, with the exception of Fabry's disease and mucopolysaccharidosis type II, which are Xlinked recessive disorders. Overall, lysosomal storage disorders are relatively common with a prevalence of 1 per 7700 live births, although some disorders are very rare.

Mucopolysaccharidoses Mucopolysaccharidosis (MPS) encompasses disorders in which undegraded or partly degraded glycosaminoglycans accumulate in the lysosomes of many tissues owing to a deficiency of specific lysosomal enzymes. Table 11-3 lists the missing enzymes and gives relevant clinical and laboratory findings. These disorders, with the exception of Hunter's syndrome, which is an X-linked trait, are inherited in the autosomal recessive manner. All of the deficient enzymes are acid hydrolases except for acetyltransferase in Sanfilippo's syndrome type C. In mucopolysaccharidoses the catabolism of heparan sulfate, dermatan sulfate, and keratan sulfate is affected. Their degradation proceeds from the nonreducing end of the carbohydrate chain by the sequential actions of lysosomal exoglycosidases, exosulfatases, and an acetyltransferase (Figures 11-11 through 11-13). These disorders are rare; collectively, they may occur in 1 in 20,000 live births. Since proteoglycans are widely distributed in human tissues, the syndromes can affect a wide variety of tissues; thus, the clinical features vary considerably. All types are characterized by reduced life expectancy, with the exception of Scheie's syndrome. All types are characterized by skeletal abnormalities, which are particularly severe in types IV and VI. Types IH, IS, IV, VI, and VII usually exhibit clouding of the

188

CHAPTER11 Heteropolysaccharides I1: Proteoglycans and Peptidoglycans

I ! Heterophagosome

~

Secondary lysosome

lysosome

....'i:~. , . . ' . : .. Iv----:"~ ... ""~ ~' ~;-:--:--3"." :: ~ t ~.~.=-~;--:.... ~1 5 ..ii,t;D,::

1

,

i

.

~

~

Autophagosome

Nucleus'""i:"~"":; " :."..

9

, ,, ~< :".:":. " ":::' " " . ;.::~')~:;,bi ~,'i~:~ ..,............~~~!,~:~!~ 9 .. '

i

~,:.,_.

:

1[: : N U itq.l~';.:~,;.~;~-a~ imr~l,.:~...: .~..:.,,c., ~

~-.:~t~:-;f.'~ :".

9

.~ cD C~ .~..~

9

9 9

> C~

~u Z

c~

189

1~0

CHAPTER11 Heteropolysaccharides I1" Proteoglycans and Peptidoglycans H2COH

COOH

O iduronate_ ~ sulfatase

O

H2CO(~) O

O~ etc

o O o etc

. co ~

COOo

O g

.2co |

o O o_etc COOo

. co |

heparan - - ~ 1 N-sulfatase ~3 H2COH

COOH

Acetyl-CoA ~ ~ -

O~ " ~ b NH2

H2COH

14

H2CO~)

O(g)

COOH

NAc

H2COH

COOH

H2COO

COOH

H2COH

o

o

etc

NAc 13 COOH

O-~,etc

H2COH O

NAc

O ~ etc NAc

/4

13-hexosaminidase A,B ,

H2CO(~)

~

COOoH

o

O~ etc o~-N-acetyl J glucosaminidase

~-glucuronidase COOH

o_

NAc

H2COH

O~

H2COH

o

12

N-acetylgalactosamine 4-sulfatase

NAc

COOH

o~

~-L-iduronidase

~

H2COH

o 11 NAc iduronate_ ~ sulfatase O (~) H2COH

NAc

~-L-iduronidase

acetyltransferase

~

o |

H2COH O~ ~ - O

~,et c NAc

15

H2CO(~) H2COH

glucuronate /~(~) I sulfatase - ' I ~6 COOoH

NAc

H2CO(~)

~ O

NAcO,wwetc

~-glucuronidase 17 CO | N-acetyl glucosamine 6-sulfatase

NAc

O,,,,~,etc

~

O

~etc NAc

F I G U R E 11-12 Stepwise degradation of dermatan sulfate. The deficiency diseases corresponding to the numbered reactions are: l = MPS II, Hunter's syndrome; 2 = MPS I, Hurler's, Scheie's, and Hurler-Scheie's syndromes; 3 = MPS VI, Maroteaux-Lamy syndrome; 4 = Sandhoff's disease; and 5 = MPS VII, Siy's syndrome. This schematic drawing depicts all structures known to occur within dermatan sulfate and does not imply that they occur in equal proportion. For instance, only a few of the L-iduronic acid residues are sulfated, and L-iduronic acid occurs much more frequently than glucuronic acid. [Reproduced with permission from E. F. Neufeld and J. Muenzer. In: Metabolic Basis of Inherited Disease, 7th ed., C. R. Scriver, A. L. Beaudet, W. S. Sly, and D. Valle (Eds). McGraw-Hill, New York, 1995, p. 2467.]

H ~ " ~ O~,wetc NAc FIGURE 11-11 Stepwise degradation of heparan sulfate. The deficiency diseases corresponding to the numbered reactions are: 1 = mucopolysaccharidosis (MPS) II, Hunter's syndrome; 2 = MPS I, Hurler's, Scheie's, and Hurler-Scheie's syndromes; 3 = MPS III A, Sanfilippo's syndrome type A; 4 = MPS III C, Sanfilippo's syndrome type C; 5 = MPS III B, Sanfilippo's syndrome type B; 6 = no deficiency disease yet known; 7 = MPS VII, Sly's syndrome; 8 = MPS III D, Sanfilippo's syndrome type D. The schematic drawing depicts all structures known to occur within heparan sulfate and does not imply that they occur stoichiometrically. Very few of the glucuronic acid residues are sulfated. [Reproduced with permission from E. F. Neufeld and J. Muenzer. In: Metabolic Basis of Inherited Disease, 7th ed., C. R. Scriver, A. L. Beaudet, W. S. Sly, and D. Valle (Eds). McGraw-Hill, New York, 1995, p. 2468.]

hypotonic environment. The contents of a bacterium can exert an osmotic pressure as high as 20 atm. At cell division, the walls rupture and reseal rapidly. Bacteria are classified into two groups on the basis of a staining reaction discovered by Gram in 1884. In this reaction, the cells do or do not retain a crystal violetiodine dye complex after an alcohol wash. Cells that retain the stain are gram-positive; those that do not are gramnegative. This empirical classification divides bacteria into two classes that differ in cell wall structure. Gram-positive bacteria (e.g., Staphylococcus aureus) are surrounded by a cytoplasmic membrane with a bilayer

SECTION 11.2

Peptidoglycans

H2CO~

191

H2CO(~)

H2COH

H2CO Q

,actose

6-sulfatase ' " ~

O ~,~ etc NAc

NAc

11 H2COH

H2CO I~)

/ o

H2COH

~

H2CO (~)

o

o _ etc

NAc ~-galactosidase

NAc

12 H2CQ.O~(~) H2COH

~

N-acetyl / ' ~ " ~ O ~ " O glucosamine "~~ 6-sulfatase NAc

.2c0.~

H2CO (~) O

O ~ etc

NAc

.2c0.

~NOAc~O ~

.2c000 O

O ,,~ etc NAc

! ~-HexosaminidaseA, B ~4 u

H2COH

H2CO I~)

oZ.

o_

tc NAc

ALTERNATEPATHWAY HzCO~) O

Q

H2COH

HzCO (~)

~

13-HexosaminidaseA H2COH

15 H2CO (~) NAc

F I G U R E 11-13 Stepwise degradation of keratan sulfate. The deficiency diseases corresponding to the numbered reactions are: 1 = MPS IV A, Morquio syndrome type A; 2 -- MPS IVB, Morquio syndrome type B; 3 = MPS III D, Sanfilippo's syndrome type D; 4 = Sandhoff's disease; and 5 = Tay-Sachs and Sandhoff's disease. The alternate pathway releases intact N-acetylglucosamine-6-sulfate, a departure from the usual stepwise cleavage of sulfate and sugar residues. [Reproduced with permission from E.F. Neufeld and J. Muenzer: In Metabolic Basis of Inherited Disease, 7th ed., C. R. Scriver, A. L. Beaudet, W. S. Sly, and D. Valle (Eds). McGraw-Hill, New York, 1995, p. 2468.]

structure similar to that of eukaryotes and a thick, bagshaped cell wall. The cell wall, about 25 nm wide, consists of peptidoglycans (Figure 11-14) and polyol phosphate polymers known as teichoic acids. Gram-negative bacteria (e.g., Escherichia coli and Salmonella typhimurium) do not contain teichoic acids but have an outer membrane system external to the plasma membrane and the peptidoglycan. The constituents of this second outer

membrane system are phospholipids, lipopolysaccharides, and proteins. The peptidoglycan layer is connected to the outer layer via lipoprotein. The region between the inner membrane and the peptidoglycan layer is known as the periplasmic space. This space contains enzymes and other proteins that digest impermeable nutrients and transport low-molecular-weight nutrients, such as amino acids, sugars, and specific ions. Lipopolysaccharides are complex amphipathic molecules located on the outer leaflet of the outer membrane. Their structure in Enterobacteriaceae comprises three regions: a phospholipid (A) (the hydrophobic portion of the molecule), a phosphorylated core oligosaccharide, and a polysaccharide chain known as the 0 side chain or 0 antigen. The core oligosaccharide and the 0 side chain regions contain several unusual sugars and form the hydrophilic portion of the molecule that projects outward. Lipopolysaccharides serve as a barrier against invading organisms, are highly toxic and are also known as endotoxins. The fever that occurs in gram-negative bacterial infections is caused by endotoxins. The outer membrane also contains a protein known as porin. The trimeric forms of porin form channels for the passage of small ionic molecules through the outer membrane. Other transport systems exist for higher molecular weight ionic substances. Both gram-negative and gram-positive bacteria contain peptidoglycan as the main structural component of their cell walls. However, gram-negative bacteria have only a single layer of peptidoglycan, whereas gram-positive cells have several layers with cross-linkages between them. The polymeric structure of peptidoglycan surrounds the entire inner membrane and the cytoplasm of the cell. Consisting of a glycan composed of amino sugars in one dimension, a cross-linked peptide moiety in the second dimension, and an interpeptide bridge in the third dimension, peptidoglycan projects in different planes (Figure 11-14). This three dimensional, covalently bound network offers considerable resistance to outward pressure and confers mechanical stability on the cell wall. The glycan is called murein (from the Latin word for "wall") and is a polymer of alternating units of Nacetylglucosamine (GlcNAc) and N-acetylmuramic acid (MurNAc). The sugar residues are linked by fl(1 --+ 4) glycosidic bonds (Figure 11-15). N-Acetylmuramic acid is the 3-O-D-lactic acid ether of N-acetylglucosamine, the carboxyl group of its lactic acid side chain being condensed with the cross-linking peptide. This peptide usually consists of four amino acid residues that are alternately L- or D-amino acids. The bacterial cell wall is one of the few structures containing a significant amount of D-amino acid residues. In S. aureus,

192

CHAPTER 11

Heteropolysaccharides I1' Proteoglycans and Peptidoglycans #

/

/

/

(Gly)5 ~~

~" ~ D-Glu'NH2 GlcMAc~ 1 L-Lys I D-Ala...~

/ -.

1

/ /

I

D-.Glu-NH2

,. ys I

GlcMAc

-""'L-Lys I D-A~(GIy),/M~~

GlcMAc

MurMAc /L-AIa

(G Y ) 5 - /

MurMAc " iGly)5, / L - ~ l a f GlcMAc

/

-Ala

/

/

D-AIa I D-Ala

MAc

/ / /

/

/

,-Lys

GIcMAc

D-Ala

/

(GIy)5

F I G U R E 11-14 Structure of peptidoglycan in the cell wall of the gram-positive bacterium Staphylococcus aureus. The structure consists of an alternating polymer of N-acetylglucosamine (GIcNAc) and N-acetylmuramic acid (MurNAc) linked by/~(1 --+ 4) glycosidic bonds in one dimension, a cross-linking peptide that contains alternating L- and D-amino acid residues in the second dimension, and a pentaglycine moiety that cross-links the peptide chains in the final dimension. This threedimensional network is continuous and surrounds the entire ceil. [Reproduced with permission from M. J. Osborn, Structure and biosynthesis of the bacterial cell wall. Annu. Rev. Biochem. 38, 301 (1969). (~)1969 by Annual Reviews Inc.]

the tetrapeptide sequence is L-alanyl-D-g-isoglutaminylL-lysyl-D-alanine. The interpeptide chain is a pentaglycine chain that connects the terminal D-alanyl residue of one tetrapeptide to an L-lysyl residue in the third amino

\

o

.

II

I

-- C-- D'Alam L'L,ysm D-isoGIn--L-Ala--N

I

\_

/

GIY

Z/~CD

Gt,,

'

I

N--H I

CH

C

O~ ~CH 3

,CH3 Ox ~1-'~4)linkage ~CH

/I

/

OH

.c/H.

ZK

O'--C. ~n \CH3 >

F I G U R E 11-15 Repeating unit of the peptidoglycan of Staphylococcus aureus.

acid position in another tetrapeptide (Figure l l-14). The chemical linkages are as follows: the amino-terminal group of pentaglycine is joined with the carboxyl group of D-alanine, and the carboxyl terminal of pentaglycine is linked with the e-NH2 of L-lysine. Both of the sequence variations of the tetrapeptide and the cross-linking pattern are characteristic of the shape and species of the bacterium. In some bacteria, in addition to the cell wall, substances form capsules or slime layers external to the cell wall. These layers are not essential for growth and multiplication but may be important for survival of the organism in harsh environments, e.g., in preventing desiccation and serving as a barrier against phage (bacterial virus) attack. The capsular layer also provides a charged surface. Streptococcus mutans, which plays a significant role in dental caries and plaque formation, produces an extracellular 1,3-glucan that enables the bacteria to adhere to teeth. In the formation of dental caries, possibly the most widespread human pathological process, dietary carbohydrates (sugar in general and sucrose in particular) promote bacterial growth and produce noxious metabolites. Penicillins prevent cell wall synthesis in susceptible organisms by inhibiting a late step in the enzymatic synthesis of peptidoglycan (Chapter 16).

193

SECTION11.2 Peptidoglycans

F I G U R E 11-16 Primary sequence of the hen egg-white lysozyme. Amino acid residues thought to be part of the active site are identified by the arrows.

Lysis of Peptidoglycans by Lysozymes Lysozyme breaks down the peptidoglycans by hydrolysis of the /3(1-+ 4) glycosidic bond between N-acetylglucosamine and N-acetylmuramic acid. Lysozyme occurs in tears, nasal and bronchial secretions, gastric secretions, milk, and tissues and may have a protective effect against air- and food-borne bacterial infections. The structure and catalytic mechanism of lysozyme have been extensively studied. Its catalytic mechanism provides an example of steric distortion induced in the substrate by the enzyme before products are formed. Hen egg-white lysozyme is abundant and easy to purify. It is a roughly ellipsoidal, compact molecule (4.5 x 3.0x 3.0 nm) of molecular weight 14,600 consisting of a single chain of 129 amino acid residues with four disulfide bridges (Figure 11-16). It is also the first enzyme whose structure was elucidated by x-ray crystallography, that contains no prosthetic group or metal ions, and that consists of regions of antiparallel /~-pleated sheet, ol-helix (small amount), and nondescriptive ("random") structure (large amount). As in hemoglobin and myoglobin (Chapter 4), its interior contains almost entirely nonpolar amino acid residues. Lysozymes and o~-lactalbumin show striking similarity in sequence and structure, which suggests a common evolutionary ancestry. However, o~-lactalbumin functions as an enzyme modifier in the

synthesis of lactose in the mammary gland after parturition (Chapter 15), so this may be an example of divergent evolution. The mechanism of lysozyme action has been elucidated by the use of synthetic substrates and inhibitors. GlcNAc-GlcNAc-GlcNAc is an inhibitor that forms a stable enzyme-inhibitor complex. The active site of the enzyme has been established by x-ray crystallography with and without inhibitor and also by model building. The enzyme contains a central crevice running horizontally across the molecule. The hexasaccharide GlcNAc-MurNAc-GlcNAc-MurNAc-GlcNAc-MurNAc A

B

C

D

E

F

binds in the cleft noncovalently, with residue C making the largest contribution. This binding distorts the normal chair conformation of sugar residue D. Since the bond between residues D and E is hydrolyzed, this binding region contains the active site of the enzyme. The rate of catalysis is not significant unless the substrate is a pentamer. The main features of a proposed mechanism for hydrolysis of the bond between D and E are shown in Figure 11-17. Ring D is distorted resulting in formation of a transition state intermediate in which the conformation lies between those of substrate and products, and which has the maximum energy. With the strained conformation (half-chair) of ring D, the susceptible bond between D and E is drawn in close proximity to Glu-35 and Asp-52 residues of the

194

CHAPTER 11

HeteropolysaccharidesI1: Proteoglycans and Peptidoglycans

profile is bell-shaped (activity rapidly falling off on either side of the optimum pH). At pH 5.0, Asp-52 (pK' ~3.8) is ionized and Glu-35 (pK' ~6.7) is un-ionized. The pK' of Glu-35 is significantly different from that of the free amino acid (4.25) because Glu-35 is in a nonpolar region of the molecule. Asp-52, on the other hand, is in a polar environment, where its ionization is facilitated. The importance of acidic amino acid residues in catalysis is further substantiated by the fact that lysozyme remains active when all carboxyl groups, except those of Glu-35 and Asp-52, are chemically modified (e.g., by esterification).

11.3 Lectins

F I G U R E 11-17 Proposed mechanism for the catalytic hydrolysis of a hexasaccharide lysosome.

enzyme. Glu-35 functions as a general acid catalyst donating a proton to the glycosidic oxygen between sugar residues D and E, which results in cleavage of the bond and release of the disaccharide E - E The remainder of the substrate (a tetrasaccharide), however, is still bound to the enzyme with D acquiring a positive charge (a carbonium ion) and continuing to be in the half-chair conformation. The negatively charged Asp-52 now stabilizes the positively charged carbonium ion intermediate. A hydroxyl ion and a proton from water then react with the carbonium ion and the enzyme to generate a tetrasaccharide and the original enzyme. In this mechanism, Asp-52 is ionized and Glu-35 is un-ionized in the enzyme, consistent with their functions. The optimum pH of the enzyme is 5.0 and the pH-rate

The lectins are a group of proteins, originally discovered in plant seeds (now known to occur more widely), that bind carbohydrates and agglutinate animal cells. They have two or more stereospecific sites that bind noncovalently with the terminal (and often penultimate) residue at the nonreducing end of an oligosaccharide chain. A number of plant lectins have been purified and their binding properties investigated. Wheat germ agglutinin binds to N-acetylglucosamine and its glycosides; concanavalin A from jack beans binds to mannose, glucose, and glycosides of mannose and glucose; peanut agglutinin binds to galactose and galactosides; and red kidney bean lectin binds to N-acetylglucosamine. Since lectins have high affinity for specific sugar residues, they have been used to identify specific carbohydrate groups and used in the purification of carbohydrate-containing compounds. Lectins may be involved in carbohydrate transport, specific cellular recognition, embryonic development, cohesion, or binding of carbohydrates. As noted in Chapter 10, hepatocytes bind to serum glycoproteins with exposed galactose residues. This binding is thought to be a lectin-mediated clearance of partially degraded glycoproteins. Hepatocytes also contain a CaZ+-dependent fucose-binding lectin. A lectin that binds to N-acetylglucosamine- and mannose-terminated glycoproteins in reticuloendothelial cells has been identified. Many lectins are glycoproteins. Some lectins cause agglutination of red blood cells and can be used in typing of blood groups. Soybean lectin, a galactose-binding protein, binds selectively to T lymphocytes and causes their agglutination. Thus, it has been used in selective removal of mature T lymphocytes from bone marrow preparations. In treatment of disorders such as immunodeficiencies, blood cancers, and some hemoglobinopathies, bone marrow grafts are made after ensuring that donor and recipient are histocompatible. If they are not, the donor cells destroy the

Supplemental Readings and References

recipient's cells, and graft-versus-host disease (GVHD) ensues. However, GVHD does not occur if the T lymphocytes are removed from the donor marrow, even without histocompatibility. A highly selective method of removal of T lymphocytes uses monoclonal antibodies (Chapter 3 5 ).

Supplemental Readings and References D. D. Bikle: Biochemical markers of bone metabolism: An overview, Clinical Biochemistry 30, 573 (1997). E H. Byers: Disorders of collagen biosynthesis and structure. In: Metabolic Basis of Inherited Disease, 7th ed., C. R. Scriver, A. L. Beaudet, W. S. Sly, and D. Valle (Eds). McGraw-Hill, New York, 1995, p. 4029. B. H. Chong and M. Eisbacher: Pathophysiology and laboratory testing of

195 heparin-induced thrombocytopenia. Seminars in Hematology 35(No. 4, Suppl. 5), 3 (1998). S. Elgavish and B. Shaanan: Lectin-carbohydrate interactions: Different folds common recognition principles. Trends in Biochemical Sciences 22, 462 (1997). E. Holtzman: Lysosomes, Plenum Press, New York, 1992. E J. Meikle, J. J. Hopwood, A. E. Clague, and W. E Carey: Prevalence of lysosomal storage disorders, Journal of the American MedicaI Association 281,249 (1999). E. E Neufeld and J. Muenzer: The mucopolysaccharidosis in: Metabolic Basis of Inherited Disease, 7th ed., C. R. Scriver, A. L. Beaudet, W. S. Sly, and D. Valle (Eds). McGraw-Hill, New York, 1995, p. 2465. E Ramirez, M. Godfrey, B. Lee, and E Tsipouras: Marfan syndrome and related disorders In: Metabolic Basis of Inherited Disease, 7th ed., C. R. Scriver, A. L. Beaudet, W. S. Sly, and D. Valle (Eds). McGraw-Hill, New York, 1995, p. 4079. J. Rosenbloom, W. R. Abrams, and R. Mecham: Extracellular matrix 4: The elastic fiber. FASEB Journal 7, 1208 (1993). J. I. Weitz: Low-molecular-weight heparins, New England Journal of Medicine 337, 688 (1997).

CHAPTER

12

Gastrointestinal Digestion and Absorption

The gastrointestinal (or alimentary) system (or tract) is responsible for digestion and absorption of nutrients and fluids. The tract extends from the mouth to the anus and in an adult human is about 10 m long. Digestion is the hydrolysis of complex food substances into simpler units such as monosaccharides, amino acids, fatty acids, and glycerol. Absorption is the transport of the products of digestion and of vitamins, minerals, and water across the intestinal epithelium to the lymphatic or blood circulatory systems. Digestion and absorption involve not only the gastrointestinal (GI) tract but also secretions from salivary glands, liver (and gallbladder), and pancreas. These processes in the GI system are interrelated. The system provides highly selective, efficient, and elaborate absorptive surfaces that contain enzymes and secretions of enzyme-containing fluids, electrolytes, and other substances required for digestion and absorption. Secretory and absorptive activities are regulated by hormonal and neural mechanisms.

12.1 Anatomy and Physiology of the GI Tract The GI tract consists of mouth and esophagus, stomach, small intestine (duodenum, jejunum, and ileum), and large intestine (colon and rectum).

Mouth and Esophagus In the mouth, food is mixed with saliva, chewed to break up large particles, and propelled into the esophagus by swallowing. Saliva is secreted by three pairs of glands (parotid, submaxillary, and sublingual) and by numerous small buccal glands and is under autonomic nervous system control. Parasympathetic nervous stimulation causes profuse secretion of saliva. Thus, atropine and other anticholinergic agents diminish salivary secretion and make the mouth dry. The presence of food in the mouth and the act of chewing stimulate secretion of saliva by reflex nervous stimulation. Salivary secretion can also be a conditioned response, so that the sight, smell, and even thought of food can elicit salivary secretion; however, in humans, this type of conditioned response is weak. The composition, pH, and volume of salivary excretion for different salivary glands vary. Usually 1-2 L of saliva are secreted per day. Saliva is a hypotonic solution with a pH of about 7.0; principal cations are Na + and K +, and anions are C1- and HCO~-. Aldosterone (a steroid hormone of the adrenal cortex) modulates Na + and K + levels in saliva as it does in the kidney (Chapter 32). Aldosterone increases reabsorption of Na + and secretion of K + by direct action on the salivary gland ducts. In hypoaldosteronism (e.g.,Addison's disease), the salivary Na+/K + ratio is high,

197

1~8 and in adrenocortical hyperfunction (e.g., aldosteronism), this ratio is low. The chief salivary organic constituent is mucin, which is a mixture of glycoproteins containing 60-85% oligosaccharide by weight. The oligosaccharide units are linked by O-glycosidic linkage to either serine or threonine residues (Chapter 9). Mucin-rich saliva is secreted by submaxillary and sublingual glands, and because of the high carbohydrate content, mucins form viscous solutions responsible for much of the lubricating action of saliva that aids swallowing. Amylase is a salivary enzyme that catalyzes the hydrolysis of or(1 --+ 4) glucosidic linkages of starch and glycogen. Its role is minimal because of the limited time spent by the food in the mouth and the inactivation of amylase in the stomach by the acid pH. Other enzymes detected in saliva include carbonic anhydrase, a lipase (lingual lipase), a phosphatase, and a protease called kallikrein. Whether these enzymes function in saliva is not known. Lingual lipase initiates hydrolysis of dietary fat in the stomach and facilitates the duodenal-jejunal hydrolysis of triacylglycerols. Even when no food is present in the mouth, saliva is secreted to keep the mouth moist and facilitates speech. Saliva also has some antibacterial action. Patients with xerostomia (dry mouth) exhibit higher than normal incidence of dental caries. Saliva maintains the oral cavity at a pH of about 7.0. The teeth do not lose calcium to oral fluids because of the high concentration of calcium in the saliva at pH 7.0. Like the thyroid gland (Chapter 33), salivary glands (and the gastric mucosa) can take up iodide ions against a concentration gradient. The mouth is the normal point of entry of food and drink. There, solid food is reduced in size by mastication, blended with saliva, and temperature-moderated before being swallowed. Conditions that interfere with any of these processes (e.g., tooth loss) can affect food choice and hence the nutritional status and health of the individual. The esophagus is a muscular tube through which masticated food is transported from the mouth to the stomach. It consists of both striated muscle (upper one third) and smooth muscle (lower two thirds). These muscles undergo periodic contractions in the form of peristaltic waves that push the swallowed boluses toward the stomach. The esophagus has no digestive function but secretes mucus to protect the esophageal mucosa from excoriation. Stomach The stomach stores food temporarily, retards its entry into the small intestine, and secretes pepsin to begin the digestion of protein. Hydrogen ions in the stomach activate pepsinogen to form pepsin and aid in maintaining

CHAPTER12 Gastrointestinal Digestion and Absorption

the sterility of the upper GI tract. The stomach also secretes intrinsic factor (a glycoprotein), which is required for vitamin B12 absorption in the ileum (Chapter 38), helps prepare some of the essential minerals (e.g., iron) for absorption by the small intestine (Chapter 29), and provides mucus for protective and lubricative functions. The stomach wall is made up of four concentric layers: the mucosa, the submucosa, the muscularis external, and the serosa. Functionally, the gastric mucosa is divided into the oxyntic gland area (which includes the body and fundus of the stomach) and the pyloric gland area. The oxyntic gland area contains tightly packed parallel glands and is covered with a layer of tall, columnar surface epithelial cells that secrete mucous and line the gastric pits. Undifferentiated cells in the region joining the gastric gland to the pit (the neck region) undergo division with some daughter cells migrating upward and differentiating into surface epithelial cells, with other daughter cells migrating downward and differentiating into neck mucus cells, and with chief and parietal cells. Thus, the mucosa renews itself rapidly, approximately every 3-5 days in humans. Replacement of the mucosa after injury occurs even more rapidly. Pepsinogen is synthesized and secreted by chief cells and HC1 by parietal cells. A proteolytic enzyme secreted by gastric mucosa of infants is chymosin (rennin), which functions to clot milk and promote its digestion by preventing rapid passage from the stomach. Chymosin hydrolyzes casein, a mixture of several related milk proteins, to paracasein, which reacts with Ca 2+ to yield the insoluble curd. Pepsin performs the same functions as chymosin. Chymosin is found in the fourth stomach of ruminants. Calf stomach is a source of this enzyme, which is used in the manufacture of cheese. Chymosin has been synthesized by recombinant DNA techniques and successfully used in the production of cheese. The pyloric mucosa contains surface epithelial cells that secrete mucus, which increases during digestion of food. Inadequate secretion of mucus can lead to stomach ulcers. The cells of the pyloric glands secrete a fluid similar in composition to an ultrafiltrate of plasma. These cells also secrete a pepsinogen that differs structurally from the pepsinogen secreted by the oxyntic gland mucosa. Secretion from all gastric glands amounts to 2-2.3 L/day and is regulated by neural and humoral systems. Neural control is mediated by the vagus nerves. The sight, smell, and thought of pleasant-tasting food can elicit gastric secretion during the cephalic phase. Neural control is also exerted by local autonomic reflexes mediated by cholinergic neurons. Anger and hostility can cause gastric secretion through nerve impulses down the vagi. Secretory signals can arises from the cerebral cortex.

SECTION 12.1

Anatomy and Physiology of the 61 Tract

Food and its digestion products stimulate receptors in the stomach that increase gastric secretion during the gastric phase, mediated primarily by secretion of gastrin. Gastrin is a gastrointestinal hormone synthesized and stored in G cells of the pyloric glands and mucosa of the proximal part of the small intestine. When food moves into the small intestine, the intestinal phase of gastric secretion involves complex interactions of stimulatory and inhibitory factors. Thus, chyme (the semifluid, homogeneous material produced by gastric digestion) in the upper jejunum inhibits gastric secretion, whereas amino acids in the middle and lower jejunum stimulate gastric secretion. These activities are primarily mediated through GI hormones (see below). The three phases of gastric secretion (cephalic, gastric, and intestinal) are interdependent and proceed more or less simultaneously. The rate of passage of food into the duodenum depends on the type of food and on the osmotic pressure it exerts in the duodenum. Food rich in fat moves most slowly, food rich in carbohydrate moves most rapidly, and food rich in protein moves at an intermediate rate. Hyperosmolality decreases gastric emptying via duodenal osmoreceptors. Some GI hormones also inhibit gastric motility and secretion.

Small Intestine Most digestion and absorption of food constituents occurs in the small intestine. The small intestine is also the major site for absorption of water and electrolytes. It is a convoluted tube about 6 m long, beginning at the pylorus and ending at the ileocecal valve. It is divided into a duodenum (a C-shaped, short, fixed segment), the mobile jejunum, and ileum. Its wall consists of the same four concentric layers as in other parts of the GI tract: m u c o s a , submucosa, muscularis externa, and s e r o s a . Finger-like projections known as villi that arise from the luminal side of the mucosal membrane provide a large surface area in the small intestine. Each villus consists of a single layer of tall, columnar epithelial absorptive cells (enterocytes) that contain microvilli which further increase the absorptive surface and give it a brush-like appearance (brush border). The surface membrane of the microvillus contains digestive enzymes and transport systems. The core of the microvillus consists of a bundle of filamentous structures that contain actin and associated proteins (e.g., myosin, tropomyosin, villin, fimbrin; see Chapter 21), which may have a structural role. Enterocytes are abundantly supplied with mitochondria, endoplasmic reticulum, and other organelles. The outermost surface of the microvillus is the carbohydrate-rich glycocalyx, which is anchored to the underlying membrane. The glycocalyx

199

is rich in neutral and amino sugars and may protect cells against digestive enzymes. Villi also contain goblet cells, which secrete mucus. The absorptive cells are held together by junctional complexes that include "tight junctions." However, the tight junctions contain pores that permit the transport of water and small solutes into the intercellular spaces according to the osmotic pressure gradient. Endocrine cells are distributed throughout the small intestine and other sections of the GI tract. About 20 different cell types have been identified. Some, easily identifiable by their staining characteristics, are argentaffin or enterochromaffin cells containing 5-hydroxytryptophan, the precursor of serotonin (Chapter 17). Neoplastic transformation of these cells leads to excessive production and secretion of serotonin, which causes diarrhea, flushing, and bronchoconstriction (carcinoid syndrome). Other types of cells lack 5-hydroxytryptophan but take up amine precursors (e.g., amino acids) to synthesize and store biologically active peptides (e.g., hormones). The mucosal cells of the small intestine and stomach are among the most rapidly replaced cells of the human body, being renewed every 3-5 days. Millions of cells are sloughed off (or exfoliated) from the tips of villi every minute; about 17 billion such cells are shed per day. These mucosal cells amount to about 20-30 g of protein, most of which is reclaimed as amino acids after hydrolysis in the lumen. These cells are replaced through division of undifferentiated cells in the crypts of the villi (crypts of Lieberktihn)and subsequent migration to the top of the villus, followed by maturation. Mitotic poisons (e.g., antineoplastic agents), injury, and infection can adversely affect mucosal cell proliferation, producing a flat mucosa devoid of villi and leading to severe malnutrition. Digestion in the small intestine requires the biliary and pancreatic secretions that are emptied into the duodenum (usually at a common site).

Formation, Secretion, and Composition of Bile Bile is formed and secreted continuously by polygonally shaped liver parenchymal cells called hepatocytes. An aqueous buffer component (e.g., HCO~-) is added to the bile by the hepatic bile duct cells that carry the secretion toward the common bile duct. The membrane of the hepatocytes in contact with the blood has microvilli that facilitate the exchange of substances between plasma and the cells. Hepatocytes are rich in mitochondria and endoplasmic reticulum. Hepatic bile flows into the gallbladder, where it is concentrated, stored, and emptied into the duodenum when the partially digested contents of the stomach enter

~00

F I G U R E 12-1 Schematic representation of the radial architecture of the plates formed by the liver cells. Blood from the portal vein and the hepatic artery flows into sinusoids and eventually enters the central vein. The bile canaliculi are located between the liver cells. Bile flows in the opposite direction and empties into the bile duct in portal triads. [Reproduced with permission from A. W. Ham Histology, 8th ed., J. B. Lippincott, Philadelphia, 1979.]

the duodenum. This movement is accomplished by contraction of the gallbladder mediated by cholecystokinin, a GI hormone. The liver is the largest gland and second largest organ in the body and weighs about 1.5 kg in an adult. It functions in synthesis, storage, secretion, excretion, and specific modification of endogenously and exogenously derived substances. The liver takes up material absorbed from the small intestine, since it is mainly supplied by venous blood arriving from the GI tract. It is uniquely susceptible to changes in alimentation and to toxic substances. The liver participates in numerous metabolic functions. The liver is organized into lobules, functional aggregates of hepatocytes arranged in radiating cords or plates surrounding a central vein (Figure 12-1). In adults, the branching plates are usually one-cell thick, and the vascular channels between them are known as sinusoids. Blood in the sinusoids passes slowly between the rows of cells, facilitating the exchange of substances between cells and the plasma. The blood supply of the sinusoids is furnished by the portal vein (75%) and by the right and left hepatic arteries (25%). Blood flows from the periphery of the lobules into the central vein. Sinusoids anastomose to form central veins that join to form larger veins which feed through the hepatic vein into the inferior vena cava. Sinusoids are larger than capillaries and are lined by reticuloendothelial cells, including the phagocytic Kupffer cells. This layer contains numerous, small fenestrations that provide access of sinusoidal plasma to the surface microvilli of hepatocytes via the intervening space, called the space of Disse. Fine collagen fibers (reticulin) within this space provide the support for the liver cell plates (Figure 12-2).

CHAPTER12 Gastrointestinal Digestion and Absorption

! F I G U R E 12-2 Schematic representation of structures within the hepatic lobule. H hepatocyte, BC -- bile canaliculus, KC - Kupffer cell, EC -- endothelial cell, N = nerve fiber, F -- reticulin fibers, S = sinusoid, D - space of Disse, X -- gap between sinusoid lining cells, and RBC -- red blood cell.

Bile produced by hepatocytes is secreted into the bile canaliculi between adjacent hepatic cells. The wall of the canaliculus is formed by the plasma membrane of the hepatocytes, which are held together by tight junctions. Canaliculi arise near central veins and extend to the periphery of the lobules. The direction of bile flow in the canaliculi is centrifugal, whereas that of the blood flow is centripetal. Canaliculi coalesce to form ducts, which are lined by epithelium, and the ducts coalesce to form the right and left hepatic ducts. Outside the liver these ducts form the common hepatic duct. The volume of hepatic bile varies in a normal human adult from 250 to 1100 mL, depending on the rate at which bile acids recirculate in the enterohepatic cycle (see below). Some GI hormones increase the volume and bicarbonate content of bile through stimulation of the duct epithelium. Hepatic bile has a pH of 7.0-7.8 and is isotonic with plasma. During the interdigestive period, it is diverted into the gallbladder, since the sphincter of Oddi is closed. Bile in the gall bladder is concentrated 12- to 20-fold by absorption of electrolytes and water. Nonetheless, gallbladder bile is isotonic with plasma because cations (mainly Na +) associate with the osmotically inactive bile acid micellar aggregates. Gallbladder capacity is 50-60 mL. Emptying requires a coordinated

SECTION 12.1

201

Anatomy and Physiology of the GI Tract

contraction of the gallbladder and relaxation of the sphincter of Oddi, both mediated by cholecystokinin. Bile contains bile acids, bile pigments, cholesterol, phosphatidylcholine (lecithin), and electrolytes. Bile pigments are glucuronide conjugates of bilirubin and biliverdin derived from the degradation of heme, a prosthetic group of many proteins. Most of the bilirubin formed in the body comes from hemoglobin catabolized in reticuloendothelial cells (liver is one of the sites where these cells are formed). The bilirubin is bound to plasma albumin and transported to hepatocytes, where it is converted to glucuronide conjugates and secreted into the bile, giving it a golden yellow color. If bilirubin remains in the gallbladder, it is oxidized to biliverdin, which is greenish brown. Bile pigments are toxic metabolites (Chapter 29). The bile acids are 24-carbon steroid derivatives. The two primary bile acids, cholic acid and chenodeoxycholic acid, are synthesized in the hepatocytes from cholesterol by hydroxylation, reduction, and side chain oxidation. They are conjugated by amide linkage to glycine or taurine before they are secreted into the bile (see cholesterol metabolism, Chapter 19). The mechanism of secretion of bile acids across the canalicular membrane is poorly understood. Bile acids are present as anions at the pH of the bile, and above a certain concentration (critical micellar concentration) they form polyanionic molecular aggregates, or micelles (Chapter 11). The critical micellar concentration for each bile acid and the size of the aggregates are affected by the concentration of Na + and other electrolytes and of cholesterol and lecithin. Thus, bile consists of mixed micelles of conjugated bile acids, cholesterol, and lecithin. While the excretion of osmotically active bile acids is a primary determinant of water and solute transport across the canalicular membrane, in the canaliculi they contribute relatively little to osmotic activity because their anions aggregate to form micelles. Bile is also a vehicle for excretion of other endogenous (e.g., bilirubin and steroids) and exogenous (e.g., drugs and dyes) compounds. The dye sulfobromophthalein is removed from the bloodstream by the hepatocytes, excreted in the bile, and eliminated through the GI tract. Its rate of removal from the circulation depends on the functional ability of the hepatocytes and of the hepatic blood flow, and so it is used to assess hepatic function. Cholesterol is virtually insoluble in water but is solubilized through the formation of micelles, which also contain phosphatidylcholine and bile acid anions in specific proportions. Cholesterol can be precipitated to form stones (known as gallstones) when the critical concentration of micellar components is altered, i.e., when the concentration ratio of cholesterol to bile acids is increased. Gallstones may also form because of precipitation of

bilirubin, e.g., with increased production of bile pigments due to hemolysis, associated with infection and bile stagnation. Presumably, under these conditions the bilirubin diglucuronide is converted to free bilirubin by /3-glucuronidase. B ilirubin is insoluble and forms calcium-bilirubinate stones (called pigment stones). Gallstones occur in 10-20% of the adult population in Western countries. Cholesterol stones account for 80% (see Chapter 19). More than 90% of the bile acids are reabsorbed, mostly in the ileum. About 5-10% undergoes deconjugation and dehydroxylation catalyzed by bacterial enzymes in the colon to form deoxycholic acid and lithocholic acid. These acids are known as secondary bile acids. Deoxycholic acid is reabsorbed, but lithocholic acid, which is relatively insoluble, is poorly absorbed. The reabsorbed bile acids enter the portal blood circulation, are reconjugated in the liver, and are secreted into bile. This cycling is known as the enterohepatic circulation and occurs about six to eight times daily. The rate of cycling increases after meals. The lithocholic acid is conjugated with either taurine or glycine, sulfated at the ring hydroxyl group, and again secreted into the bile. Sulfated lithocholate is not reabsorbed. Free lithocholic acid is highly toxic to tissues and causes necrosis. The bile acid pool normally consists of about 2 - 4 g of conjugated and unconjugated primary and secondary bile acids. Daily loss of bile acids in feces, mostly as lithocholate, is about 0.2-0.4 g. Hepatic synthesis of bile acids equals this amount, so that the size of the bile acid pool is maintained at a constant level.

Exocrine Pancreatic Secretion The pancreatic gland is 10-15 cm long and resides in the retroperitoneal space on the posterior wall of the abdomen, with its broad and flat end ("head") fitting snugly into the loop formed by the duodenum. The endocrine function of the pancreas consists of synthesis, storage, and secretion of the hormones insulin, glucagon, and somatostatin into the venous circulation that drains the pancreas. These hormones are essential in the regulation of metabolism of carbohydrates and other substrates (Chapter 22). The endocrine cells are located in the islets of Langerhans, distributed throughout the pancreas but separated from the surrounding exocrine-acinar tissue by collagen fibers. The exocrine function of the pancreas consists of synthesis, storage, and secretion of digestive enzymes and bicarbonate-rich fluids. The acinar cells of the pancreas are arranged in ellipsoidal structures known as acini. Each acinus contains about a dozen cells arranged around a lumen, which forms

202

CHAPTER12 Gastrointestinal Digestion and Absorption

the beginning of the pancreatic duct system, and centroacinar cells, which secrete a bicarbonate solution. The acinar cells are joined to one another by tight junctions, desmosomes, and gap junctions. The tight junctions probably prevent leakage of pancreatic secretions into the extracellular spaces. Acinar cells are pyramidal in shape with the nucleus located away from the lumen and secretory granules located toward the lumen. Centroacinar and acinar cells have microvilli on their free borders. Enzymes are synthesized in the acinar cells by ribosomes situated on the rough endoplasmic reticulum and sequestered in membrane-bound granules (Chapter 25) stored in the apex of the cell. Upon stimulation by the vagus nerve or by cholecystokinin, the contents of the granules are released into the lumen. The centroacinar and duct cells secrete bicarbonate ions under the control of gastrointestinal hormones. The bicarbonate ions are formed by the action of carbonic anhydrase: carbonic anhydrase C 0 2 -Jl-H20 < > H2CO3 Z H + + HCO 3

Large Intestine The large intestine extends from the ileocecal valve to the anus. It is wider than the small intestine except for the descending colon, which when empty may have the same diameter as the small intestine. Major functions of the colon are absorption of water, Na +, and other electrolytes, as well as temporary storage of excreta followed by their elimination. The colon harbors large numbers of mostly anaerobic bacteria that can cause disease if they invade tissues. These bacteria metabolize carbohydrates to lactate, short-chain fatty acids (acetate, propionate, and butyrate), and gases (CO2, CH4, and H2). Ammonia, a toxic waste product, is produced from urea and other nitrogenous compounds. Other toxic substances are also produced in the colon. Ammonia and amines (aromatic or aliphatic) are absorbed and transported to the liver via the portal blood, where the former is converted to urea (Chapter 17) and the latter is detoxified. The liver thus protects the rest of the body from toxic substances produced in the colon. Colonic bacteria can also be a source of certain vitamins (e.g., vitamin K, Chapter 36).

The exact mechanism of HCOf secretion into ductules is not known.

12.2 Gastrointestinal Hormones Composition of Pancreatic Juice Pancreatic juice is alkaline because of its high content of HCO 3. It neutralizes the acidic chyme from the stomach, aided by the bile and small intestinal juices. In the jejunum, chyme is maintained at a neutral pH as required for the optimal functioning of the pancreatic digestive enzymes. A normal pancreas secretes about 1.5-2 L of juice per day. Pancreatic juice contains the proenzymes trypsinogen, chymotrypsinogen, procarboxypeptidases, and proelastase. All are activated by trypsin in the intestinal lumen. Enteropeptidase located in the brush border of the jejunal mucosa converts trypsinogen to trypsin. A trypsin inhibitor in pancreatic juice protects against indiscriminate autodigestion from intraductal activation of trypsinogen. Other enzymes of pancreatic juice and their substrates are listed below.

Enzyme Prophospholipase, activated to phospholipase Cholesteryl ester hydrolase Lipase and procolipase, converted to colipase Amylase Ribonuclease Deoxyribonuclease

Substrate Phospholipids Cholesteryl esters Triacylglycerols Starch and glycogen RNA DNA

Gastrointestinal hormones are chemical messengers that regulate intestinal functions (e.g., motility and secretion), which are also regulated by the autonomic nervous system. The endocrine cells of the GI tract are interspersed with mucosal cells and do not form discrete glands. The GI tract has been described as the largest endocrine organ in the body. In addition, the GI tract is noted for its special significance in the development of endocrinology. The first hormone discovered and characterized as a chemical messenger by Bayliss and Starling in 1902 was the GI hormone secretin. The GI hormones are a heterogeneous group of peptides that are released into the bloodstream in response to specific stimuli, bind to specific receptors on target cells to produce biological responses, and are subject to feedback regulation. Some may affect neighboring cells by being transported through intercellular spaces (paracrine secretion), and others serve as neurotransmitters in peptidergic neurons (Chapter 30). Peptidergic neurons are present in the GI tract and in the peripheral and central nervous systems. These peptides are synthesized in the GI tract and in the brain, although in the GI tract some may originate from vagal nerve endings. This dual localization of "brain-GI tract" peptides is an example of biological conservation. Of the many regulatory gastrointestinal peptides, four are well understood and are considered to be authentic

203

SECTION 12.2 Gastrointestinal Hormones

TABLE 12-1

Selected Peptide Candidate Hormones of the Gut* Name

Bombesin

Intestinal mucosa

Bulbogastrone Enteroglucagon Motilin

Mucosa of duodenal bulb Duodenal mucosa Duodenum and jejunum

Somatostatin

Intestinal mucosa, pancreas and nerves; hypothalamus GI tract (highest concentrations in small intestine and colon) GI tract (all layers and nerve fibers)

Substance P

Vasoactive intestinal peptide

Mechanism of Release and Putative Function

Source

Mechanism of release unknown; stimulates gastric and pancreatic secretions directly and by release of gastrin and CCK, respectively; inhibits absorption of water, Na +, and CI- from the jejunum. Released by acid; inhibits gastric secretion. Functions like glucagon (Chapter 22). Released by vagal stimulation and passage of nutrients; modulates interdigestive motility, clearing the intestine between meals and preparing it for another nutritional bolus. Mechanism of release not clear; multiple inhibitory actions.

Released after food ingestion, contraction of intestinal smooth muscle and gall bladder, reduction of bile flow, and increase in pancreatic juice output. Mechanism of release not clear; inhibits gastric acid and pepsin secretion, potent stimulator of intestinal secretion, vasodilator, possibly neurotransmitter.

*A number of these peptides are found in the gut (in both enteroendocrine cells and enteric neurons) and brain.

hormones: secretin, cholecystokinin (CCK), gastrin, and gastric inhibitory peptide. The hormonal status of other peptides (sometimes referred to as candidate hormones, Table 12-1) is not yet established. On the basis of structural similarity, these hormones fall into two families: 1. The gastrin family, which consists of gastrin, CCK, cerulein, enkephalin, and motilin, and 2. the secretin family, which consists of secretin, glucagon, vasoactive intestinal peptide (VIP), gastric inhibitory peptide (GIP), and bombesin. Other candidate hormones (e.g., neurotensin, substance P, and somatostatin) belong to neither family. Somatostatin, considered a candidate hormone in the GI tract, is an authentic hypothalamic hormone (Chapter 31). The amino acid sequences of some members of the gastrin and secretin families are shown in Tables 12-2 and 12-3, respectively. The hormones exhibit both macroand microheterogeneity. For example, gastrin exists in many forms, probably including the principal hormone, biosynthetic precursors, and metabolites. The sulfate group on tyrosine at position 12 of gastrin may be missing without significant change in biological activity. The biological activity of gastrin and CCK resides in the carboxyterminal tetrapeptide and heptapeptide, respectively.

Gastrin

Gastrin is synthesized and stored in G cells in the antral mucosa of the stomach; it is also found, to a lesser extent, in the mucosa of the duodenum and jejunum. During fetal life, gastrin is also found in the pancreatic islet cells. Gastrin-secreting tumors, known as gastrinomas (e.g., Zollinger-Ellison syndrome), may develop in the pancreas. The main form of gastrin obtained from G cells is a heptadecapeptide. At the N-terminal end of the heptadecapeptide., there is a pyroglutamyl residue, and at the C-terminal end, there is a phenylalaninamide residue, both of which protect the peptide from amino and carboxy-peptidase activity, respectively. Two forms of the heptadecapeptide exist: one in which the tyrosyl residue at position 12 is free (gastrin 1) and the other in which it is sulfated (gastrin 2). Though larger and smaller forms of gastrin have been identified, the biological activity of gastrin resides in the C-terminal tetrapeptide. A synthetic pentapeptide is available (pentagastrin, Table 12-2). Gastrin is released in response to chemical, mechanical, or neural stimuli on the G cell. Hypoglycemia (plasma glucose concentration ~ 200,000) that are integral constituents of the cell membrane (Chapter 10). Their active sites project toward the luminal side. They have pH optima at about 6 and Km values for substrates in the range of 3-20 mmol/L. Some oligosaccharidases are the following:

1. Exo-l,4-a-D-glucosidase, also called glucoamylase or maltase, catalyzes sequential hydrolysis of terminal glucosyl units linked in a(1 -+ 4) linkages from the nonreducing ends of malto-oligosaccharides or maltose. 2. Sucrose a-D-glucohydrolase, also called sucrose-a-dextrinase or sucrase, catalyzes hydrolysis of the a(1 --> 2) linkage of sucrose to release glucose and fructose; of the a(1 -+ 4) linkage of maltose to release two glucose units; and of the a(1 -+ 6) linkage in a-limit dextrins and isomaltose. It is a single gene product that is posttranslationally cleaved to form two distinct polypeptide chains, one chain catalyzing the hydrolysis of an a(1 --> 4) linkage and the other of an a(1 --> 6) linkage. 3. fl-D-Galactoside galactohydrolase, also called lactase, catalyzes hydrolysis of the/3(1 --> 4) linkage of lactose (or terminal nonreducing fl-D-galactose units in fl-D-galactosides) into galactose and glucose. 4. a,a-Trehalose glucohydrolase, also called trehalase, catalyzes the hydrolysis of trehalose into two glucose units. Hydrolysis of branched-chain oligosaccharides (a-limit dextrins) to glucose thus requires sequential action of three enzyme activities (Figure 12-8). Hydrolysis of oligosaccharides is rapid and is not the rate-limiting step in their absorption. However, accumulation of monosaccharides in the lumen is limited by end-product inhibition of the oligosaccharidases. The rate-determining step in absorption is the monosaccharide transport system, with the following exception. Mucosal lactase activity is the lowest oligosaccharidase activity, and so hydrolysis rather than absorption is rate limiting. Oligosaccharide digestion is virtually complete by midjejunum.

Transport of Monosaccharides into the Enterocyte Glucose and galactose compete for a common transport system. This system is an active transport system; i.e., the monosaccharides are absorbed against a concentration gradient, it is saturable and obeys Michaelis-Menten

Surface membrane LumenNa+

~,t" /

60%

site ~, site ~ Glucose--Gluc~C ) .~~ Na+ site

..... Serosal carrier

~ 15% ".. ~C) ~

~ Mucosal "Na+ carrier Ka+K+-KA~

25%

Diffusion y

ATP /" ADP + Pi / "~Na+ K+

FIGURE 12-9 Schematic representation of glucose (or galactose) transport by the enterocyte. Glucose binds to the receptor, facilitated by the simultaneous binding of two Na+ at separate sites. The glucose and Na+ are released in the cytosol as the receptor affinity for them decreases. The Na+ are actively extruded at the basolateral surface into the intercellular space by Na+,K+-ATPase, which provides the energy for the overall transport. Glucose is transported out of the cell into the intercellular space and thence to portal capillaries, both by a serosal carrier and by diffusion. (Reproduced with permission from G. M. Gray, Carbohydrate Absorption and Malabsorption in Gastrointestinal Physiology. Raven Press, New York, 1981.]

kinetics, and it is carrier-mediated and Na+-dependent. Translocation of glucose (or galactose) is shown in Figure 12-9. On the luminal side, one molecule of glucose and two sodium ions bind to the membrane carrier (presumably, Na + binding to the carrier molecule increases the affinity for glucose because of a conformational change). The carrier-bound Na + and glucose are internalized along the electrochemical gradient that results from a low intracellular Na + concentration. Inside the cell, the sodium ions are released from the carrier, and the diminished affinity of the carrier for glucose releases the glucose. The sodium ions that enter in this manner are transported into the lateral intercellular spaces against a concentration gradient by the free energy of ATP hydrolysis catalyzed by a Na+,K+-ATPase. Thus, glucose and Na + are transported by a common carrier, and energy is provided by the transport of Na + down the concentration and electrical gradient. The low cytoplasmic Na + concentration is maintained by the active transport of Na + out of the cell coupled to K + transport into the cell by the Na+,K+-ATPase. Since the sugar transport does not use ATP directly, it can be considered as secondary active transport. Although this mode of glucose transport is the most significant, passive diffusion along a concentration gradient may also operate if the luminal concentration of glucose exceeds the intracellular concentration. The intracellular glucose is transferred to the portal capillary blood by passive diffusion and by a carrier-mediated system. Intracellular glucose can be converted to lactate

212 (Chapter 13), which is transported via the portal blood system to the liver, where it is reconverted to glucose (gluconeogenesis, Chapter 15). The quantitative significance of this mode of glucose transport is probably minimal. Fructose transport is distinct from glucose-galactose transport and requires a specific saturable membrane carrier (facilitated diffusion).

Na+,K+-ATPase Na+,K+-ATPase, in addition to participating in Na+-driven uptake of glucose and amino acids (see below), is responsible for maintaining high intracellular concentrations of K + and low concentrations of Na + (the reverse of the relative concentrations of these ions in the extracellular fluid). The Na + and K + gradient across the cell membrane is involved in the maintenance of osmotic equilibrium, propagation of nerve impulses, reabsorption of solutes by the kidney, and other processes that require the electrochemical energy of the ion gradients. Thus, Na+,K+-ATPase plays a critical role in many important functions of the body. Na+,K+-ATPase is a transmembrane protein found almost exclusively in the plasma membrane. It has two major subunits: ct (M.W. ~ 95,000) and/3 (M.W. ~ 55,000). The latter is a glycoprotein and is exposed to the exterior of the cell. The former spans the entire membrane (Figure 12-10). Minimum subunit stoichiometry required for the activity of the enzyme is c~/3, and the most probable native enzyme structure is (ct/~)2. The enzyme is asymmet-

CHAPTER 12

Gastrointestinal Digestion and Absorption

rically oriented and drives active transport only in one direction. The ATP binding site is located on the cytoplasmic aspect of the ot-subunit. Ouabain, a cardiac glycoside (similar to digitalis glycosides), which inhibits the enzyme, also binds to the ot-subunit but at a site that projects to the exterior of the cell. The inhibition of Na +, K+-ATPase activity indirectly leads to an increase in intracellular Ca 2+ concentration, which stimulates contraction in muscle cells (Chapter 21), thus accounting for the therapeutic effect of cardiac glycosides on the heart. Na+,K +ATPase requires the presence of Na +, K +, Mg 2+, and ATE Each cycle of enzyme activity results in the extrusion of three Na + coupled to the transport of two K + into the cell, with the hydrolysis of one molecule of ATE Thus, the enzyme utilizes the energy derived from ATP hydrolysis to transport K + into the cell and Na + out of the cell, against concentration gradients. Since unequal numbers of monovalent cations are transferred across the plasma membrane, a transmembrane electric current is generated. A model for the mechanism of action of the enzyme is shown in Figure 12-11. It proposes that Na+,K+-ATPase can exist in two (or more) conformational states: one binding Na + or ATP (or both) and the other binding K + or phosphate (or both). On the cytoplasmic side, Na + binding initiates transient phosphorylation of an aspartate residue at the active site, resulting in a cyclic process with translocation of Na + from inside to outside and of K + from outside to inside. The vectorial equation for the transport is 3Na + + 2K + + ATP 4- + H20 3 Nao+ + 2K + + ADP 3- + HPO 2+ + H + where i = inside and o = outside. Thyroid hormone increases Na+,K+-ATPase activity (Chapter 33). Other ATPases participate in the transport of other ions (e.g., K+,H+-ATPase, above; Ca2+-ATPase, Chapter 21).

Disorders of Carbohydrate Digestion and Absorption Carbohydrate malabsorption can occur in a number of diseases that cause mucosal damage or dysfunction (e.g., gastroenteritis, protein deficiency, gluten-sensitive enteropathy). Disorders due to deficiencies of specific oligosaccharidases are discussed below. F I G U R E 12-10 Schematic representation of Na +, K+-ATPase. The minimum subunit stoichiometry of the active enzyme is c~/3, and the native enzyme most likely has (c~/~)2 structure. The enzyme spans the plasma membrane, with the smaller/3 glycoprotein subunits projecting outside the cell. Each functional unit has binding sites on both sides of the membrane; the outer surface has K + and cardiac glycoside binding sites, and the inner surface has Na + and ATP binding sites. [Reproduced with permission from K. J. Sweadner and S. M. Goldin, Active transport of sodium and potassium ions. N. Engl. J. Med. 302, 777 (1980).]

Lactose Intolerance (Milk Intolerance) Lactose intolerance is the most common disorder of carbohydrate absorption. Lactase deficiency occurs in the majority of human adults throughout the world and appears to be genetically determined. The prevalence is high in persons of African and Asian ancestry (~> 65%) and low in persons of Northern European ancestry. Lactase deficiency in which mucosal lactase levels are low or absent at birth

213

SECTION12.3 Digestion and Absorption of Major Food Substances Outside

3 No+

Outside

/ns,de

No* No+ No+

\ ATP

t2K*~

+Pi.

F I G U R E 12-11 Model for transport of Na + and K + by Na +,K +-ATPase. Inside the cell, Na + initiates the one-way ion exchange cycle by the phosphorylation of an aspartate residue at the active site by ATE which eventually leads to the translocation of Na + and K +. Conformational changes of the enzyme occur during the exchange of ions. [Reproduced, with permission, from K. J. Sweadner and S. M. Goldin, Active transport of sodium and potassium ions. New Engl. J. Med. 302, 777 (1980).]

is rare and is transmitted as an autosomal recessive trait. Prompt diagnosis and a lactose-free diet assure the infant's normal growth. Since hydrolysis of lactose by lactase is rate-limiting, any mucosal damage will cause lactose intolerance. In full-term human infants, lactase activity attains peak values at birth and remains high throughout infancy. As milk intake decreases, lactase levels drop and lactose intolerance may develop. The extent of the decrease of lactase activity distinguishes lactose-tolerant from intolerant populations. Ingestion of milk (or lactose) by individuals who have lactose intolerance leads to a variety of symptoms (bloating, cramps, flatulence, and loose stools). Severity of the symptoms depends on the amount of lactose consumed and the enzyme activity. Symptoms disappear with elimination of lactose from the diet. Eight ounces of milk contains about 12 g of lactose. Some individuals show symptoms with consumption of as little as 3 ounces of milk. Some lactose-intolerant individuals can drink milk if it is consumed along with meals and show no reaction to milk added to cereal or coffee. Lactose-depleted milk or fermented milk products with negligible amounts of lactose are good substitutes for milk. The intestinal problems are due primarily to osmotic effects of lactose and its metabolites in the colon. The lactose not absorbed in the small intestine increases the osmolarity and causes water to be retained in the lumen. In the colon, it is metabolized by bacterial enzymes to a number of short-chain acids, further increasing osmolarity and aggravating fluid reabsorption. The bacterial fermentation also yields gaseous products (H2, CO2, and CH4), hence the bloating, flatulence, and sometimes frothy diarrhea.

A lactose tolerance test is performed by oral administration of a load of lactose (100 g in an adult), measurement of blood glucose at specific time intervals, and occurrence of characteristic symptoms. The values for blood glucose are compared with those from an oral glucose tolerance test performed the previous day on the same individual. The hydrogen breath test can also be used in the assessment of lactose malabsorption. Hydrogen is a product of bacterial fermentation of undigested sugar in the colon and is not a product of human metabolism. Some of this hydrogen is absorbed and excreted by the lungs. Measurement of the amount of hydrogen in the expired air by gas chromatographic studies provides a measure of the unabsorbed carbohydrate in the colon. Both tests can be adapted for the diagnosis of malabsorption disorders for other carbohydrates.

Intolerance of Other Carbohydrates Intolerance to sucrose and o~-limit dextrins may be due to deficiency of sucrase-o~-dextrinase or to a defect in glucose-galactose transport. These disorders are rare autosomal recessive traits; clinical problems can be corrected by removing the offending sugars from the diet. Lactulose, a, synthetic disaccharide consisting of galactose and fructose with a /3(1 --> 4) linkage, is hydrolyzed not in the small intestine but in the colon and is converted to products similar to those derived from lactose fermentation. It has been used in the treatment of patients with liver disease. Normally, ammonia (NH3) produced in the GI tract is converted in the liver to urea (Chapter 17), hence, in patients with severe liver disease, blood ammonia levels increase. Absorption of ammonia can be decreased by administration of lactulose, which acidifies the colonic constituents so that NH3 is trapped as NH4+ ions (Chapter 9).

~1~

CHAPTER12 Gastrointestinal Digestion and Absorption

Proteins Protein is an essential nutrient for human growth, development, and homeostasis. The nutritive value of dietary proteins depends on its amino acid composition and digestibility. Dietary proteins supply essential amino acids, which are not synthesized in the body. Nonessential amino acids can be synthesized from appropriate precursor substances (Chapter 17). In human adults, essential amino acids are valine, leucine, isoleucine, lysine, methionine, phenylalanine, tryptophan, and threonine. Histidine (and possibly arginine) appears to also be required for support of normal growth in children. In the absence from the diet of an essential amino acid, cellular protein synthesis does not occur. The diet must contain these amino acids in the proper proportions. Thus, quality and quantity of dietary protein consumption and adequate intake of energy (carbohydrates and lipids) are essential. Protein constitutes about 10-15% of the average total energy intake. Animal proteins, with the exception of collagen (which lacks tryptophan), provide all of the essential amino acids. Vegetable proteins differ in their content of essential amino acids (Table 12-4), but a mixture of these proteins will satisfy the essential amino acid requirement. For example, the lysine lacking in grains can be provided by legumes. This combination also corrects for the methionine (which is supplied in corn) deficiency of legumes. Such combinations (e.g., lentils and rice, chick-peas and sesame seeds, spaghetti and beans, corn and beans) are widely used in different cultures to provide for an optimal protein requirement. The recommended allowance for mixed proteins in an adult in the U.S.A. is 0.85 g per kilogram of body weight per day (Chapter 5). The allowances

TABLE 12-4 Limiting Essential Amino Acids in Plant Proteins Source of Plant Proteins Grain Maize or corn Millet, oats, wheat, or rice Legumes Beans, immature Beans, mature Peas Peanuts Nuts and oil seeds Coconut Vegetables

Limiting Amino Acids Lysine, tryptophan Lysine, threonine Methionine, isoleucine Methionine, valine Methionine, tryptophan Lysine, threonine Lysine, threonine Lysine, threonine Methionine, isoleucine

are increased during childhood, pregnancy, and lactation (Appendix IV). Besides dietary protein, a large amount of endogenous protein undergoes digestion and absorption. Endogenous protein comes from three sources: 1. Enzymes, glycoproteins, and mucins secreted from the salivary glands, stomach, intestine, biliary tract, and pancreas, which together constitute about 20-30 g/day; 2. Rapid turnover of the gastrointestinal epithelium, which contributes about 30 g/day; and 3. Plasma proteins that normally diffuse into the intestinal tract at a rate of 1-2 g/day. In several disorders of the GI tract (e.g., protein-losing gastroenteropathy), loss of plasma proteins is considerable and leads to hypoproteinemia.

Digestion Protein digestion begins in the stomach, where protein is denatured by the low pH and is exposed to the action of pepsin. The low pH also provides the optimal H + concentration for pepsin activity. The zymogen precursor pepsinogen (M.W. 40,000) is secreted by the chief cells and is converted to pepsin (M.W. 32,700) in the acid medium by removal of a peptide consisting of 44 amino acid residues. This endopeptidase hydrolyzes peptide bonds that involve the carboxyl group of aromatic amino acid residues, leucine, methionine, and acidic residues (Table 12-5). The products consist of a mixture of oligopeptides. Chyme contains potent secretagogues for various endocrine cells in the intestinal mucosa. CCK and secretin cause release of an alkaline pancreatic juice containing trypsinogen, chymotrypsinogen, proelastase, and procarboxypeptidases A and B. Activation begins with that of trypsinogen to trypsin by enteropeptidase (previously called enterokinase) present in the brush-border membranes of the duodenum. Enteropeptidase cleaves between Lys-6 and Ile-7 to release a hexapeptide from the N-terminus. Trypsin autocatalytically activates trypsinogen and activates the other zymogens. The importance of the initial activation of trypsinogen to trypsin by enteropeptidase is manifested by children with congenital enteropeptidase deficiency who exhibit hypoproteinemia, anemia, failure to thrive, vomiting, and diarrhea. Trypsin, elastase, and chymotrypsin are endopeptidases. Carboxypeptidases are exopeptidases (Table 12-5). The combined action of these enzymes produces oligopeptides having two to six amino acid residues and free

SECTION 12.3

Digestion and Absorption of Major Food Substances

215

TABLE 12-5

Specificities of the Proteolytic Enzymes Involved in Gastrointestinal Protein Digestion Peptide bond cleaved o

H2N. . . . . N H - C H -

//o

oiI

N H - C H - C . . . . . C\

I R1

I

R2

OH

Enzyme*

Preferentially cleaves peptide bonds in which

Trypsin Chymotrypsin Carboxypeptidase A Carboxypeptidase B Elastase Aminopeptidase Pepsin Dipeptidases

R1R1R1R1R1R1R] =

Arg or Lys; R 2 = any amino acid residue aromatic amino acid (Phe, Tyr, Trp); R 2 - any amino acid residue any amino acid residue; R2 - any C-Terminal residue except Arg, Lys, or Pro any amino acid residue; R2 - Arg or Lys at the C-terminus of a polypeptide Neutral (uncharged) residues; R 2 - a n y amino acid residue Most N-terminal residues of a polypeptide chain; R 2 - any residue except Pro Trp, Phe, Tyr, Met, Leu; R 2 = any amino acid residue H2N-CH - O

NH-CH-C,~ O

I

I

R2

R1

OH

Peptide bond cleaved Tripeptidases

O II

O

H2N-CI H-

N H - C HI- C

R1 R 1, R2, R 3 - any amino acid residue

R2

,O

NH-CH-C// I "

R3

OH

Choice of bond cleaved depends on specific enzyme. In a given molecule, only one bond is cleaved by a tripeptidase.

*Trypsin, chymotrypsin, elastase, and pepsin are endopeptidases (trypsin is the most specific and pepsin is the least specific)" carboxypeptidaseA and B and aminopeptidase are exopeptidases.

amino acids. Hydrolysis of oligopeptides by the brushborder aminopeptidases releases amino acids. Leucine aminopeptidase, a ZnZ+-containing enzyme, is an integral transmembrane glycoprotein with a carbohydrate-rich hydrophilic portion and active site protruding into the luminal side. It cannot degrade proline-containing dipeptides, which are largely hydrolyzed intracellularly. Dipeptidases and tripeptidases are associated with the brush-border membranes, but their functions are not clearly understood. The major products of intraluminal digestion of protein are mixtures of amino acids (30-40%) and small peptides (60-70%). Chymotryptic activity can be measured by oral administration of N-benzoyl-L-tyrosyl-p-amino acid (Figure 12-12). On hydrolysis, p-aminobenzoic acid (PABA) and N-benzoyl-L-tyrosine are released; PABA is absorbed, conjugated to glycine by the liver, and excreted

in the urine to give an index of exocrine pancreatic function.

Absorption of Amino Acids and Oligopeptides Dipeptides and tripeptides that escape brush border membrane peptidases are actively transported against a concentration gradient by Na+-dependent mechanisms. Inside the cell they are hydrolyzed. Free amino acids are transported into enterocytes by four active, carrier-mediated, Na+-dependent transport systems remarkably similar to the system for glucose. These systems transport, respectively, neutral amino acids; basic amino acids (Lys, Arg, His) and cystine; aspartic and glutamic acids; and glycine and imino acids. Some amino acids (e.g., glycine) have affinities for more

216

CHAPTER12 Gastrointestinal Digestion and Absorption OH

1

S/,teo, c,eavaoe by chymotrypsin

mNHmCH uC--NH

COOH

N-Benzoyl-L-tyrosyl-p-aminobenzoicacid H20 OH

I

I--I=N~ C O O H

~

O

II

CH= O

I

II

C--NH --CH --C--OH N-BenzoyI-L-tyrosine

p-Aminobenzoic acid (PABA) Absorbed and excretedin urine as p-aminobenzoylglycine

F I G U R E 12-12 Hydrolysis of a synthetic tripeptide by chymotrypsin in the small intestine. The hydrolysis yields PABA, which is absorbed and eventually excreted in the urine. The concentration of PABA is a measure of exocrine pancreatic function.

than one transport system; within each group, amino acids compete with each other for transport. These systems are specific for L-Amino acids. D-Amino acids are transported by a passive diffusion process. Amino acid transport in the renal cells may use similar systems (Chapter 39). Entry of amino acids into cell compartments elsewhere in the body may require different transport systems (e.g., the y-glutamyl cycle, Chapter 17). In the enterocyte, amino acids may be metabolized or transported to the liver. Glutamate, glutamine, aspartate, and asparagine are metabolized in the enterocyte (Chapter 17). Small amounts of protein (e.g., dietary, bacterial, and viral) may be absorbed intact by nonselective pinocytosis. This absorption may be more common in neonatal life. Absorption of food proteins (or antigenic peptides from them) can cause allergic manifestations, whereas bacterial and viral antigens stimulate immunity by production and secretion of secretory IgA (Chapter 35). Since thyrotropin-releasing hormone (pGlu-His-Pro-NH2) is resistant to hydrolysis, it is effective if taken orally (Chapter 33).

Disorders of Protein Digestion and Absorption The principal causes of protein maldigestion and malabsorption are diseases of the exocrine pancreas and

small intestine. Primary isolated deficiency of pepsinogen or pepsin, affecting protein assimilation, has not been described. Deficiencies of trypsinogen and enteropeptidase are rare. Defects in neutral amino acid transport (Hartnup disease), in basic amino acids and cystine (cystinuria), dicarboxylic aminoaciduria, and aminoglycinuria have been reported. The clinical severity of these disorders is usually minimal and relates to the loss of amino acids or relative insolubility of certain amino acids in the urine. In cystinuria, for example, cystine can precipitate in acidic urine to form stones. In Hartnup disease, severe nutritional deficiencies are uncommon, since the essential amino acids are absorbed as dipeptides or oligopeptides. Tryptophan, a precursor of nicotinamide (a vitamin; see Chapter 38), and NAD + and NADP + (Chapter 17) are lost in this disease; skin and neuropsychiatric manifestations characteristic of nicotinamide deficiency respond to oral nicotinamide supplementation.

Lipids Dietary fat provides energy in a highly concentrated form and accounts for 40--45% of the total daily energy intake (100 g/day in the average Western diet). Lipids contain more than twice the energy per unit mass than carbohydrates and proteins (Chapter 5). The efficiency of fat absorption is very high; under normal conditions, almost all ingested fat is absorbed, with less than 5% appearing in the feces. The predominant dietary lipid is triacylglycerol (previously called triglyceride), which contains three long-chain (16-carbon or longer) fatty acids (Chapter 18). The dietary lipids include essential fatty acids (Chapter 18) and the lipid-soluble vitamins A, D, E, and K (Chapters 36-38). Digestion and absorption of lipids involves the coordinated function of several organs but can be divided into three phases: luminal, intracellular, and secretory. Intraluminal Phase

Digestion of lipid in the mouth and stomach is minimal. However, lipases secreted by lingual glands at the base of the tongue are active at acid pH and initiate the hydrolysis of triacylglycerol without a requirement for bile acids. The fatty acids released'stimulate the release of CCK and a flow of bile and pancreatic juice. The free fatty acids also stabilize the surface of triacylglycerol particles and promote binding of pancreatic colipase. This phase aids in the optimal action of pancreatic lipase and is particularly important in disorders of pancreatic function or secretion (e.g., in prematurity, cystic fibrosis, congenital deficiency of pancreatic lipase).

SECTION12.3 Digestion and Absorption of Major Food Substances O

II

1

O

H=?2--O--C--R II R--C--O---CH O

I~

II

0

Pancreatic

,lipase

+ 21"120

l

3

Fatty acid

O

II

O

H~?2--O---C-- R,

R~---C--O~H

+ 2RCOO- + 2H+

H2C--OH 2-Monoacylglycerol

Triacylglycerol

1

H2?--OH

II R--C--O--CH

H2C--O--C--R

~)

217

O

II

II

CH3

I+

H --O--P--O-I_ CH2---CH2--'?-CH3 O

H2?--O--C--R, phospholipase A2 + H20

CH 3

Phosphatidycholine

HC--OH O

CH~

II I§ H~C--O--l~yO-- CHF--CHF-- ? --CH3 I

O Lysophosphatidylcholine

+ ~COO-+ H+ Fatty acid

CH 3

F I G U R E 12-13 Hydrolysis of triacylglycerol and phosphatidylcholine by pancreatic lipase and phopholipase A2, respectively.

The major functions of the stomach in fat digestion are to store a fatty meal, to contribute to emulsification by the sheafing actions of the pylorus, and to gradually transfer the partially digested emulsified fat to the duodenum by controlling the rate of delivery. The hydrolysis of triacylglycerol in the duodenum and jejunum requires bile and pancreatic juice. Bile acids are powerful detergents that, together with monoacylglycerol and phosphatidylcholine, promote the emulsification of lipids. Emulsification is also aided by the churning action of the GI tract which greatly increases the area of the lipid-water interface that promotes the action of pancreatic lipase. This triacylglycerol lipase hydrolyzes ester linkages at the 1- and 3-positions to yield 2-monoacylglycerol and fatty acids (Figure 12-13). The products of digestion are relatively insoluble in water but are solubilized in micelles. Micelles also contain lipidsoluble vitamins, cholesterol, and phosphatidylcholine. Pancreatic lipase functions at the lipid-water interface, its activity being facilitated by colipase (M.W. ~ 10,000), also secreted by the pancreas as procolipase activated by tryptic hydrolysis of an Arg-Gly bond in the N-terminal region. Colipase anchors the lipase to the triacylglycerol emulsion in the presence of bile salts by forming a 1:1 complex with lipase and protects lipase against denaturation. Colipase deficiency (with normal lipase) is accompanied by significant lipid malabsorption, as is pancreatic lipase deficiency. Pancreatic juice contains esterases that act on short-chain triacylglycerols and do not require bile salts, as well as cholesteryl esterase. Phosphatidylcholine in the diet (4-8 g/day) and in bile secretions (17-22 g/day) is hydrolyzed to lysophosphatidylcholine and fatty acid by

phospholipase A2, a pancreatic enzyme with an absolute requirement for Ca 2+ ions and for bile acids. The secreted form, prophospholipase A2, is activated by tryptic hydrolysis of an Arg-Ala bond in the N-terminal region. Phospholipase A2 also hydrolyzes fatty acids esterified at the 2-position of phosphatidylethanolamine, phosphatidylglycerol, phosphatidylserine, phosphatidylinositol, and cardiolipin but has no effect on sphingolipids. Lipid absorption in the duodenum and jejunum appears to be a passive diffusion process. Lipid-laden micelles migrate to the microvilli, and the fatty acids, monoacylglycerols, and lysophosphoglycerols are transferred across the membrane according to their solubility within the lipid bilayer of the cell surfaces. Bile acids are not absorbed into the enterocyte but migrate to the ileum, where they are actively absorbed and transferred to the liver via the portal venous system. The bile acid pool is recycled several times daily (enterohepatic circulation) to meet the demands of lipid digestion, and disorders that interfere with this process lead to malabsorption of lipids. A cytoplasmic fatty acid-binding protein with high affinity for long-chain fatty acids transports fatty acids to the smooth endoplasmic reticulum for resynthesis of triacylglycerol. Digestion and absorption of triacylglycerols with medium-chain fatty acids (~

D-fructose-l,6-bisphosphate 4- + ADP 3+ + H +

I (~lnsulin~--------------------~ ATP

Glucokinase gene expression

Via Blockage of ATP sensitive K§ channel Insulin secretion (~

I Plasma Insulin(~ Insulin-Sensitive Tissues Skeletal and Cardiac Muscle, Adipose tissue GLUT-4 Transporter(~ Utilization ~of Glucose(~)

F I G U R E 13-2 The role of tissue-specificglucokinasesof liver and pancreatic fl cells. During the periods of abundant supplyof glucose (e.g., postprandial conditions), the plasmalevels of glucose are increased. This induces glucokinase expressionin pancreatic islet/3 cells with eventualinsulin secretion. In the hepatocytes,insulin induces the glucokinaseexpression with accompanyingincreased metabolism.Insulin also enhances glucose utilization by recruiting glucosetransporters, GLUT-4in insulin-sensitive tissues, j" = Increased; (~ = positiveeffect.

channels. The increased cytosolic Ca 2+ triggers the secretion of insulin (Chapter 22). Tissue-specific glucokinases expressed in liver and pancreatic islet/3 cells are differentially regulated. The glucokinase gene consists of two different transcription control regions. Other examples of genes that contain different transcription control regions (promoters) yet produce mRNAs that code for identical proteins, are or-amylase and O~l-antitrypsin genes. In the hepatocytes, insulin promotes glucokinase expression, whereas in /3 cells insulin has no effect but glucose promotes enzyme expression. In the hepatocytes, cAMP (and thus glucagon) and insulin have opposing effects on the expression of glucokinase (Chapter 22). Tissuespecific glucose transporters and glucokinases play critical roles in glucose homeostasis (Figure 13-2). Mutations that alter these proteins may lead to inherited forms of diabetes mellitus (Chapter 22).

Isomerization of Glucose-6-Phosphate to Fru cto s e-6- Pho sp hate This freely reversible reaction requires Mg 2+ and is specific for glucose-6-phosphate and fructose-6-phosphate. It is catalyzed by glucose-phosphate isomerase. ot-D-glucose-6-phosphate ~ oe-D-fructose-6-phosphate

The reaction is essentially irreversible under physiological conditions and is a major regulatory step of glycolysis. PFK-1 is an inducible, highly regulated, allosteric enzyme. In its active form, muscle PFK-1 is a homotetramer (M.W. 320,000) that requires K + or NH~-, the latter of which lowers Km for both substrates. When adenosine triphosphate (ATP) levels are low during very active muscle contraction, PFK activity is modulated positively despite low concentration of fructose-6-phosphate. Allosteric activators of muscle PFK-1 include adenosine monophosphate (AMP), adenosine diphosphate (ADP), fructose-6-phosphate, and inorganic phosphate (Pi); inactivators are citrate, fatty acids, and ATP. The most potent regulator of liver PFK-1 is fructose2,6-bisphosphate, which relieves the inhibition of PFK-1 by ATP and lowers the Km for fructose-6phosphate. Fructose-2,6-bisphosphate is a potent inhibitor of fructose-l,6-bisphosphatase (which is important in gluconeogenesis) and thus ensures the continuation of glycolysis. Metabolism of fructose-2,6-bisphosphate, its role as activator of PFK-1 and inhibitor of fructose-l,6bisphosphatase, and its allosteric and hormonal regulation are discussed in Chapter 15.

Cleavage of Fructose-l,6-Bisphosphate into Two Triose Phosphates In this reversible reaction, aldolase (or fructose-l,& bisphosphate aldolase) cleaves fructose-l,6-bisphosphate into two isomers: D-Fructose- 1,6-bisphosphate 4- ~ D-Glyceraldehyde 3-phosphate 2- + dihydroxyacetone phosphate 2-

Isomerization of Dihydroxyacetone Phosphate to Glyceraldehyde 3-Phosphate In this reversible reaction, triose-phosphate isomerase converts dihydroxyacetone phosphate to D-glyceraldehyde 3-phosphate, which is the substrate for the next reaction. Thus, the net effect of the aldolase reaction is to yield

CHAPTER13 Carbohydrate Metabolism I: Glycolysis and TCA Cycle

230

~

~

~176 Ribose

o

I 0-- P LDH-2

Oxidized NBT ~

Reduced NBT

Elevation of LDH-5

(colored and insolublel

As long as the total activity of LDH is not very high so that the substrates are not limiting, the intensity of the color is approximately proportional to the various isoenzyme activities. Thus, densitometric scanning of an electropherogram provides an estimate of the activities of the individual enzymes as well as quantitation of their relative intensities. LDH-1 is the most negatively charged and fastest moving isoenzyme (mobility comparable to that of the ot 1-globulin region in serum protein electrophoresis). LDH-5 is the least negatively charged and slowest migrating isoenzyme (mobility comparable to that of ),-globulin); the other isoenzymes possess intermediate mobilities. The normal relative intensity of LDH isoenzyme fractions in serum is LDH-1 < LDH-2 > LDH-3 > LDH-4 < = > LDH-5 Since myocardium is rich in LDH-1, injury to that tissue results in the elevation of LDH- 1 and the ratio of LDH- 1 to LDH-2; similarly, since liver is rich in LDH-4 and LDH-5, elevations of these isoenzymes suggests liver disease. Because a particular isoenzyme pattern may result from injury to more than one tissue (Table 13-3), further diagnostic tests are required. Forexampte, measurement of creatine kinase and its isoenzyme determination and troponin I in the serum is helpful in the evaluation of acute injury to the myocardium. Because of differences in halflives in serum, serial determination of total enzyme activity and relative isoenzyme composition aids in the diagnosis and assessment of the magnitude of the injury of the tissue in question (Chapter 8). In patients with acute myocardial infarction, serial analysis every 8-12 hours during the first 48 hours after the onset of symptoms is useful. In vitro, LDH-I(H4) has a low Km for pyruvate and is strongly inhibited by high concentration of pyruvate. This form may favor the oxidation of lactate to pyruvate in aerobic tissues such as the heart. LDH-5(M4) exhibits a higher Km value for pyruvate and is less susceptible to inhibition by high concentrations of pyruvate than is LDH-1.

Elevation of LDH-3, frequently LDH-3 > LDH-2 Elevation of LDH-2 and LDH-3 All isoenzymes elevated

Disorder Myocardial infarction Renal cortical infarction Pernicious anemia Hemolysis Muscular dystrophy (later stages) Liver disease Skeletal muscle damage Some cancers Some neoplastic diseases Lymphoproliferative disorders Platelet-related disorders Pulmonary infarction Widespread tissue injury

*Normal distribution: LDH-I < LDH-2 > LDH-3 > LDH-4 < = > LDH-5

This form may occur in anaerobic tissues, in which lactate is the end product of glycolysis. However, in vitro differences in kinetic properties of the isoenzyme may be inappropriate to explain actual physiological actions for several reasons: differences in kinetic properties between LDH-1 and LDH-5 are less marked at 37~ (body temperature) than at 25~ high intracellular concentrations of the enzyme are present, and differences in the actual ratio of ketopyruvate to enolpyruvate exist (the enol form may be the more potent inhibitor). Furthermore, the occurrence of similar isoenzyme patterns in widely different tissues with divergent metabolic goals (e.g., LDH-5 in liver and muscle; LDH-1 in heart and erythrocytes) points out our tack of understanding of their precise role.

Alternative Substrates of Glycolysis The glycolytic pathway is also utilized by fructose, galactose, mannose, glycogen, and glycerol. The metabolism of the monosaccharides and glycogen is discussed in Chapter 15. Glycogenolysis catalyzed by phosphorylase in a nonequilibrium reaction yields glucose-1-phosphate, which is then converted to glucose-6-phosphate (Chapter 15), bypassing the initial phosphorylation reaction of glycolysis. Therefore, the conversion of a glucosyl unit of glycogen to two lactate molecules yields three ATP, which is 50% more than the yield from a glucose molecule. Glycerol released by hydrolysis of

SECTION 13.2

235

Pyruvate Metabolism

triacylglycerol enters the glycolytic pathway by way of dihydroxyacetone phosphate, as follows: Glycerol + ATP4-

glycerol kinase, Mg2+

> glycerol 3-phosphate2+ ADP3- + H+ glycerol-3-phosphate dehydrogenase

Glycerol 3-phosphate2- + NAD+


dihydroxyacetone phosphate2- + NADH + H+

Role of Anaerobic Glycolysis in Various Tissues and Cells In tissues that lack mitochondria or function under limiting conditions of oxygen (e.g., lens and erythrocytes), glycolysis is the predominant pathway providing ATE On the basis of speed of contraction and metabolic properties, skeletal muscle fibers may be classified into three types: type I (slow-twitch, oxidative), type IIA (fast-twitch, oxidativeglycolytic), and type IIB (fast-twitch, glycolytic). Shortterm, sudden energy output is derived from glycolytic fibers. The glycolytic fibers (particularly type IIB) have few mitochondria, with relative enrichment of myofibrils and poor capillary blood supply. Thus, these fibers can perform a large amount of work in a short period of time, in contrast to slow-twitch fibers, which are rich in mitochondria, have good capillary blood supply, and power sustained efforts (Chapter 21). A high rate of glycolysis also occurs in lymphocytes, kidney medulla, skin, and fetal and neonatal tissues. The anaerobic capacity of lymphocytes is increased for growth and cell division. Kidney medulla (Chapter 39), which contains the loops of Henle and collecting tubules, receives much less blood than the cortex and is rich in glycogen. In contrast, kidney cortex, which contains the glomeruli, proximal tubules, and parts of the distal tubules, receives a large amount of blood through the renal arterioles. The renal cortex derives its energy from oxidation of glucose, fatty acids, ketone bodies, or glutamine to CO2 and H20. Some rapidly growing tumor cells exhibit a high rate of glycolysis. Inadequate oxygen supply (hypoxia) causes tumor cells to grow more rapidly than the formation of blood vessels that supply oxygen. Thus, in hypoxic tumor cells, glycolysis is promoted by increased expression of glucose transporters and most glycolytic enzymes by hypoxia-inducible transcription factor (HIF- 1).

reduce their normal 120-day life span. For example, deficiency of hexokinase, glucose-phosphate isomerase, 6-phosphofructokinase, aldolase, triose-phosphate isomerase, or phosphoglycerate kinase is associated with hemolytic anemia, whereas lactate dehydrogenase deficiency is not. The most common deficiency is that of pyruvate kinase, (PK), which is inherited as an autosomal recessive trait. The PK deficiency occurs worldwide, however, it is most commonly found in kindreds of Northern European ancestry. Erythrocyts of PK individuals have elevated levels of 2.3-DPG and decreased ATP level. Several mutations of the PK gene have been identified. Individuals with PK deficiency suffer from lifelong chronic hemolysis to a varying degree. Splenectomy ameliorates hemolytic process in severe cases. Some enzymopathies of erythrocytes may be associated with multisystem disease (e.g., aldolase deficiency with mental and growth retardation). Individuals with 6-phosphofructokinase deficiency exhibit hemolysis and myopathy and have increased deposition of muscle glycogen (a glycogen storage disease; see Chapter 15). The myopathy is usually characterized by muscle weakness and exercise intolerance. (See also Chapters 10, 15, and 28.)

13.2 Pyruvate Metabolism Pyruvate has several metabolic fates. It can be reduced to lactate, converted to oxaloacetate in a reaction important in gluconeogenesis (Chapter 15) and in an anaplerotic reaction of the TCA cycle (see below), transminated to alanine (Chapter 17), or converted to acetyl-CoA and CO2. Acetyl-CoA is utilized in fatty acid synthesis, cholesterol (and steroid) synthesis, acetylcholine synthesis, and the TCA cycle (Figure 13-6).

ALANINE

GLUCOSE

y/Glycolysis PYRUVATE

OXALOACETATE

-.,_

ACETYL CoA - ~

Gluconeogenesis

Glycolytic Enzyme Deficiencies in Erythrocytes The only pathway that provides ATP in mature erythrocytes is glycolysis. Because these cells lack mitochondria, a nucleus, and other organelles required for protein synthesis, deficiency of glycolytic enzymes may

GLUCOSE

~

~ LACTATE

Fatty acids

~ TCA cycle CO 2 + H20

FIGURE 13-6 Major pathwaysof pyruvatemetabolism. Pyruvateis metabolizedthrough four majorenzymepathways:Lactatedehydrogenase(LDH),pyruvate dehydrogenasecomplex(PDH),pyruvatecarboxylase(PC), and alanine aminotransferase (ALT). Arrowsindicate multiple steps.

~36

CHAPTER13

Conversion of pyruvate to ethanol by certain yeast strains occurs in two steps. It is first decarboxylated to acetaldehyde by pyruvate decarboxylase, which utilizes thiamine pyrophosphate (TPP) as coenzyme. m g 2+

CH3COCOOH - - + CH3CHO + C02 TPP

In the second step, acetaldehyde is reduced to ethanol by alcohol dehydrogenase, an NAD-dependent enzyme. CH3CHO + NADH + H + ~ CH3CH2OH + NAD + The NADH is derived from glyceraldehyde 3-phosphate dehydrogenation. Thus, yeast fermentation yields ethanol rather than lactate as an end product of glycolysis. Small amounts of ethanol are produced by the microbial flora of the gastrointestinal tract. Other types of fermentation using similar reactions occur in microorganisms and yield a variety of products (e.g., acetate, acetone, butanol, butyrate, isopropanol, hydrogen gas). Lactic Acidemia and Lactic Acidosis

As discussed earlier, some tissues produce lactate as an end product of metabolism. The lactate produced is L-lactate and is commonly referred to simply as lactate. As we will see in the following discussion, D-lactate is also produced under certain pathological conditions, which presents a unique clinical problem. Under normal conditions, lactate is metabolized in the liver and the blood lactate level is between 1 and 2 mM. Lactate accumulation in body fluids can be due to increased production and/or decreased utilization. Blood lactate-to-pyruvate ratio below 25 suggests defects in a gluconeogenic enzyme (Chapter 15) or pyruvate dehydrogenase (discussed later). A common cause of lactic acidosis is tissue hypoxia caused by shock, cardiopulmonary arrest, and hypoperfusion. Inadequate blood flow leads to deprivation of oxygen and other nutrients to the tissue cells as well as to the removal of waste products. Oxygen deprivation leads to decreased ATP production and accumulation of NADH, which promotes conversion of pyruvate to lactate. A major cause of acidosis that occurs during inadequate cellular oxygen delivery is continual hydrolysis of the available supply of ATP that releases protons:

Carbohydrate Metabolism I: Glycolysis and TCA Cycle

D-Lactic Acidosis

This unusual form of lactic acidosis is due to increased production and accumulation of D-lactate in circulation. The normal isomer synthesized in the human body is L-lactate but the D-lactate isomer can occur in patients with jejunoileal bypass, small bowel resection, or other types of short bowel syndrome. In these patients, ingested starch and glucose bypass the normal metabolism in the small intestine and lead to increased delivery of nutrients to the colon where gram-positive, anaerobic bacteria (e.g., Lactobacilli) ferment glucose to D-lactate. The D-lactate is absorbed via the portal circulation. A limited quantity of D-lactate is converted to pyruvate by a mitochondrial flavoprotein enzyme D-2-hydroxy acid dehydrogenase. Thus, the development of D-lactate acidosis requires excessive production of D-lactate and an impairment in its metabolism. The clinical manifestations of D-lactic acidosis are characterized by episodes of encephalopathy after ingestion of foods containing carbohydrates. The diagnosis of D-lactic acidosis is suspected in patients with disorders of the small intestine causing malabsorption and when the serum anion gap (Chapter 39) is elevated in the presence of normal serum levels of L-lactate and other organic acids. Measurement of serum D-lactate requires special enzymatic procedures utilizing D-lactate dehydrogenase and NADH. As D-lactate is converted to pyruvate, NADH is oxidized to NAD + which is detected spectrophotometrically (Chapter 8). The treatment of D-lactic acidosis consists of oral administration of antibiotics, limitations of oral carbohydrate intake, and recolonization of the colon by bacterial flora which do not produce D-lactate.

Oxidation of Pyruvate to Acetyl-CoA Pyruvate must first be transported into mitochondria by a specific carrier that cotransports a proton to maintain electrical neutrality. Inside mitochondria pyruvate undergoes oxidative decarboxylation by three enzymes that function sequentially and are present as a complex known as the pyruvate dehydrogenase complex. The overall reaction is physiologically irreversible, has a high negative A G O' ( - 8 . 0 kcal/mol, or - 3 3 . 5 kJ/mol), and commits pyruvate to the formation of acetyl-CoA:

ATP 4- + H20 --+ ADP 3- + HPO 2- + H + Pyruvate + NAD + + CoASH --> Laboratory assessment includes measurements of blood lactate, pyruvate, /~-hydroxybutyrate, and acetoacetate (Chapter 39). The primary treatment of lactic acidosis involves correcting the underlying cause such as reversal of circulatory failure.

acetyl-CoA + NADH + H + + CO2 In the above reaction CoASH stands for coenzyme A (Figure 13-7), and it contains the vitamin pantothenic acid (Chapter 38). Coenzyme A functions as a carrier of

SECTION13.2 Pyruvate Metabolism

237

NH, 4"-Phosphopantetheine

HI

'

I 3 CH

I O-

HS--OH i--'CH [-- N H - - I - - CH T'- CH2"--"N H - - OII - - CI- - C -I - C H ' - - O - ,

I,

O OH CH3 Pantothenic acid

O

I~-Mercaptoethylamine

I ' O-

H

P II - - O - - I - - O - - C H ' , O

N ~NtC'N~CH

O

O

[ 7

Adenine

]

-

H H

Ribose 3"-phosphate

OH

I

O--P--O-

I_

0 F I G U R E 13-7 Coenzyme A. The terminal sulfhydryl group is the reactive group of the molecule.

acetyl (and acyl groups) by formation of thioesters, and the acetyl-sulfur bond has a AG o' comparable to that of the phosphoanhydride bond of ATE Dehydrogenation and decarboxylation of pyruvate occur in five reactions requiring three enzymes and six coenzymes, prosthetic groups, and cofactors (Mg 2+, TPP, lipoic acid, CoASH, flavin adeninedinucleotide [FAD], and NAD+). The mammalian complex (M.W. 7-8.5 x 106) consists of 20 or 30 units of pyruvate dehydrogenase (depending on the source), 60 units of dihydrolipoyl transacetylase (also called dihydrolipoamide acetyltransferase), and 5 or 6 units of dihydrolipoyl dehydrogenase (also called dihydrolipoamide reductase). A kinase and a phosphatase are tightly bound to the pyruvate dehydrogenase subunit and participate in the regulation of the activity. In the first reaction, pyruvate is decarboxylated by reaction with TPP bound to pyruvate dehydrogenase (El), with formation of a hydroxyethyl group attached to the thiazole ring of TPP:

H CH3--C--TPP--E1 +

I

OH E2

H:S~ O---CI ICH

E2

H

I

CH3--C--TPP--E1 + CO2

I

OH

In the second step, also catalyzed by pyruvate dehydrogenase, the hydroxyethyl group is transferred to the oxidized form of the lipoyl-lysyl prosthetic group of dihydrolipoyl transacetylase (E2):

E2

A detailed mechanism of the first two reactions is shown in Figure 13-8. In the third step, catalyzed by dihydrolipoyl transacetylase, the acetyl group attached to E2 is transferred to CoASH, with formation of the dithiol form of the lipoyl group and acetyl-CoA.

S-- --OH 3

O II CH3--C--COO- + TPP--E1

+ E1--TPP

+co,sH . SH

O

II

SH

O

E2

+ CHaC--S--CoA AcetyI-CoA

In the fourth step, catalyzed by the same enzyme, the hydrogen atoms of the dithiol group of E2 are transferred to the FAD prosthetic group of dihydrolipoyl dehydrogenase (E3).

SH + E3__FAD SH E2

;

+ E3__FADH2 E2

CHAPTER13 Carbohydrate Metabolism I: Glycolysis and TCA Cycle

238

NH`, i 9

H3C~, .

(i)~1 TPP , .....CH~_.._CHi___.O~PmO~PmO-

N~-~'" ,"h----- CH2--"--N, H3C-~~N J

I ""-... s

~IT H

(removal of this hydrogen forms an ylide that acts as a nucleophilicreagent)

~

R~N.+

O

"~Thiazole ring R`, ~

1

O

\\ ~ .~., S ~ 8 +7 ~ H3C--C--COO ~ Pyruvate

+H+

\ /~ -R~----N~ +/ \~ yS

/ ,o+"~'Y

- R`,

H3C~ C- - C~ O -

OH I

CO,, R._._N~) s

R

~

Ri--

H3CJ'C~OH

o~-HydroxyethyI-TPP (active acetaldehyde)

H3C~ C m OH

/s _/~,s\

~.......~~o

/~

S

*

oII

..CH-- (CH2);--C--N--Enzyme ~C ~ H H= Oxidized lipoic acid

'a`,

a`,

9.

I

s

"s

I

/ CIH ~

H~CH ~

o

(CH`,)----~--N--Enzyme H

S

I

H

S

I

S

I o H`, .CH II CH"~2~(CH`,),---C--N -- Enzyme

I

H S-Acetylhydrolipoyl-enzyme F I G U R E 13-8 Steps involved in the decarboxylation and formation of the S-acetylhydrolipoyl enzyme of the dehydrogenase complex. TPP = Thiamine pyrophosphate.

The structure of FAD that contains the vitamin riboflavin is given in Figure 13-9. In the last step, catalyzed by dihydrolipoyl dehydrogenase, the hydrogens (or reducing equivalents) are transferred to NAD +, with the formation of NADH and the oxidized flavoprotein. E3-FADH2 + NAD + ~ E3-FAD + NADH + H +

Thus, the flow of reducing equivalents in the pyruvate dehydrogenase complex is from pyruvate to lipoyl to FAD to NAD +. Conversion of pyruvate to acetyl-CoA requires four vitamins: thiamine, pantothenic acid, riboflavin, and niacin. In contrast, in glycolysis, niacin is the only vitamin used. NADH generated in this reaction is oxidized to NAD + in the electron transport

SECTION13.2 Pyruvate Metabolism

239 NH=

_

_

o II

O--P-0I i ~

o

I

o--P-=-~

1

I

OH

?

OH

H~OH HCOH I

Riboflavin

H~OH

~

H2

9

10

I

~

~.NH 0

F I G U R E 13-9 Flavin adenine dinucleotide (FAD).

chain, with the production of ATP (Chapter 14). Dihydrolipoyl transacetylase plays a central role in transferring hydrogen atoms and acetyl groups from one enzyme to the next in the pyruvate dehydrogenase complex. This is made possible by the lipoyl-lysyl swinging arm of the transacetylase, which is about 1.4 nm long (Figure 13-10). In a similar way, other o~-keto acids, e.g., ot-ketoglutarate (in the TCA cycle; see below) and branched-chain o~-keto acids derived by transamination from the branched-chain amino acids valine, leucine, and isoleucine (Chapter 17), undergo decarboxylation and dehydrogenation catalyzed by enzyme complexes. These enzyme complexes differ in specificity of E1 and E2, but all contain the same E3 (the dihydrolipoyl dehydrogenase).

Regulation of Pyruvate DehydrogenaseActivity The pyruvate dehydrogenase complex catalyzes an irreversible reaction that is the entry point of pyruvate into the TCA cycle (see below) and is under complex regulation by allosteric and covalent modification of the pyruvate dehydrogenase component of the complex. The end products of the overall reaction (NADH and acetyl-CoA) are potent allosteric inhibitors of the pyruvate dehydrogenase

F I G U R E 13-10 Schematic representation of the relationship between the three enzymes of the pyruvate dehydrogenase complex. The lipoyl-lysyl moiety of the transacetylase delivers the acetyl group to CoA and the reducing equivalents to FAD. TPP = Thiamine pyrophosphate.

component of the complex. They also function as effectors in a non-cAMP-dependent reversible phosphorylation and dephosphorylation cycle of the dehydrogenase. Phosphorylation occurs by an ATP-specific pyruvate dehydrogenase kinase at three serine residues of the o~-subunit of the enzyme and leads to inactivation. The kinase is activated by elevated [acetyl-CoA]/[CoA], [NADH]/[NAD +], and [ATP]/[ADP] and inhibited by increases in [pyruvate], [Ca2+], and [K+]. The phospho enzyme is converted to the active dephospho enzyme by a pyruvate dehydrogenase phosphatase, an MgZ+/CaZ+-stimulated enzyme that is also stimulated by insulin in adipocytes. The kinase and phosphatase are associated with pyruvate dehydrogenase. The regulation of pyruvate dehydrogenase is shown in Figure 13-11. In general, when the levels of ATE NADH, or acetyl-CoA are high, the oxidation of pyruvate to acetyl-CoA is markedly decreased. For example, fatty acid oxidation (Chapter 18) provides all three metabolites and thus decreases the need for pyruvate oxidation. An analogue of pyruvate, dichloroacetate (CHClzCOO-), inhibits pyruvate dehydrogenase kinase and maintains the pyruvate dehydrogenase complex

CHAPTER 13 CarbohydrateMetabolismI: GlycolysisandTCACycle

240

[ATP] [NADH][AcetylCoA] [A~P] [NAD"].~A-]~ ,.m../Pyruvate ~P Oki~naD/ i c h l ~ 1 7 6 a2+ DephosphoPhosphopyruvate pyruvate dehydrogenase dehydrogenase (acti~~ ( i n a c t i v e ) Phospho-PDphosphatase Ca2+,~(~2+ i~nsulin , NADHj+ H+ F I G U R E 13-11 Regulation of pyruvate dehydrogenase (PD) by inactivation and reactivation by a non-cAMP-dependent phosphorylation-dephosphorylation cycle. Although PD kinase phosphorylates three specific seryl residues in the c~-subunit of PD, phosphorylation at any of these sites inactivates PD. The kinase and the phosphatase are under the influence of several regulators, and the dephospho-active PD is also regulated by end products. (~ = Activation; Q = inhibition; E2 -- dihydrolipoyl transacetylase; E3 -- dihydrolipoyl dehydrogenase.

in the active state. In experimental animals and in humans with lactic acidemia due to a variety of causes, dichloroacetate administration lowers the concentration of lactate through promotion of pyruvate oxidation. Compounds like dichloroacetate have potential applications in disorders associated with lactic acidemia (e.g., diabetes mellitus).

Abnormalities of Pyruvate Dehydrogenase Complex Thiamine deficiency causes decreased pyruvate oxidation, leading to accumulation of pyruvate and lactate, particularly in the blood and brain, and is accompanied by impairment of the cardiovascular, nervous, and gastrointestinal systems (Chapter 38). Inherited deficiency of pyruvate dehydrogenase complex is accompanied by lactic acidemia and abnormalities of the nervous system (e.g., ataxia and psychomotor retardation). Pyruvate carboxylase deficiency causes similar abnormalities (Chapter 15). Both inherited disorders of pyruvate utilization are autosomal recessive. Nervous system abnormalities may be attributed in part to diminished synthesis of neurotransmitters rather than to inadequate synthesis of ATE In pyruvate dehydrogenase complex deficiency, diminished levels of acetylCoA cause decreased production of acetylcholine; in pyruvate carboxylase deficiency, decreased production of

oxaloacetate may lead to deficiency of amino acid neurotransmitters (e.g., F-aminobutyrate, glutamate, aspartate). The interrelationships of these amino acids are discussed in Chapter 17. Although no particular therapeutic method is well established in treatment of the inherited disorders of pyruvate metabolism, ketogenic diets have been beneficial in pyruvate dehydrogenase complex deficiency, since they provide the product of the deficient reaction (acetyl-CoA). Administration of large doses of thiamine may be of benefit because mutations of pyruvate dehydrogenase complex may give rise to decreased affinity for thiamine pyrophosphate. In one patient, administration of dichloroacetate was beneficial even in the absence of a ketogenic diet. In pyruvate carboxylase deficiency, administration of diets supplemented with aspartate and glutamate demonstrated sustained improvement in neurological symptoms. These two amino acids cross the blood-brain barrier after amidation to asparagine and glutamine in nonneural tissues. The toxicity of organic arsenicals and arsenite (AsO~-)is due to their abiltiy to bind functional sulfhydryl groups of enzymes. One important target is the dithiol form of the lipoyl group of pyruvate dehydrogenase and ot-ketoglutarate dehydrogenase complexes. In the earlier times arsenicals were used in the treatment of syphilis, however due to their toxicity it has been replaced with better drugs such as penicillin.

SECTION 13.3 Tricarboxylic Acid (TCA) Cycle

13.3 Tricarboxylic Acid (TCA) Cycle

241 SCoA

I

*C=O

I

The tricarboxylic acid (TCA) cycle (also called the citric acid cycle or Krebs cycle) consists of eight enzymes that oxidize acetyl-CoA with the formation of carbon dioxide, reducing equivalents in the form of NADH and FADH2, and guanosine triphiosphate (GTP): CH3COSCoA + 3NAD + + FAD + GDP 3- + HPO 2+ 2H20 --+ CoASH + 2CO2 + 3NADH + FADH2 + GTP 4- + 2H + The reducing equivalents are transferred to the electron transport chain, ultimately to reduce oxygen and generate ATP through oxidative phosphorylation (Chapter 14). Thus, oxidation of acetyl-CoA yields a large supply of ATE Since the intermediates in the cycle are not formed or destroyed in its net operation, they may be considered to play catalytic roles. However, several intermediates are biosynthetic precursors of other metabolites (amphibolic role) and hence may become depleted. They are replenished (anaplerotic process) by other reactions to optimal concentrations. The TCA cycle is the major final common pathway of oxidation of carbohydrates, lipids, and proteins, since their oxidation yields acetyl-CoA. AcetylCoA also serves as precursor in the synthesis of fatty acids, cholesterol, and ketone bodies. All enzymes of the cycle are located in the mitochondrial matrix except for succinate dehydrogenase, which is embedded in the inner mitochondrial membrane. Thus, the reducing equivalents generated in the cycle have easy access to the electron transport chain. TCA cycle enzymes, with the exception of ot-ketoglutarate dehydrogenase complex and succinate dehydrogenase, are also present outside the mitochondria. The overall TCA cycle is shown in Figure 13-12.

~CH 3

AcetyI-CoA

tl

o

II

H2o

+ C--COO-

L

I

+

, HO--C--COO-

I CH 2--0OO

-

OH 2__000

Oxaloacetate

-

Citrate

The condensation reaction is thought to yield a transient enzyme-bound intermediate, citroyl-CoA, which undergoes hydrolysis to citrate and CoASH with loss of free energy. This reaction is practically irreversible and has a AG o' of - 7 . 7 kcal/mol ( - 3 2 . 2 kJ/mol). Formation of citrate is the committed step of the cycle and is regulated by allosteric effectors. Depending on the cell type, succinylCoA (a later intermediate of the cycle), NADH, ATE or long-chain fatty acyl-CoA functions as the negative allosteric modulator of citrate synthase. Citrate formation is also regulated by availability of substrates, and citrate is an allosteric inhibitor. Citrate provides the precursors (acetyl-CoA, NADPH) for fatty acid synthesis and is a positive allosteric modulator of acetyl-CoA carboxylase, which is involved in the initiation of long-chain fatty acid synthesis (Chapter 18). It regulates glycolysis by negative modulation of 6-phosphofructokinase activity (see above). All of the above reactions occur in the cytoplasm, and citrate exits from mitochondria via the tricarboxylate carrier. Isomerization of Citrate to Isocitrate

In this reaction, the tertiary alcoholic group of citrate is converted to a secondary alcoholic group that is more readily oxidized. The isomerization catalyzed by aconitate dehydratase (or aconitase) occurs by removal and addition of water with the formation of an intermediate, cis-aconitate: CH2_CO O-

I

HO--C--COOCH2__CO O-

Citrate

"CH2--COO-" H20

)

. CI- - C O 0 II

H20 k._

.

CH--COOcis-Aconitate

Condensation of Acetyl-CoA with Oxaloacetate to

CH2--COO-

Form Citrate

CH--COO-

The first reaction of the TCA cycle is catalyzed by citrate synthase and involves a carbanion formed at the methyl group of acetyl-CoA that undergoes aldol condensation with the carbonyl carbon atom of the oxaloacetate:

CoASH+ H +

I

I

Reactions of T C A Cycle

CH2--~OO-

I I HO--CH--CO0Isocitrate

The cis-aconitate may not be an obligatory intermediate, since the enzyme presumably can isomerize

~2

CHAPTER13 Carbohydrate Metabolism I: Glycolysis and TCA Cycle

F I G U R E 13-12 Reaction of the tricarboxylic acid (TCA) cycle.

citrate to isocitrate directly. Aconitate dehydratase has an iron-sulfur center, the exact function of which is not known. The reaction is considered to be near-equilibrium, even though under physiological conditions the ratio of citrate to isocitrate is about 9:1 owing to rapid conversion of isocitrate to subsequent products of the cycle. Although citrate is a symmetrical molecule, its two -CHzCOOH groups are not identical with regard to the orientation of t h e - O H a n d - C O O H groups. Thus, citrate reacts in an asymmetrical manner with the enzyme, so that only t h e - C H z C O O H group derived from oxaloacetate is modified. A three-point attachment of the enzyme to the substrate accounts for the asymmetrical nature of the reaction.

A powerful competitive inhibitor (highly toxic) of aconitate dehydratase is fluorocitrate, an analogue of citrate:

CHF--COO-

I

HO--CH--COO-

I CH2--CO0-

In vivo, fluorocitrate can be formed by condensation of fluoroacetyl-CoA with oxaloacetate by action of citrate synthase. Fluoroacetyl-CoA itself is synthesized from fluoroacetate by acetyl-CoA synthase.

SECTION13.3

Tricarboxylic Acid (TCA) Cycle

243

Oxidative Decarboxylation of lsocitrate

CH2_CO O-

I

to ~-Ketoglutarate Isocitrate dehydrogenase catalyzes the first of two decarboxylations and dehydrogenations in the cycle. Three different isocitrate dehydrogenases are present: one specific for NAD + and found only in mitochondria, the other two specific for NADP + and found in mitochondria and cytoplasm. The NAD+-specific enzyme is the primary enzyme with regard to TCA cycle operation. All three require Mg 2+ or Mn 2+. The reaction yields o~-ketoglutarate (2-0xoglutarate), NAD(P)H, and CO2 and involves enzyme-bound oxalosuccinate as an intermediate. CH2--COONAD +

I CH--COO

+ CoASH + NAD + - ~

CH2

NADH + H +

-

I HO--CH--COO-

Isocitrate

C H 2__CO O -

I CH--COO-

I O--C--COO -

Oxalosuccinate (enzyme-bound)

H 2C__CO O -

~

I ;

CH2

I O - - C-- C O O -

0r (2-oxoglutarate)

The reaction is nonequilibrium in type and has a AG O' o f - 5 . 0 kcal/mol ( - 2 0 . 9 kJ/mol). Although in mitochondria both NAD +- and NADP+-linked enzymes are involved in the cycle, the NAD+-linked enzyme, which is also under allosteric regulation, is the more predominant. Positive effectors are ADP and Ca 2+, and negative effectors are ATP and NADH. Thus, under conditions of abundance of energy the enzyme is inhibited, and under conditions of low energy the enzyme is stimulated.

Oxidative Decarboxylation of ~-Ketoglutarate to Succinyl-CoA The o~-ketoglutarate dehydrogenase complex is analogous to the pyruvate dehydrogenase complex and consists of three enzymes: o~-ketoglutarate dehydrogenase, dihydrolipoyl transsuccinylase, and dihydrolipoyl dehydrogenase (the latter dehydrogenase is identical in both complexes). The cofactor, prosthetic groups, and coenzyme requirements are identical to those of pyruvate oxidation.The overall reaction is

I O - - - C - - C O O

-

=-Ketoglutarate CH2--COO+ NADH + H +

I

CH2CO--SCoA

The reaction is of the nonequilibrium type, with a AG O' o f - 8 kcal/mol (-33.5 kJ/mol). Unlike the pyruvate dehydrogenase complex, the ot-ketoglutarate dehydrogenase complex does not possess a complex regulatory mechanism involving a kinase and a phosphatase. However, activity is inhibited by high ratios of [ATP]/[ADP], [succinyl-CoA]/[CoASH], and [NADH]/[NAD +] and stimulated by Ca 2+. ot-Ketoglutarate is reversibly transaminated to glutamate and is utilized in the hydroxylation of prolyl or lysyl residues of collagen (Chapter 25).

Conversion of Succinyl-CoA to Succinate Coupled to Formation of GTP In this complex reaction catalyzed by succinylCoA synthase (succinate thiokinase), the energy-rich thioester linkage of succinyl-CoA is hydrolyzed with release of free energy that is conserved in the substrate phosphorylation of GDP with phosphate ~to form GTP: CH2COO-

I

+ G DP3- + Pi2-

CH2CO-SCoA SuccinyI-CoA CH2COOI + GTP 4- + CoASH CH2COO-

Succinate

This reaction is readily reversible (a near-equilibrium reaction) and has a AG o' o f - 0 . 7 kcal/mol ( - 2 . 9 kJ/mol). It proceeds with the formation of intermediates of the enzyme with succinyl phosphate and with phosphate (Pi), the latter being linked to histidyl residue of the enzyme (E): E + succinyl-CoA + p2+ 9

-N-----

E-succinyl phosphate + CoASH E-succinyl phosphate ~-- E-phosphate + succinate E-phosphate + GDP ~- E + GTP

244

CHAPTER 13

GTP is converted to ATP by nucleoside-diphosphate kinase: GTP + ADP ~- GDP + ATP Succinyl-CoA can also be synthesized from propionylCoA by way of methylmalonyl-CoA, which is formed in the oxidation of branched-chain amino acids (e.g., valine, isoleucine) and in the terminal stage of oxidation of oddchain-length fatty acids (Chapter 18). Succinyl-CoA is utilized in the activation of acetoacetate (Chapter 18) and the formation of 6-aminolevulinate, a precursor of prophyrin (Chapter 29).

Dehydrogenation of Succinate to Fumarate Succinate is dehydrogenated to the trans-unsaturated compound fumarate by succinate dehydrogenase, an FAD enzyme: C H 2COO CH2COO-

HCCOO + E--FADH 2 , II -OOCCH

Succinate

Fumarate

I

+ E--FAD

This reaction is the only dehydrogenation reaction of the TCA cycle in which NAD + does not mediate the transport of reducing equivalents. The FAD is covalently linked to a histidyl residue of the enzyme, which is an integral protein of the inner mitochondrial membrane and contains iron-sulfur centers that undergo redox changes (Fe3++ e- ~ FEZ+). Reducing equivalents from FADH2 enter the electron transport chain at the coenzyme Q level, bypassing one of the sites of oxidative phosphorylation (Chapter 14). The enzyme is stereospecific for the trans hydrogen atoms of the methylene carbons of the substrate, so that only the trans isomer is produced (the cis isomer is maleate). Succinate dehydrogenase is competitively inhibited by malonate, the next lower homologue of succinate (Chapter 6).

Hydration of Fumarate to Malate In this reaction, water is added across the double bond of fumarate by fumarate hydratase (fumarase) to yield the hydroxy compound L-malate: HC--COO-

II

-OOC--CH Fumarate

HO--CH--COO+

H20 ~

I

CH2--COOH L-Malate

The enzyme has absolute specificity for the double bond of the trans-unsaturated acid and for the formation of

Carbohydrate Metabolism I: Glycolysis and TCA Cycle

L-hydroxy acid. The reaction is freely reversible (nearequilibrium).

Dehydrogenation of Malate to Oxaloacetate In the last reaction of the cycle, L-malate is oxidized to oxaloacetate by malate dehydrogenase, an NAD+-linked enzyme: HO--CHCOO-

I

+ NAD +

CH2--COOL-Malate

o II

C--COO-

+ NADH + H +

l

CH2--COO-

Oxaloacetate

Although the equilibrium of this reaction favors malate formation, in vivo the reaction proceeds toward the formation of oxaloacetate, since the latter is rapidly removed by the citrate synthase reaction to initiate the next round of the cycle.

Stereochemical Aspects of the TCA Cycle Aconitate dehydratase, succinate dehydrogenase, and fumarase yield stereospecific products. Labeling experiments with 14C in methyl and carboxyl carbons of acetylCoA or in all of the carbons of oxaloacetate yield the following results in terms of the intermediates or product formed. When only labeled acetyl-CoA is used, the label does not appear in CO2 during the first revolution of the cycle, and upon completion of the first revolution, two of the four carbon atoms of oxaloacetate are labeled. Thus, during the first revolution, neither carbon atom of acetylCoA is oxidized to CO2 and the carbon atoms of the CO2 produced are derived from oxaloacetate. This finding is due to discrimination of the two -CHzCOO- groups of citrate by aconitate dehydratase, so that citrate is treated as a chiral molecule at the asymmetrical active center of the enzyme. However, during the succeeding revolutions of the cycle, the label is randomized because succinate, a symmetrical compound, is treated as such by the enzyme without discriminating between its two carboxyl groups. If only labeled oxaloacetate is used, half of the label is retained in the oxaloacetate at the end of the first revolution of the cycle.

Amphibolic Aspects of the TCA Cycle At each turn of the TCA cycle, oxaloacetate is regenerated and can combine with another acetyl-CoA

245

Supplemental Readings and References

molecule. The TCA cycle is amphibolic; i.e., it serves as a catabolic and an anabolic pathway. Reactions that utilize intermediates of the cycle as precursors for the biosynthesis of other molecules are listed below. Some of these reactions occur outside the mitochondria. 1. Citrate + ATP + CoA -+ acetyl-CoA + oxaloacetate + ADP + Pi. This reaction takes place in the cytoplasm and is a source of acetyl-CoA for fatty acid biosynthesis. 2. ot-Ketoglutarate + alanine ~- glutamate + pyruvate. 3. ot-Ketoglutarate --+ succinate + CO2. This reaction is involved in the hydroxylation of prolyl and lysyl residues of protocollagen, a step in the synthesis of collagen. 4. Succinyl-CoA + glycine --+ 6-aminolevulnic acid (ALA). ALA is then utilized for the synthesis of heme. 5. Succinyl-CoA + acetoacetate --+ acetoacetyl-CoA + succinate. This reaction is important in the activation of acetoacetate (a ketone body) and hence for its utilization in extrahepatic tissues. 6. Oxaloacetate + alanine ~ aspartate + pyruvate. Mg 2+

7. Oxaloacetate + GTP GDP § CO2.

> phosphoenolpyruvate +

Utilization of intermediates in these reactions leads to their depletion and hence to a slowdown of the cycle unless they are replenished by the replacement or anaplerotic reactions listed below: biotin, Mg 2+

1. Pyruvate § C O 2 § ATP > oxaloacetate § ADP + Pi. This reaction, catalyzed by pyruvate carboxylase, is the most important anaplerotic reaction in animal tissues and occurs in mitochondria. Pyruvate carboxylase is an allosteric enzyme that requires acetyl-CoA for activity (see gluconeogenesis; Chapter 15). 2. Pyruvate § C O 2 § NADPH § H + ~- L-malate § NADP + . 3. Oxaloacetate and o~-ketoglutarate may also be obtained from aspartate and glutamate, respectively, by transaminase reactions. 4. Oxaloacetate obtained in the reverse of reaction (7), above.

Regulation of the TCA Cycle TCA cycle substrates oxaloacetate and acetyl-CoA and the product NADH are the critical regulators. The availability of acetyl-CoA is regulated by pyruvate dehydrogenase complex. The TCA cycle enzymes citrate synthase,

isocitrate dehydrogenase and the ot-ketoglutarate dehydrogenase complex are under regulation by many metabolites (discussed above) to maintain optimal cellular energy needs. Overall fuel homeostasis is discussed in Chapter 22.

Energetics of the TCA Cycle Oxidation of one molecule of acetyl-CoA in the cycle yields three NADH, one FADH2, and one GTP. Transport of reducing equivalents in the electron transport chain from one NADH molecule yields three ATP and from FADH2, two ATP (see Chapter 14). Thus, oxidation of one molecule of acetyl-CoA yields 12 ATE Since one glucose molecule yields two acetyl-CoA, the yield of ATP is 24. In addition, in the oxidation of glucose to acetyl-CoA, two other steps yield four NADH (i.e., glyceraldehyde3-phosphate dehydrogenase and pyruvate dehydrogenase reactions), since one glucose molecule yields two triose phosphate molecules, which accounts for 12 more ATE Oxidation of one molecule of glucose to two of pyruvate yields two ATP (see under glycolysis) by substratelevel phosphorylation. Thus, complete oxidation of one molecule of glucose can yield 38 ATE In cells dependent only on anaerobic glycolysis, glucoseconsumption has to be considerably greater to derive an amount of energy equal to that obtainable from aerobic oxidation (two ATP versus 38 ATP per glucose). However, in cells capable of both aerobic and anaerobic metabolism, glycolysis, the TCA cycle, and oxidation in the electron transport chain are integrated and regulated so that only just enough substrate is oxidized to satisfy the energy needs of a particular cell. In the presence of oxygen, a reduction in glucose utilization and lactate production takes place, a phenomenon known as the Pasteur effect. The depression of rate of flux through glycolysis can be explained in part by the accumulation of allosteric inhibitors (e.g., ATP) of 6-phosphofructokinase and pyruvate kinase, together with the supply of cofactors and coenzymes of certain key enzymes (e.g., glyceraldehyde-3-phosphate dehydrogenase).

Supplemental Readings and References D. C. De Vivo, R. R. Trifiletti, R. I. Jacobson, et al.: Defective glucose transport across the blood-brain barrier as a cause of persistent hypoglycorrhachia, seizures and developmental delay. New England Journal of Medicine 325, 703 (1991). O. N. Elpeleg, W. Ruitenbeek, C. Jakobs, et al.: Congenital lacticacidemia caused by lipoamide dehydrogenase defciency with favorable outcome. Journal of Pediatrics 126, 72 (1995). R. A. Fishman: The glucose-transporter protein and glucopenic brain injury. New England Journal of Medicine 325, 731 (1991).

246 R. S. Hotchkiss and I. E. Karl: Reevaluation of the role of cellular hypoxia and bioenergetic failure in sepsis. Journal of the American Medical Association 267, 1503 (1992). S. J. Hunter and T. Garvey: Insulin action and insulin resistance: Diseases involving defects in insulin receptors, signal transduction, glucose transport effector system. American Journal of Medicine 105, 331 (1998). M. A. Magnuson: Glucokinase gene structure. Diabetes 39, 523 (1990). A. Mattevi, M. Bolognesi, and G. Valentini: The allosteric regulation of pyruvate kinase. FEBS Letters 389, 15 (1996). M. A. Permutt, K. C. Chiu, and Y. Tanizawa: Glucokinase and NIDDM. Diabetes 41, 1367 (1992).

CHAPTER13 Carbohydrate Metabolism I' Glycolysis and TCA Cycle S. J. Pilkis, I. T. Weber, R. W. Harrison, and G. I. Bell: Glucokinase: Structural analysis of a protein involved in susceptibility to diabetes. Journal of Biological Chemistry 269, 2192 (1994). B. H. Robinson: Lactic acidemia. In: The Metabolic and Molecular Bases of Inherited Disease, 7th ed. C. R. Scriver, A. L. Bendet, and S. Sly (Eds). McGraw-Hill, New York, 1995, p. 1479. K. R. Tanaka and D. E. Paglia: Pyruvate kinase and other enzymopathies of the erythrocyte. In: The Metabolic and Molecular Bases of Inherited Disease, 7th ed. C. R. Scriver, A. L. Bendet, and S. Sly (Eds). McGraw-Hill, New York, 1995, p. 3485. J. Uribarri, M. S. Oh, and H. J. Carroll: D-Lactic acidosis. Medicine 77, 73 (1998).

CHAPTER

14

Electron Transport and Oxidative Phosphorylation

The energy requirements of aerobic cells are met by the energy released in the oxidation of carbohydrates, fatty acids, and amino acids by molecular oxygen. In these oxidation processes, the reducing equivalents from substrates are transferred to NAD +, flavin mononucleotide (FMN), or flavin adenine dinucleotide (FAD). The hydrogens, electrons, or hydride (H-) ions are removed catalytically from NADH and reduced flavin nucleotides and transferred through a series of coupled reductionoxidation (redox) reactions to oxygen, which serves as the ultimate electron acceptor. The reduced oxygen is then converted to water. The entire process is known as cell respiration. The numerous coupled redox reactions are tightly linked and take place in the inner mitochondrial membrane. This reaction sequence is called the respiratory chain electron transport. Electrons in substrate or cofactor begin with a high potential energy and end at oxygen with a lower potential energy. During this electron flow, a portion of the free energy liberated is conserved by an energy-transducing system (by which electrical energy is changed to chemical energy). Since the energy is conserved in the terminal phosphoanhydride bond of ATP through phosphorylation of ADP to ATE the overall coupled process is known as oxidative

phosphorylation. The following reactions yield NADH or FADH2 during the breakdown of glucose by glycolysis and in the

TCA cycle (Chapter 13). The NADH-producing reactions are Glyceraldehyde 3-phosphate --+ 1,3-bisphosphoglycerate Pyruvate --+ acetyl-CoA Isocitrate --+ ot-ketoglutarate ot-Ketoglutarate --+ succinyl-CoA Malate --+ oxaloacetate The FADHz-producing reaction is Succinate --+ fumarate Except for the conversion of glyceraldehyde 3-phosphate to 1,3-bisphosphoglycerate occurring in the cytoplasm, all of the above reactions take place in the mitochondria. Since mitochondria are not permeable to NADH, two shuttle pathways transport the reducing equivalents of cytoplasmic NADH into the mitochondria. The TCA cycle functions for both the oxidation and the production of reducing equivalents from a variety of metabolites such as carbohydrates, amino acids, and fatty acids. In the mitochondria fatty acids undergo successive removal of two carbon units in the form of acetylCoA, which is converted in the TCA cycle to CO2 and reducing equivalents found in NADH and FADH2. The latter two substances are then fed into the electron transport chain. The formation of each acetyl-CoA molecule

247

248

CHAPTER14 Electron Transport and Oxidative Phosphorylation

requires the removal of four H atoms and is catalyzed by two dehydrogenases (Chapter 18). One dehydrogenase yields FADH2; the other yields NADH. The term "one reducing equivalent" means that 1 mol of electrons is present in the form or one gram-equivalent of a reduced electron carrier. One mole of NAD +, when reduced to 1 mol of NADH, utilizes 2 mol of electrons from 1 mol of substrate (Chapter 13):

Inner membrane Intermembrane

~

space

~

Matrix

Cristae

~f

Outer membrane

NAD + + SH2 (reduced substrate) ~NADH + H + + S (oxidized substrate) In this reaction, the 2 mol of electrons is transferred in the form of 1 mol of hydride ion (H:)-.

14.1 Mitochondrial Structure and Properties Mitochondria are present in the cytoplasm of aerobic eukaryotic cells. They are frequently found in close proximity to the fuel sources and to the structures that require ATP for maintenance and functional activity (e.g., the contractile mechanisms, energy-dependent transport systems, and secretory processes). The number of mitochondria in a single cell varies from one type of cell to another; a rat liver cell contains about 1000, while one giant amoeba has about 10,000. In a given cell, the number of mitochondria may also depend on the cell's stage of development or functional activity. The size and shape of mitochondria vary considerably from one cell type to another. Even within the same cell, mitochondria can undergo changes in volume and shape depending on the metabolic state of the cell. In general, they are 0.5-1.0 # m wide and 2-3 # m long and are known to aggregate end to end, forming long filamentous structures. Mitochondria consist of two membranes, one encircling the other, creating two spatial regions: the intermembrane space and the central space, called the matrix. The outer membrane is 6-7 nm thick, smooth, unfolded (Figure 14-1), and freely permeable to molecules with molecular weights below 10,000. It contains a heterogeneous group of enzymes that catalyze certain reactions of lipid metabolism as well as hydroxylation reactions (Table 14-1). The intermembrane space (5-10 nm) contains the enzymes that catalyze interconversion of adenine nucleotides. The inner membrane (6-8 nm thick) has many folds directed toward the matrix. These invaginations, known as cristae, increase the surface area of the inner membrane. The lipid component, almost all of which is phospholipid, constitutes 30-35% by weight of the inner membrane.

~~ - ~ 1 I ~

Inner

II~-.~ ~ ~ n~embra~e sphere I ~-------~-In;emrbran e

Intermembrane

membrane F I ( ; U R E 14-1 Morphology of a mitochondrion (transverse section).

Phospholipids are asymmetrically distributed in the lipid bilayer, with phosphatidylethanolamine predominating on the matrix side and phosphatidylcholine on the cytoplasmic side. Seventy-five percent of the cardiolipin is present on the matrix side of the membrane. The fatty acid composition of the phospholipids depends on the species, tissue, and diet. In all cases, sufficient unsaturated fatty acids are contained in the phospholipids to provide a highly fluid membrane at physiological temperatures. The inner membrane is studded with spheres, each 8-10 nm in diameter, that are attached via stalks 4-5 nm in length. These inner membrane spheres are present on the matrix side (M-side) but absent from the cytoplasmic side (C-side). The components of the inner membrane include respiratory chain proteins, a variety of transport molecules, and a part of the ATP-synthesizing apparatus (the base piece of ATP synthase). The ATP-ADP translocase and

SECTION 14.1

Mitochondrial Structure and Properties

249

TABLE 14-1

Distribution of Enzymes in Mitochondrial Compartments Outer Membrane

Intermembrane Space

NADH cytochrome b 5 reductase Cytochrome b 5 Monoamine oxidase Kynurenine hydroxylase Glycerolphosphate acyltransferase Lysophosphatidyl transferase Phosphatidate phosphatase Phospholipase A Nucleoside diphosphokinase Fatty acid elongation enzyme system

Adenylate kinase (myokinase) Nucleoside diphosphokinase Nucleoside monophosphokinase Sulfite oxidase

Inner Membrane

Matrix

NADH-coenzyme Q reductase Succinate-coenzyme Q reductase Coenzyme QH2_ cytochrome c reductase Cytochrome oxidase Oligomycin-sensitive ATPase /3-Hydroxybutyrate dehydrogenase Pyridine nucleotide transhydrogenase Carnitine palmitoyltransferase Ferrochelatase (heine synthase) Adenine nucleotide carrier and other carrier proteins Carbamoyl phosphate synthetase I Dihydroorotate dehydrogenase (C-side) Glycine cleavage enzyme complex

Pyruvate dehydrogenase complex c~-Ketoglutarate dehydrogenase complex Citrate synthase Aconitase Malate dehydrogenase Isocitrate dehydrogenase [NAD +] Isocitrate dehydrogenase [NADP +] Fumarase Glutamate dehydrogenase Pyruvate carboxylase Aspartate aminotransferase Ornithine carbamoyltransferase Fatty acyl-CoA synthetase Fatty acyl-CoQ dehydrogenase Enoyl hydrase /3-Hydroxyacyl-CoA dehydrogenase /3-Ketoacyl-CoA thiolase Amino acid activating enzymes RNA polymerase DNA polymerase 6-Aminolevulinic acid synthase HMG-CoA synthase HMG-CoA lyase Acetyl-CoA acetyltransferase /3-Ketoacid CoA-transferase Rhodanese (detected in both inner membrane and matrix) *Coproporphyrinogen oxidase *Protoporphyrinogen oxidase

*The exactlocation is not yet known.

the ATP synthase together make up at least two thirds of all protein within the inner mitochondrial membrane. Small uncharged molecules (e.g., water, oxygen, carbon dioxide, ammonia, and ethanol) can diffuse through the inner membrane, but all other molecules that pass through require specific transport systems. The mitochondrial outer and inner membranes are vastly different in their constituents and function:

1. The outer membrane contains two to three times more phospholipids per unit of protein; 2. Cardiolipin is localized in the inner membrane; and 3. Cholesterol is found predominantly in the outer membrane. The permeability of the outer membrane to charged or uncharged substances up to a molecular weight of 10,000

250

CHAPTER14 Electron Transport and Oxidative Phosphorylation

F I G U R E 14-2 Formation of submitochondrial particles. These resealed inside-out inner membrane vesicles are formed when mitochondria are subjected to a variety of disruptive forces; they can support both reducing-equivalent transport and phosphorylation of ADP. Removal of the inner membrane spheres from the vesicles leads to the loss of ability to phosphorylate ADP.

is due to pore-like structures that consist of a protein (M.W. 30,000) embedded in the phospholipid bilayer. In vitro, addition of the purified pore protein renders phospholipid bilayers permeable to a variety of substances. An analogous pore protein, known as porin, has been identified in the outer membrane of gram-negative bacteria (Chapter 11). The matrix is viscous and contains all TCA cycle enzymes except succinate dehydrogenase, which is a component of electron transport complex II and is located within

the inner membrane. Enzymes of the matrix or the inner mitochondrial membrane mediate reactions of fatty acid oxidation, ketone body formation and oxidation, and biosynthesis of urea, heme, pyrimidines, DNA, RNA, and protein. Mitochondria are involved in programmed cell death apoptosis, (see Chapter 26). The structure, mechanism of replication of mitochondrial DNA, and processes of transcription and translation are unique in several respects (discussed later).

SECTION 14.1 M i t o c h o n d r i a l

251

S t r u c t u r e and P r o p e r t i e s

Submitochondrial Particles

Complex l

Submitochondrial particles (SMPs) are produced by disruption of the mitochondria by mechanical, osmotic, or sonic shock treatment. The fragmentation results in the release of water-soluble components and inner membrane fragments that re-form into vesicles (Figure 14-2). The components are then separated by differential centrifugation. The membranes of SMPs have the characteristic inner membrane spheres on their outside. SMPs are capable of electron transport and oxidative phosphorylation (i.e., the synthesis of ATP from ADP and phosphate). Removal of the inner membrane spheres by further mechanical treatment, urea, or trypsin results in the dissociation of the electron transport assembly from ATP synthesis. The ability to synthesize ATP resides in the overall structure, which includes the spheres (called F1), the stalks, and a membrane protein subunit (called Fo). The Fo-subunit spans the inner membrane and thus is retained in the vesicles. The spheres removed from SMPs do not support ATP synthesis but do hydrolyze ATP to ADP and phosphate. Thus, ATP synthesis is carried out by Fo/F1-ATPase (ATP synthase). The subscript "o" in Fo indicates that it contains the site at which a potent antibiotic inhibitor, oligomycin, binds and inhibits oxidative phosphorylation. Oligomycin does not bind F1-ATPase and does not inhibit ATP hydrolysis to ADP and phosphate. FoF1-ATPase

(ATP synthase; inhibited by oligomycin)

ADP + Pi,

, ATP + H20 F1-ATPase

(Not inhibited by oligomycin)

The fully active SMPs can be reconstituted by adding F~ATPase to depleted vesicles under appropriate conditions.

The electron transport system can be reconstituted into discrete enzyme complexes that catalyze the following four reactions: NADH-coQ reductase

>

Succinate-CoQ reductase

2. Succinate + Q

> fumarate + QH2 coQ cytochrome c reductase

3. QH2 + 2ferricytochrome c 2 ferrocytochrome c + 2H +

- -

I

Complex II

--" Complex III

/

- - " Cyt a,a3-Cu

I_i O,

Complex IV

F I G U R E 14-3 Diagram of the functional complexes of the electron transport system within the respiratory chain. FNAD ----NADH dehydrogenase flavoprotein; Fs = succinate dehydrogenase flavoprotein; Fe(n.h.) = nonheme iron.

ation procedures, whereas the bonds holding unlike complexes are relatively weak and can be dissociated. The functional organization of the four complexes in the inner mitochondrial membrane is shown in Figure 14-3. In addition to these complexes, the F0/F1-ATPase, which is required for ATP synthesis, may be considered as complex V. The relative ratios of complexes I, II, III, IV, and V have been estimated to be 1:2:3:6:6. Complexes I, II, III, and IV can be combined in the presence of cytochrome c (which separates during fractionation) to form a single unit with all of the enzymatic properties of the intact electron transport system except coupled phosphorylation. The individual electron carriers of the four complexes of the respiratory chain, shown in Figure 14-4, are arranged in accordance with their redox potentials, with the transfer of electrons from NADH to oxygen associated with a potential drop of 1.12 V, and that of succinate to oxygen of 0.8 V. In the electron transport system, the electrons can be transferred as hydride ions (H:)- or as electrons (e.g., in the cytochromes).

Electron Transport Complexes Complex I catalyzes an NADH-CoQ reductase activity, and it contains the NADH dehydrogenase flavoprotein. It has two types of electron-carrying structures: FMN and several iron-sulfur centers. FMN is a tightly bound prosthetic group of the dehydrogenase enzyme, and it is reduced to FMNH2 by the two reducing equivalents derived from NADH: NADH + H + + E-FMN ~-- NAD + + E-FMNH2

>Q + cytochrome c oxidase

4. 2 Ferrocytochrome c + 2H + + 1/202 2 ferricytochrome c + H20

S u c c , r, at,

"'Q//--"

Complex I

Components of the Electron Transport Chain

1. NADH + H + + coenzyme Q (Q) NAD + + QH2

NDH - I

,,

>

Each complex can be considered as a functional unit composed of a fixed number of electron carriers. The individual components of the four complexes are firmly bonded together and are not dissociated by mild fraction-

The electrons from FMNH2 are transferred to the next electron carrier, coenzyme Q, via the iron-sulfur centers of the NADH-CoQ reductase. The iron-sulfur centers consist of iron atoms paired with an equal number of acid-labile sulfur atoms. The respiratory chain iron-sulfur clusters are of the FezS2 or Fe4S4 type. The iron atom, present as nonheme iron, undergoes oxidation-reduction cycles (Fe z+ ~ Fe 3+ + e -). In the Fe4S4 complexes, the centers

m I n

7
phosphoenolpyruvate 3- +

C02

+ GDp3-(or IDP 3-)

In this reaction, inosine triphosphate (ITP) can substitute for guanosine triphosphate (GTP), and the CO2 lost is the one fixed in the carboxylase reaction. The net result of these reactions is Pyruvate + ATP + GTP (or ITP) -+ phosphoenolpyruvate + ADP + Pin t- GDP (or IDP) Pyruvate carboxylase is a mitochondrial enzyme in animal cells, whereas PEPCK is almost exclusively mitochondrial in some species (e.g., pigeons) and cytosolic in others (e.g., rats and mice). In humans (and guinea pigs), PEPCK occurs in both mitochondria and cytosol. An interesting consequence of this species differences is that quinolinate, an inhibitor of cytoplasmic PEPCK, causes hypoglycemia in rats but is less active in humans and guinea pigs. In humans, oxaloacetate must be transported out of the mitochondrion to supply the cytosolic PEPCK. Because there is no mitochondrial carrier for oxaloacetate and its diffusion across the mitochondrial membrane is slow, it is transported as malate or asparate (Figure 15-2). The malate shuttle carries oxaloacetate and reducing equivalents, whereas the aspartate shuttle, which does not require a preliminary reduction step, depends on the availability of glutamate and ot-ketoglutarate in excess of tricarboxylic acid (TCA) cycle requirements.

277

SECTION15.1 Gluconeogenesis INNERMITOCHONDRIAL MEMBRANE MATRIX CYTOSOL [ Oxaloacetate Oxaloacetate --NADH + H+ NADH+ H+~,,{3

Cytoplasm Glucose 6-phosphat

~

\

I ~",,...~NAD+ Malate

NAD+---JI

Malate,

H20~

Malate shuttle

Oxaloacetate Glutamate~ [ 7 ot-KetoglutarateJ[ Aspartate--

/61

NAD+ Glutamate ~ ate (z-Keto- - ~ N A D H + H+ glutarate Aspartate ~ NH+

Oxal~

Glucose6-phosphatase Glucose6-phosphatase Regulatoryprotein(SP)

P~ ucose/X~-.J 4 ~7 GI

'

~~ ~

~

Endoplasmic reticulum membrane

Aspartate shuttle

F I G U R E 15-2 Shuttle pathways for transporting oxaloacetate from mitochondria into the cytosol. The shuttles are named for the molecule that actually moves across the mitochondrial membrane. 1 and 3 = malate dehydrogenase; 2 -- malate translocase; 4 and 7 = aspartate aminotransferase; 5 = glutamate dehydrogenase; 6 = aspartate translocase.

The proportion of oxaloacetate carried by each shuttle probably depends on the redox state of the cytosol. If most of the pyruvate is derived from lactate, the NADH / NAD + ratio in the cytosol is elevated. In this situation, there is no need to transport reducing equivalents out of the mitochondria, and the asparate shuttle predominates. However, if alanine is the principal source of pyruvate, no cytosolic reduction occurs, and the glyceraldehyde-phosphate dehydrogenase reaction (which requires NADH) requires transport of reducing equivalents via the malate shuttle. In species in which oxaloacetate is converted in mitochondria to phosphoenolpyruvate (which is readily transported to the cytosol, perhaps via its own carrier system), no transport of oxaloacetate or reducing equivalents is required. In pigeons, which have virtually no cytosolic PEPCK, the rate of gluconeogenesis from pyruvate is much slower than it is from lactate, since there is no source of cytosolic reducing equivalents. Phosphoenolpyruvate is converted to fructose-l,6bisphosphate by reversal of glycolysis in the cytosol via reactions that are at near-equilibrium and whose direction is dictated by substrate concentration. Conversion of fructose-1,6-bisphosphate to fructose-6-phosphate is a nonequilibrium step, catalyzed by fructose-l,6bisphosphatase: Mg +

Fructose- 1,6-bisphosphate 4- + H20

>

fructose_6_phosphate 2- + pZFructose-6-phosphate is then converted to glucose-6phosphate by reversal of another near-equilibrium reaction of glycolysis. In the last reaction in gluconeoge-

F I G U R E 15-3 Schematic representation of the hepatic microsomal glucose-6-phosphatase system. The hydrolysis of glucose phosphate to glucose and inorganic phosphate (Pi) consists of (1) glucose-6-phosphate transport protein (T1), (2) glucose-6-phosphatase with its catalytic site located on the luminal side of the membrane, (3) glucose-6-phosphatase CaZ+-binding stabilizing protein (SP) also known as regulatory protein, (4) phosphate transport protein (T2fl), (5) phosphate, pyrophosphate, and carbamoyl phosphate transport protein (T2fl), and (6) glucose transport protein (GLUT7).

nesis, glucose-6-phosphate is converted to glucose by glucose-6-phosphatase: Glucose-6-phosphate 2- + H20--+ glucose + p2Glucose-6-phosphatase is part of a multi-component integral membrane protein system (Figure 15-3) located in the endoplasmic reticulum of liver, kidney, and intestine but not of muscle or adipose tissue. This system consists of six different proteins. These are 1. Glucose-6-phosphate transport protein (T1), which transports glucose-6-phosphate into the lumen of the endoplasmic reticulum; 2. Catalytic subunit of glucose-6-phosphatase (M.W. 36,500), which hydrolyzes glucose-6-phosphate into glucose and phosphate at the luminal surface; 3. Glucose-6-phosphatase regulating protein (M.W. 21,000), which stabilizes the activity of glucose-6-phosphatase; 4. Microsomal phosphate transport protein (Tzot) which mediates the efflux of Pi, an inhibitor of the glucose-6-phosphatase, from the lumen of the endoplasmic reticulum to the cytosol; 5. Microsomal phosphate/pyrophosphate transport protein (Tzfl, M.W. 37,000), which transports phosphate, pyrophosphate, and carbamoyl phosphate, which are substrates for glucose-6-phosphatase; and 6. Microsomal glucose transport protein (GLUT7), which is a member of the family of facilitative glucose transport proteins (Chapter 13) and which transports glucose into the cytosol.

~

CHAPTER15

CarbohydrateMetabolism I1: Gluconeogenesis, Glycogen Synthesis and Breakdown, and Alternative Pathways

These actions of all four transport proteins are reversible. Phosphate transport protein Tzfl can transport pyrophosphate and carbamoyl phosphate as well as Pi, providing substrates and removing products for several other reactions (of unknown metabolic significance), which are also catalyzed by glucose-6-phosphatase. The importance of each member of the glucose-6-phosphate hydrolysis system is reflected in the occurrence of glycogen storage diseases. Deficiency of any of these five proteins leads to glycogen storage diseases (discussed later). Thus, gluconeogenesis requires the participation of enzymes of the cytosol, mitochondrion, and smooth endoplasmic reticulum, as well as of several transport systems, and it may involve more than one tissue. The complete gluconeogenic pathway, culminating in the release of glucose into the circulation, is present only in liver and kidney. Most tissues contain only some of the necessary enzymes. These "partial pathways" are probably used in glycerogenesis and in replenishing tricarboxylic acid (TCA) intermediates. Muscle can also convert lactate to glycogen, but this probably takes place only in one type of muscle fiber and only when glycogen stores are severely depleted and lactate concentrations are high, such as after heavy exercise. Under normal conditions, the liver provides 80% or more of the glucose produced in the body. During prolonged starvation, however, this proportion decreases, while that synthesized in the kidney increases to nearly half of the total, possibly in response to a need for NH3 to neutralize the metabolic acids eliminated in the urine in increased amounts (Chapter 22). Gluconeogenesis is a costly metabolic process. Conversion of two molecules of pyruvate to one of glucose consumes six high-energy phosphate bonds (4ATP + 2GTP --+ 4ADP + 2GDP + 6Pi) and results in the oxidation of two NADH molecules (Figure 15-1). In contrast, glycolytic metabolism of one molecule of glucose to two of pyruvate produces two high-energy phosphate bonds (2ADP + 2Pi ~ 2ATP) and reduces two molecules of NAD +. For gluconeogenesis to operate, the precursor supply and the energy state of the tissue must be greatly increased. Using some gluconeogenic precursors to provide energy (via glycolysis and the TCA cycle) to convert the remainder of the precursors to glucose would be inefficient, even under aerobic conditions. Usually, the catabolic signals (catecholamines, cortisol, and increase in glucagon/insulin ratio) that increase the supply of gluconeogenic precursors also favor lipolysis, which provides fatty acids to supply the necessary ATE When amino acid carbons are the principal gluconeogenic precursors, the metabolic and physiological debts are particularly large compared to those incurred

when lactate or glycerol is used. Amino acids are derived by breakdown of muscle protein, which is accompanied by a loss of electrolytes and tissue water. One kilogram of muscle contains about 150 g of protein, which can be used to form about 75 g of glucose. However, 1 kg of muscle also contains 1,200 mM of K +, 27 mM of phosphate, and 8 mM of Mg 2+, all of which must be excreted by the kidney. The ammonia released from catabolism of the amino acids is converted to urea in an energy-requiring process, and the urea further increases the osmotic load on the kidney. Renal excretion of these solutes necessitates mobilization of at least 2 L of water from other tissues in addition to the 750 mL released from the muscle, and osmotic diuresis may result from catabolism of even a relatively small amount of muscle. Finally, if fat metabolism is stimulated, as in starvation and diabetes mellitus, increased plasma levels of fatty acids and ketone bodies causes acidemia. To prevent severe acidosis, renal proton excretion is increased by increasing secretion of ammonia into the renal tubules. Excretion of nitrogen as ammonia avoids energy consumption due to urea synthesis, but it doubles the osmotic load per nitrogen excreted, causing even greater diuresis. Without a compensatory increase in water intake, profound depletion of blood volume can occur (Chapter 22). Gluconeogenic Precursors Gluconeogenic precursors include lactate, alanine, and several other amino acids, glycerol, and propionate (Chapter 22). Lactate, the end product of anaerobic glucose metabolism, is produced by most tissues of the body, particularly skin, muscle, erythrocytes, brain, and intestinal mucosa. In a normal adult, under basal conditions, these tissues produce 1,300 mM of lactate per day, and the normal serum lactate concentration is less than 1.2 mM/L. During vigorous exercise, the production of lactate can be increased several fold. Lactate is normally removed from the circulation by liver and kidney. Because of its great capacity to use lactate, liver plays an important role in the pathogenesis of lactic acidosis, which may be thought of as an imbalance between the relative rates of production and utilization of lactate (Chapter 39). Alanine, derived from muscle protein and also synthesized in the small intestine, is quantitatively the most important amino acid substrate for gluconeogenesis. It is converted to pyruvate by alanine aminotransferase (Chapter 17): B6-PO4

Alanine + ot-ketoglutarate 2- ~

> pyruvate- + glutamate-

279

SECTION15.1 Gluconeogenesis

Glycerol 3-phosphate is oxidized to dihydroxyacetone phosphate by glycerol-3-phosphate dehydrogenase:

I Cysteine Alanine Glycine Hydroxyproline

Serine

Tryptophan Glucose

I t

Pyruvate

Oxaloacetate

j / Aspartate

,,,,,...Oxaloacetate~~~__._ AcetyI-CoA

CH2OH I CHOH I

+ NAD+ -

CH2OPO3 z-

Glycerol 3-phosphate M te

Citrate

Tyrosine] PhenylalanineJ--* Fumarate

or

1

CH2OH I + NADH+ H+ C=O I CH2OPO32Dihydroxyacetone phosphate

,~

Succiny L;

/ i.,.oA~,,"

~ [ Glutamate Arginine Glutamine

r

Histidine Proline

Isoleucine Methionine Valine Threonine

FIGURE 15-4 Points of entryof aminoacidsintothe pathwayof gluconeogenesis.

where B6-PO4--pyridoxal phosphate. An amino acid is classified as ketogenic, glucogenic, or glucogenic/ ketogenic depending on whether feeding it to a starved animal increases plasma concentrations of ketone bodies (Chapter 18) or of glucose. Leucine and lysine are purely ketogenic because they are catabolized to acetyl-CoA, which cannot be used to synthesize glucose (Chapter 17). Isoleucine, phenylalanine, tyrosine, and tryptophan are both glucogenic and ketogenic, and the remaining amino acids (including alanine) are glucogenic. Entry points of the amino acids into the gluconeogenic pathway are shown in Figure 15-4. Glycerol is more reduced than the other gluconeogenic precursors, and it results primarily from triacylglycerol hydrolysis in adipose tissue. In liver and kidney, glycerol is converted to glycerol 3-phosphate by glycerol kinase: CH2OH I CHOH + ATP 4I CH2OH

Dihydroxyacetone phosphate is the entry point of glycerol into gluconeogenesis. Glycerol cannot be metabolized in adipose tissue, which lacks glycerol kinase, and the glycerol 3-phosphate required for triacylglycerol synthesis in this tissue is derived from glucose (Chapter 22). During fasting in a resting adult, about 210 mM of glycerol per day is released, most of which is converted to glucose in the liver. During stress or exercise, glycerol release is markedly increased (Chapter 22). Propionate is not a quantitatively significant gluconeogenic precursor in humans, but it is a major source of glucose in ruminants. It is derived from the catabolism of isolecucine, valine, methionine, and threonine; from ~-oxidation of odd-chain fatty acids; and from the degradation of the side chain of cholesterol. Propionate enters gluconeogenesis via the TCA cycle after conversion to succinyl-CoA (Chapter 18).

Regulation of Gluconeogenesis Gluconeogenesis is regulated by the production and release of precursors and by the activation and inactivation of key enzymes. Glucagon and glucocorticoids stimulate gluconeogenesis, whereas insulin suppresses it. See Chapter 22 for a discussion of the overall metabolic control and physiological implications of the several seemingly competing pathways.

Carboxylation of Pyruvate to Oxaloacetate

Glycerol CH2OH I CHOH + ADP3- + H+ I CH2OPO32Glycerol 3-phosphate

The pyruvate carboxylase reaction is activated by Mg 2+ and, through mass action, by an increase in either the [ATP]/[ADP] or the [pyruvate]/[oxaloacetate] ratio. It is virtually inactive in the absence of acetyl-CoA, an allosteric activator. The enzyme is allosterically inhibited by glutamate, since oxaloacetate formed in excess would flood the TCA cycle and result in a buildup of ot-ketoglutarate and glutamate. Pyruvate carboxylase

280

CHAPTER 15

Carbohydrate Metabolism I1: Gluconeogenesis, Glycogen Synthesis and Breakdown, and Alternative Pathways

activity is indirectly stimulated by the catabolic hormones glucagon (via cAMP) and epinephrine (independent of cAMP), in four ways: 1. By increasing substrate availability through stimulation of mitochondrial respiration. This decreases the intramitochondrial concentration of H + and increases the rate of pyruvate transport. The [ATP]/[ADP] ratio also rises. 2. By increasing the rate of fatty acid oxidation, which results in an increase in the mitochondrial concentration of acetyl-CoA, an allosteric activator of pyruvate carboxylase. 3. By decreasing the mitochondrial concentration of glutamate, an inhibitor of pyruvate carboxylase, through stimulation of the TCA cycle (secondary to the increase in mitochondrial acetyl-CoA) and the aspartate shuttle (secondary to the increase in cytosolic PEPCK induced by glucagon). 4. By decreasing the activity of glycolytic enzymes competing for the same substrate; in this case, by inactivating pyruvate kinase in the cytosol and pyruvate dehydrogenase in the mitochondria (via cAMP stimulation of protein kinase).

Conversion of Oxaloacetate to Phosphoenolpyruvate Short-term regulation of this reaction is accomplished by changes in the relative proportions of substrates and products. Increased concentrations of oxaloacetate and GTP (or ITP) increase the rate, and accumulation of phosphoenolpyruvate and GDP (or IDP) decreases it. The cytosolic PEPCK is also under long-term regulation by hormones. Its synthesis is increased by corticosteroids. Starvation and diabetes mellitus increase the synthesis of cytosolic PEPCK, whereas refeeding and insulin have the opposite effect. The net effect of increasing the ratio ofcytosolic PEPCK to mitochondrial PEPCK is to maintain gluconeogenesis even during periods of increased fatty acid metabolism. When gluconeogenesis is strongly stimulated, lipolysis and intramitochondrial fatty acid oxidation increase with the activity of cytosolic PEPCK. The increase in fatty acid oxidation elevates the [NADH]/[NAD +] ratio within the mitochondria, inhibiting phosphoenolpyruvate formation by channeling oxaloacetate into formation of malate or aspartate. At higher [NADH]/[NAD +] ratios, aspartate synthesis is favored because of increased formation of glutamate by mitochondrial glutamate hydrogenase: c~-Ketoglutarate + NH~- + NADH ~ glutamate + NAD+ + H20

The increase in glutamate favors transamination of oxaloacetate and limits oxaloacetate availability for phosphoenolpyruvate synthesis. When the [NADH]/[NAD +] ratio is low, malate formation occurs more readily. The cytosolic PEPCK is relatively unaffected by the mitochondrial [NADH]/[NAD +] ratio. Once malate and aspartate are transported to the cytosol and they are reconverted to oxaloacetate, cytosolic PEPCK can convert it to phosphoenolpyruvate.

Conversion of Fructose-l,6-Bisphosphate to Fructose-6-Phosphate This reaction is catalyzed by fructose-l,6-bisphosphatase (FBPase-1). This enzyme is a tetramer (M.W. 164,000) of identical subunits. It is inhibited by AMP, inorganic phosphate, and fructose-2,6-bisphosphate, all of which are allosteric activators of 6-phosphofructo1-kinase (PFK-1), the competing glycolytic enzyme (Figure 15-5). This inhibition prevents the simultaneous activation of the two enzymes and helps prevent a futile ATP cycle. However, this futile cycle apparently does operate at a low rate in mammalian muscle, and in the bumblebee it is essential for maintaining the flight muscle at 30~ Activation of the futile cycle occurs after exposure to certain anesthetic agents such as suxamethonium and/or volatile halogenated compounds. In susceptible persons, malignant hyperthermia is characterized by a hypermetabolic state and is accompanied by a catastrophic rise in body temperature (to 42~ or higher), massive increase in oxygen consumption, rhabdomyolysis, (disintegration of muscle, associated with excretion of myoglobin in the urine) and metabolic acidosis. Rhabdomyolysis is assessed by measurement of serum creatine kinase levels (Chapter 8). In susceptible persons, the hypermetabolic events can also be triggered by excessive exercise under hot conditions, infections, and neuroleptic drugs.

Fructose 6-phosphate

(~ AMP ],_ . (~ F 2,6-BPJ I-rucl~

P,

~ PFK-~[QATP IQAuP . ,6.bisphosphat ~,~ase)l /,--ATP H2OJI , " ' A D P (-_)Citrate I(.t)F 2,6-BP Fructose 1,6-bisphosphate

FIGURE 15-5 Regulation of liver6-phosphofructokinaseand fructose-1,6bisphosphatase. Thesemultimodulatedenzymescatalyzenonequilibrium reactions, the formerin glycolysisand the latterin gluconeogenesis.Note the dual action of fructose-2,6-bisphosphate (F-2,6-BP),whichactivates phosphofructokinase (PFK-1)and inactivatesfructose-l,6-bisphosphatase. The activityof F-2,6-BPis underhormonaland substrateregulation (Figure 15-6). Q = positiveeffectors; G = negativeeffectors.

281

SECTION15.1 Gluconeogenesis

The primary biochemical abnormality of malignant hyperthermia is a membrane defect that leads to a rapid and sustained rise in Ca 2+ levels in the cytosol of skeletal muscle cells. Sarcoplasmic reticulum is the main storage site of intracellular Ca 2+ and it is released into the myoplasm by excitation-contraction coupling (Chapter 21). Dantrolene, a drug useful in the treatment of malignant hyperthermia, inhibits excitation-contraction coupling. However, dantrolene does not affect neuromuscular transmission or the electrical properties of skeletal muscle. In the animal model, pigs susceptible for malignant hyperthermia exhibit a genetic defect in a Ca 2+ release channel receptor located in the sarcoplasmic reticulum. This receptor protein is named ryanodine receptor (RYR) because it binds to a plant alkaloid ryanodine and upon binding causes Ca 2+ release. The mutation that alters the receptor protein causes a substitution of cysteine for arginine at position 615. The concentration of fructose-2,6-bisphosphate is controlled by two competing enzyme activities in a single

Insulin

Glucagon

t

A ....~

+ PP~

cAMP

o,

L..

R AMP F-6-P

..,

ATP

Proteinphosphatase

/ f

_ //~,~

Cell membrane

.................p.

' cAMP-dependent protein kinase

|

ou,s,o,

Acti vatiohof adenylata cvclasa

2ATP 2ADP ~. ) ~J

|

1 /

I~

~r~-l.,.

ot-GP Citrate PEP

~

ADP

F-2,6-BP f J l 1 H=O I..

..................................

:_/~_

/o '.,,.e

PEP ot-Gp

o-.or

?o,

F-6-P P~

F I G U R E 15-6 Regulation of fructose-2,6-bisphosphate (F-2,6-BP) concentration in liver. F-2,6-BP is a major factor controlling the relative activities of fructose- 1,6-bisphosphatase (FBPase- 1) and 6-phosphofructo-l-kinase (PFK-1), key enzymes in gluconeogenesis and glycolysis, respectively. 6-Phosphofructo-2-kinase (PFK-2) and fructose-2,6-bisphosphatase (FBPase-2) are separate enzymes expressed by a single multifunctional protein. Insulin can antagonize the effect of glucagon on cAMR but the mechanism is not known. Dotted lines = regulatory factors; @ = inhibitory; @ = stimulatory. F6P = fructose-6-phosphate; PEP = phosphoenolpyruvate; ot-GP = o~-glycerol phosphate; Pi = inorganic phosphate; F-6-P -- fructose-6-phosphate.

multifunctional protein (M.W. 110,000) with two identical subunits (Figure 15-6). Phosphorylation by cAMPdependent protein kinase produces one phosphoserine per subunit, inhibiting 6-phosphofructo-2-kinase (PFK-2) activity and stimulating fructose-2,6-bisphosphatase (FBPase-2) activity. The phosphotransferase inactivation results from an increased Km for fructose-6-phosphate and a small decrease in Vmax. An increase in Vmax and a decrease in Km for fructose-2,6-bisphosphate increases the phosphohydrolase activity. The protein phosphatase that reverses the cAMP-dependent phosphorylation has not yet been characterized. The nomenclature of these enzyme activities (PFK-1, PFK-2, FBPase-1, and FBPase-2) reflects both the order of their discovery and the carbon atom of fructose that is phosphorylated or dephosphorylated. Phosphorylation is regulated by insulin and glucagon. Diabetes mellitus (in which the ratio of glucagon to insulin is increased) and glucagon therapy reduce the hepatic activity of PFK-2 and increase that of FBPase-2. The concentration of fructose-2,6-bisphosphate is thus reduced, thereby stimulating gluconeogenesis. The concentration of fructose-2,6-bisphosphate is also influenced by nonhormonal factors. PFK-2 is allosterically stimulated by inorganic phosphate, AMP, and fructose-6-phosphate and inhibited by citrate, fructose2,6-bisphosphate, phosphoenolpyruvate, and u-glycerol phosphate. FBPase-2, on the other hand, is stimulated by phosphoenolpyruvate and u-glycerol phosphate and inhibited by phosphate and fructose-6-phosphate. Glucose increases the concentration of fructose-2,6-bisphosphate in vivo, probably by increasing the availability of fructose6-phosphate, thereby stimulating PFK-2, the kinase for which this is a substrate and inhibiting the phosphatase, FBPase-2. The effect is to increase glycolysis and inhibit gluconeogenesis. The concentration of u-glycerol phosphate in vivo can be altered by ischemia consumption of ethanol, and feeding of various sugars; it may be an important regulatory molecule in determining the levels of fructose-2,6-bisphosphate. A rise in orglycerol phosphate decreases fructose-2,6-bisphosphate concentration, decreasing glycolysis and increasing gluconeogenesis.

Conversion of Glucose-6-Phosphate to Glucose Glucose-6-phosphatase and the corresponding glycolytic enzyme, glucokinase, are not controlled by the metabolites that affect PFK-1 and FBPase-1. Glucose-6-phosphatase seems to be regulated only by the concentration of glucose-6-phosphate, which is

282

CHAPTER 15

Carbohydrate Metabolism I1: Gluconeogenesis, Glycogen Synthesis and Breakdown, and Alternative Pathways

increased by glucocorticoids, thyroxine, and glucagon. The concentration of glucose-6-phosphate, which is also elevated in diabetes mellitus and following ethanol administration, is increased 300-fold after a 48-hour fast. Consumption of a low-protein diet reduces the concentration of glucose-6-phosphate. A futile cycle involving glucokinase and glucose-6-phosphatase does not occur because glucose-6-phosphatase is active only when the concentration of glucose-6-phosphate is high, and glucokinase is active only when glucose-6-phosphate concentration is low. ATP

Glucose ~ ~

yP ~ Glucose 6-phosphate,

Gluconeogenesis

P,

For extrahepatic tissue utilization

Glycogen

Glycolysis

synthesis

Pentose phosphate pathway

Abnormalities of Gluconeogenesis Glucose is the predominant fuel for cells that depend largely on anaerobic metabolism, cells that lack mitochondria, and tissues such as brain that normally cannot use other metabolic fuels. An adult brain represents only 2% of total body weight, but it oxidizes about 100 g of glucose per day, accounting for 25% of the basal metabolism. Brain cannot utilize fatty acids because they are bound to serum albumin and so do not cross the blood-brain barrier. Ketone bodies are alternative fuels, but their concentration in blood is negligible, except in prolonged fasting or diabetic ketoacidosis (Chapters 18 and 22). Brain and liver glycogen stores are small relative to the needs of the brain, and gluconeogenesis is essential for survival. Decreased gluconeogenesis leads to hypoglycemia and may cause irreversible damage to the brain. Blood glucose levels below 40 mg/dL (2.2 raM/L) in adults or below 30 mg/dL (1.7 mM/L) in neonates represent severe hypoglycemia. The low levels may result from increased glucose utilization, decreased glucose production, or both. Increased glucose utilization can occur in hyperinsulinemia secondary to an insulinoma. In a diabetic pregnancy, the mother's hyperglycemia leads to fetal hyperglycemia and causes fetal and neonatal hyperinsulinemia. Insufficiency of glucocorticoids, glucagon, or growth hormone and severe liver disease can produce hypoglycemia by depressing gluconeogenesis (Chapters 22 and 32). Ethanol consumption can cause hypoglycemia, since a major fraction of ethanol is oxidized in the liver by cytosolic alcohol dehydrogenase, and the NADH and acetaldehyde generated inhibit

gluconeogenesis: CH3CH2OH + NAD + -+ CH3CHO + NADH + H + Excessive NADH decreases the cytosolic [NAD+]/ [NADH] ratio, increasing lactate dehydrogenase activity and thereby increasing conversion of pyruvate to lactate. The decrease in pyruvate concentration inhibits pyruvate carboxylase. Acetaldehyde inhibits oxidative phosphorylation, increasing the [ADP]/[ATP] ratio, promoting glycolysis, and inhibiting gluconeogenesis. Hypoglycemia and lactic acidosis are common findings in chronic alcoholics. Pyruvate carboxylase deficiency can cause intermittent hypoglycemia, ketosis, severe psychomotor retardation, and lactic acidosis. The deficiency of either cytosolic or mitochondrial isoenzyme form of phosphoenolpyruvatecarboxykinase (PEPCK), is characterized by a failure of gluconeogenesis. Both these disorders are rare and the mitochondrial PEPCK deficiency is inherited as an autosomal recessive trait. The major clinical manifestations include hypoglycemia, lactic acidosis, hypotonia, hepatomegaly and failure to thrive. The treatment is supportive and is based upon symptoms. Fructose1,6-bisphosphatase deficiency, inherited as an autosomal recessive trait, severely impairs gluconeogenesis, causing hypoglycemia, ketosis, and lactic acidosis. Glucose6-phosphatase deficiency, also an autosomal recessive trait, causes a similar condition but with excessive deposition of glycogen in liver and kidney (discussed later). Hypoglycin A (2-methylenecyclopropylalanine), the principal toxin of the unripe akee fruit, produces severe hypoglycemia when ingested. A less toxic compound, hypoglycin B, is a y-glutamyl conjugate of hypoglycin A. The akee tree is indigenous to western Africa and grows in Central America and the Caribbean. Hypoglycin causes vomiting ("vomiting sickness") and central nervous system depression. In Jamaica, symptoms apparently occur in epidemic proportions during colder months when the akee fruit is unripe and other food sources are limited. The edible portion of the ripe akee fruit, which contains only small amounts of hypoglycin, is a main dietary staple in Jamaica. Hypoglycin A causes hypoglycemia by inhibiting gluconeogenesis. In the liver, hypoglycin A forms nonmetabolizable esters with CoA (shown below) and carnitine, depleting the CoA and carnitine pools, thereby inhibiting fatty acid oxidation (Chapter 18). Since the principal source of ATP for gluconeogenesis is mitochondrial oxidation of long-chain fatty acids, gluconeogenesis is stopped. Intravenous administration of glucose relieves the hypoglycemia.

SECTION15.2 Glycogen Metabolism OH2

/\ H2C--C

283

NH2

,

CHuCH2mCH--COOH

Hypoglycin A

=-Ketoglutarate smination Glutamate

o

II H2C~CH~CH~CH2~C~COOH Methylenecyclopropyl ~-ketopropionic acid

If'-"'~

CoASH -~l~V' Oxidative

k decarboxylation I~._~ NAOH + H + ~ CO2

o

II

H2C'--C

CHCH2C~S~CoA

Methylenecyclopropyl 0c-ketoproprionyI-CoA

Overactivity of gluconeogenesis due to increased secretion of catecholamines, cortisol, or growth hormone or an increase in the glucagon/insulin ratio (Chapter 22) leads to hyperglycemia and causes many metabolic problems.

15.2 Glycogen Metabolism Animals have developed a method of storing glucose that reduces the need to catabolize protein as a source of gluconeogenic precursors between meals. The principal storage form of glucose in mammals is glycogen. Storage of glucose as a polymer reduces the intracellular osmotic load, thereby decreasing the amount of water of hydration and increasing the energy density of the stored glucose. In muscle, glycogen is stored in the cytosol and endoplasmic reticulum as granules, called fl-particles, each of which is an individual glycogen molecule. In the liver, the fi-particles aggregate, forming larger, rosette-shaped a-particles that can be seen with the electron microscope. The average molecular weight of glycogen is several million (10,000-50,000 glucose residues per molecule). The storage granules also contain the enzymes needed for glycogen synthesis and degradation and for regulation of these two pathways. Glycogen is present in virtually every cell in the body, but it is especially abundant in liver and skeletal muscle. The amount stored in tissues varies greatly in response to metabolic and physiological demands, but in a resting individual after a meal, liver usually contains roughly 47% of its wet weight as glycogen and muscle about 1%.

Since the body contains 10 times more muscle than hepatic tissue, the total amount of glycogen stored in muscle is greater than that in liver. Muscle needs a rapidly available supply of glucose to provide fuel for anaerobic glycolysis when, as during bursts of muscle contraction, blood may provide inadequate supplies of oxygen and fuel. The liver, under most conditions, oxidizes fatty acids for this purpose; however, liver is responsible for maintaining blood glucose levels during short fasts, and it integrates the supply of available fuels with the metabolic requirements of other tissues in different physiological states. Liver glycogen content changes primarily in response to the availability of glucose and gluconeogenic precursors. Muscle glycogen stores vary less in response to dietary signals, but they depend on the rate of muscular contraction and of oxidative metabolism (tissue respiration). Glycogenesis (glycogen synthesis and storage) and glycogenolysis (glycogen breakdown) are separate metabolic pathways having only one enzyme in common, namely, phosphoglucomutase. Glycogen synthesis and breakdown are often reciprocally regulated, so that stimulation of one inhibits the other. The control mechanisms are more closely interrelated than are those for glycolysis and gluconeogenesis, perhaps because the glycogen pathways ensure the availability of only one substrate, whereas intermediates of several other metabolic pathways are produced and metabolized in glycolysis and gluconeogenesis (Chapter 13). Glycogen Synthesis Glycogenesis begins with the phosphorylation of glucose by glucokinase in liver and by hexokinase in muscle and other tissues (Chapter 13): Mg 2+

G l u c o s e + ATP 4-

> g l u c o s e - 6 - p h o s p h a t e 2- + A D P 3- + H +

The second step in glycogenesis is conversion of glucose6-phosphate to glucose-1-phosphate by phosphoglucomutase in a reaction similar to that catalyzed by phosphoglyceromutase. The phosphoryl group of the enzyme participates in this reversible reaction in which glucose-1,6bisphosphate serves as an intermediate: E-P at- g l u c o s e - 6 - p h o s p h a t e E + g l u c o s e - l , 6 - b i s p h o s p h a t e ~ E-P + g l u c o s e - l - p h o s p h a t e

In the third step, glucose-1-phosphate is converted to uridine diphosphate (UDP) glucose, the immediate precursor of glycogen synthesis, by reaction with uridine triphosphate (UTP). This reaction is catalyzed by glucose-lphosphate uridylyltransferase (or UDP-glucose pyrophosphorylase):

~4

CHAPTER15 Carbohydrate Metabolism I1: Gluconeogenesis, Glycogen Synthesis and Breakdown, and Alternative Pathways

O CH2OH . ,F---o. UTP4-+ Glucose 1-phosphate 2- ,------

H~)~

uo

HN~I

y

Jf L--O--P--O--P~O--CH2

l

+ PPi 4-

-0,

ON ON

The reaction is freely reversible, but it is rendered practically irreversible by hydrolysis of pyrophosphate (PPi) through the action of pyrophosphatase. PPi + H20--+ 2Pi Thus, two high-energy phosphate bonds are hydrolyzed in the formation of UDP-glucose. Nucleoside diphosphate sugars are commonly used as intermediates in carbohydrate condensation reactions. ADP-glucose is the precursor for the synthesis of starch in plants and of other storage polysaccharides in bacteria. In each case, the high-energy bond between the sugar and the nucleoside diphosphate provides the energy needed to link the new sugar to the nonreducing component of another sugar or polysaccharide. The next step in the biogenesis of glycogen is addition of a glucosyl residue at the C-1 position of

UDP-glucose to a tyrosyl group located at position 194 of the enzyme glycogenin, which is a MgZ+-dependent autocatalytic reaction. Glycogenin (M.W. 37,000) extends the glucan chain, again by autocatalysis, by six to seven glucosyl units in or(1 --+ 4) glycosidic linkages using UDPglucose as substrate (Figure 15-7). The glucan primer of glycogenin is elongated by glycogen synthase using UDP-glucose. Initially, the primer glycogenin and glycogen synthase are firmly bound in a 1:1 complex. As the glucan chain grows, glycogen synthase dissociates from glycogenin. The branching of the glycogen is accomplished by transferring a minimum of six or(1 ~ 4) glucan units from the elongated external chain into the same or a neighboring chain by or(1 --+ 6) linkage. This reaction of c~(1 --+ 4) to or(1 --+ 6) transglucosylation creates new non-reducing ends and is catalyzed by a branching enzyme. A mature glycogen particle is spherical, containing one molecule of

lycogenin~-- Tyrosyl Group

Mg 2+ Autocatalysis

(.--UDP-glucose "----~ UDP ~~)f-Glucosyl Unit

Autocatalysis

~ ~UDP-glucose

Glycogen Synthase

I (--UDP-glucose ""---~UDP

Branching Enzyme i

Glycogen Synthase and Branching Enzyme

~' ~ cUDP-glucose "----~ UDP Mature Glycogen (60,000 Glucosyl Units) (I] particle)

FIGURE 15-7 Schematic representation of glycogen biosynthesis. ~, Glucosyl units, (1 ~ 6) linkage. A

285

SECTION15.2 Glycogen Metabolism

C

~~~)/~

a(1 ~ 6) bonds;all other linka~s are a(1 ~ 4)

Glycogen

C~% ~

Inaccessible to

Debranching enzyme

( ~

(al,4 -, al,4 glu.can

.204

16 G-1-P

~i

(~x_

/]

Debranching enzyme

(amy.lo-l,6-glucosidase)

Glycogen

phosphorylase 16P. I

Glucose

Glycogen phosphorylase

Debranching

enzyme

H20

10 G-1-P Debranching enzyme

F I G U R E 15-8 Catabolism of a small glycogen molecule by glycogen phosphorylase and debranching enzyme. Pi = inorganic phosphate; G-1-P -- glucose-l-phosphate.

glycogenin and up to 60,000 glucosyl units (/3-particles). In the liver, 20-40/3-particles are aggregated into rosettes, known as (or-particles). Glycogen Breakdown Glycogenolysis is catalyzed by two enzymes unique to the pathway: glycogen phosphorylase and debranching enzyme. The former normally regulates the rate of glucose release from glycogen. The progressive degradation of glycogen is illustrated in Figure 15-8. Glycogen phosphorylase catalyzes the release of glucose-l-phosphate from the terminal residue of a nonreducing end of a glyco-

gen branch, by means of phosphorolysis. A molecule of inorganic phosphate attacks the C1 side of an or(1 --+ 4) glycosidic bond, leaving a hydroxyl group on C4 that remains in the glycogen polymer. The reaction is analogous to hydrolysis, in which water attacks and cleaves bonds. The phosphorolysis reaction is summarized below. (Glucose)n + Pi --+ (glucose)n_l 4-glucose-l-Phosphate The energy stored in the or(1 --+ 4) glycosidic bond during the condensation reaction in glycogen synthesis is sufficient to permit the formation of a glucose-phosphate bond without using ATE

~6

CHAPTER15

Carbohydrate Metabolism I1: Gluconeogenesis, Glycogen Synthesis and Breakdown, and Alternative Pathways

Glucose-1-phosphate is next converted by phosphoglucomutase to glucose-6-phosphate. The latter may then enter the glycolytic pathway, but if glucose-6-phosphatase is present, free glucose can be formed. Glycogen phosphorylase sequentially removes the glucosyl residues from a glycogen branch until further action is sterically hindered by a branch point. This occurs when the branch is four residues long from the branch point. Debranching enzyme, a multifunctional protein, first removes the trisaccharide "stump" on the branch and then removes the branch point itself. The or(1 --+ 4) glycosidic bond linking the trisaccharide to the branch point residue is first cleaved, and the trisaccharide is transferred to the nonreducing end of an adjacent branch. This elongated branch can now be cleaved, one residue at a time, by glycogen phosphorylase. The glucose residue that remains, linked by an or(1 --+ 6) glycosidic bond, is then cleaved by hydrolysis to yield free glucose. Thus, one molecule of glucose is released for each branch point removed, even in muscle, which lacks glucose-6-phosphatase. Roughly 7% of the glycosidic bonds in glycogen are or(1 --+ 6) linkages. Under normal conditions, glycogen phosphorylase and debranching enzyme act simultaneously at different regions of the glycogen molecule. Deficiency of either enzyme prevents complete glycogen degradation. Glycogen phosphorylase deficiency leaves the original glycogen molecule untouched. Deficiency of debranching enzyme results in a glycogen molecule smaller than the original, with very short chains on the outer branches but with the inner core unchanged (a limit dextrin).

Regulation of Glycogen Metabolism Metabolism of glycogen in muscle and liver is regulated primarily through control of glycogen synthase and glycogen phosphorylase. The activities of these enzymes vary according to the metabolic needs of the tissue (as in muscle) or of other tissues that use glucose as a fuel (as in liver). Proximal control is exerted on synthase and phosphorylase by phosphorylation/dephosphorylation and by allosteric effectors such as glucose, glucose-6-phosphate, and several nucleotides (ATE ADP, AMP, and UDP). The concept that allosteric effectors are of primary importance in regulating synthase and phosphorylase activities was based mostly on in vitro experiments. For example, inactive (phosphorylated) glycogen synthase can be activated, without dephosphorylation, by glucose-6phosphate. For this reason, the active and inactive forms of this enzyme were formerly called glycogen synthase I (glucose-6-phosphate independent) and glycogen synthase D (glucose-6-phosphate dependent). It now appears that the concentration of glucose-6-phosphate may not

vary widely enough, particularly in muscle, to change synthase activity significantly, although glucose-6-phosphate binding may help determine the basal activity of the enzyme. The two forms are now known as glycogen synthase a (dephosphorylated) and b (phosphorylated). The same type of nomenclature is used for glycogen phosphorylase, except that phosphorylase a, the active form, is phosphorylated, while phosphorylase b is dephosphorylated. The regulation of glycogen metabolism in liver and in muscle differs in several ways. Although the control mechanisms are not completely understood, particularly in liver, the differences probably are due to the receptors in each tissue and to the presence of glucose-6-phosphatase in liver rather than to differences in the intrinsic regulatory properties of the enzymes involved. This aspect of tissue differences in glycogen metabolism between muscle and liver probably also applies to that in brain, myocardium, and other tissues. In all tissues, the rate of glycogen synthesis must be inversely proportional to the rate of glycogenolysis to avoid futile cycling. Muscle In muscle, glycogen is used as a fuel for anaerobic metabolism during brief periods of high-energy output (e.g., sprinting). Glycogenolysis is initiated and glycogenesis inhibited by the onset of muscle contraction and by factors such as epinephrine that signal a need for muscular activity. Since muscle glycogen is not a source of glucose for other tissues, it is not sensitive to blood glucose levels.

Control of Glycogen Synthase Muscle glycogen synthase (M.W. ~340,000) is a tetramer of identical subunits that exists in several forms, which differ in catalytic activity and degree of covalent modification. Glycogen synthase a, an active dephosphorylated form, can interconvert with several less active, phosphorylated forms, collectively called glycogen synthase b. The enzyme contains at least nine serine residues located near the extremities of the molecule, which can be phosphorylated by protein kinases (Figure 15-9). For the most part, the sites can be phosphorylated in any order. An exception is site C42, which undergoes phosphorylation by glycogen synthase kinase-3 only after phosphorylation at site C46 by casein kinase-2. Once C46 and C42 are phosphorylated in that order, glycogen synthase kinase-3 phosphorylates at sites C38, C34, and C30. In general, phosphorylation reduces synthase activity, and an increase in the number of phosphorylated sites additively decreases the activity. Reduced synthase activity may be manifested as increased Km for UDP-glucose, increased

SECTION15.2 Glycogen Metabolism

287

5. Glycogen synthase kinase-4 (GSK-4; site N7; relatively specific for glycogen synthase). 6. Casein kinase-2 (site C46; broadly specific; physiologically important substrates not yet known). 7. A novel protein kinase that may be activated by cAMP-dependent protein kinase (site N 10).

F I G U R E 15-9 Regulation of glycogen synthase by multisite phosphorylation. The location of phosphorylation sites (*) and the protein kinases that phosphorylate at these sites (boxes) are shown. Phosphorylations occur only at N- and C-terminal regions of the enzymes, as indicated by CB-N and CB-C, respectively. The single-letter abbreviations for amino acids are used (see Chapter 2). cAMP-PK -- cyclic AMP-dependent protein kinase; CAM-MPK = Ca2+/calmodulin-dependent multiprotein kinase; PhK -phosphorylase kinase; GSK -- glycogen synthase kinase; CK -- casein kinase; N10-PK -- A novel protein kinase. [Reproduced with permission from E Cohen, Protein phosphorylation and hormone action. Proc. R. Soc. Lond. (Biol.)234, 115 (1988).]

Ka for glucose-6-phosphate, or decreased Ki for ADR inorganic phosphate, or other inhibitors, or a change in Vmax. At least seven different protein kinases can phosphorylate muscle glycogen synthase in vitro, but whether all are involved in vivo is not established. Some are moderately specific for glycogen synthase, while others phosphorylate a variety of proteins. They are listed below, together with the sites that they phosphorylate (Figure 15-9). 1. cAMP-dependent protein kinase (sites N7, C87, and C100, broad specificity; mediates effects of many hormones). 2. Phosphorylase kinase (site N7; main function in vivo probably is activation of glycogen phosphorylase). 3. CaZ+/calmodulin-dependent multiprotein kinase (sites N7 and C100; relatively specific for glycogen synthase). 4. Glycogen synthase kinase-3 (GSK-3; sites C30, C34, C38 and C42; also phosphorylates inhibitor-2 (see below), thereby activating protein phosphatase-1).

In muscle, cAMP-dependent protein kinase (Chapter 30) mediates the inhibitory effect of epinephrine on glycogenesis by phosphorylating protein phosphatase inhibitor-1 and phosphorylating sites N7, C87, and C100. The inhibitory effect of cytosolic Ca 2+ on glycogen synthesis may be mediated by calmodulin-dependent multiprotein kinase. GSK-3 may be a key control enzyme; it phosphorylates sites on glycogen synthase that have the greatest effect on synthase activity. GSK-3 also activates protein phosphatase-1 by phosphorylating inhibitor-2, however, and these seemingly opposing effects have not been fully explained. Stimulation by insulin of glycogen synthesis in muscle may be mediated by GSK-3 or phosphatases that remove the phosphate group at the GSK-3 sites. Reactivation of glycogen synthase requires removal of the phosphate groups by a phosphatase. Four enzymes that dephosphorylate glycogen synthase or one or more kinases in vitro have been isolated from skeletal muscle. 1. Protein phosphatase-1 (Mg2+/ATP-dependent phosphatase; multisubstrate protein phosphatase; M.W. of catalytic subunit is 35,000; major enzyme in regulation of glycogen metabolism in skeletal muscle; dephosphorylates glycogen phosphorylase,/~-subunit of phosphorylase kinase, and at least three sites of glycogen synthase; regulated by inhibitor-1, inhibitor-2, and GSK-3 + Mg 2+. ATP). 2. Protein phosphatase-2A (polycation-dependent phosphatase, M.W. ~200,000; M.W. of catalytic subunit ~38,000; function unclear). 3. Protein phosphatase-2B (calcineurin; dimer; catalytic subunit of M.W. 60,000 has Ca2+ binding site; CaZ+-binding subunit of M.W. ~ 18,000, partially homologous with calmodulin and troponin C; N-terminal glycine blocked by amide linkage with myristic acid; absolute requirement for Ca2+; regulated by CaZ+/calmodulin; dephosphorylates ot-subunit of phosphorylase kinase and inhibitory but not glycogen phosphorylase or synthase or/~-subunit of phosphorylase kinase). 4. Protein phosphatase-2C (monomer; M.W. 43,000; represents a small fraction of glycogen synthase phosphatase in skeletal muscle; may primarily regulate other metabolic pathway in vivo).

288

CHAPTER15 Carbohydrate Metabolism I1: Gluconeogenesis, Glycogen Synthesis and Breakdown, and Alternative Pathways

Other proteins involved in glycogen synthase control are calmodulin, inhibitor-1, and inhibitor-2. Troponin C, when complexed with Ca 2+, can activate phosphorylase kinase, but phosphorylase kinase may have little or no role in the inactivation of glycogen synthase in vivo. Inhibitor-1 (M.W. 18,600), when phosphorylated on threonine 35 by cAMP-dependent protein kinase, binds to and inhibits protein phosphatase-1. The concentration of protein phosphatase-1 is lower than that of inhibitors, and its activity is sensitive to the latter, suggesting that inhibitor- 1 phosphorylation may be an important regulatory mechanism for this phosphatase. Inhibitor-2 (M.W. 30,500) appears to be an integral part of protein phosphatase1, forming an inactive 1:1 complex, called Mg2+/ATP dependent phosphatase, with the catalytic subunit of this enzyme. Phosphorylation of inhibitor-2 (also at a threonine residue) activates protein phosphatase-1. This phosphorylation, which requires Mg2+-ATR is catalyzed by an "activating factor" (FA), GSK-3.

Control of Glycogen Phosphorylase Regulation of glycogen phosphorylase in muscle is accomplished by many of the same enzymes that control glycogen synthesis. Phosphorylase kinase converts the dimeric phosphorylase from the inactive to the active form by Mg 2+ and ATP-dependent phosphorylation of two identical serine residues. The principal enzyme that removes this phosphate may be protein phosphatase-1 (phosphorylase phosphatase). Phosphorylase kinase is a hexadecamer having four different subunits: c~ (M.W. 145,000) or or' (M.W. 133,000), fl (M.W. 128,000), ?, (M.W. 44,700), and 6 (M.W. 16,700), with the stoichiometry ota(or Of~)j~aY4~ 4 (Figure 15-10). Whether the oe- or oe'-subunit is present depends on the tissue. The subunits apparently differ in Ca 2+ sensitivity, since the 6'-subunit, discussed later, will not bind to the o~'-isozyme. A catalytic site on the ?'-subunit has considerable homology with the catalytic subunit of cAMP-dependent protein kinase (Chapter 30). Evidence for active sites on the or- and fl-subunits is weak. Phosphorylase kinase activity has an absolute requirement for Ca 2+, which binds to the 6-subunit. The amino acid sequence of this subunit is nearly identical to that of calmodulin, with four calcium binding sites, but unlike calmodulin, the 3-subunit is an integral part of the enzyme and does not dissociate from it in the absence of Ca 2+. In the presence of Ca 2+, kinase activity is further increased by phosphorylation of the or- and fi-subunits, catalyzed by cAMP-dependent protein kinase and several other kinases. Phosphorylation may activate the enzyme by disinhibiting

i

G

G

Tmponin C, calmodulin, ) olher related calcium-

)

1

binding proteins

F I G U R E 15-10 Subunit structure of muscle phosphorylase kinase. The oe- and fl-subunits are regulatory proteins containing the sites at which phosphorylation can activate the kinase. The V-subunit contains the catalytic site. The ~-subunit is a Ca2+-binding protein, essentially identical with calmodulin, Ca e+. Calcium binding to the &subunit causes a change in the quaternary structure of the enzyme, allowing binding of the 6'-subunit, thereby increasing kinase activity even in the absence of phosphorylation of the fl-subunit. [Modified and reproduced with permission from A. K. Campbell: Intercellular Calcium: Its Universal Role as Regulator, Wiley, New York, 1983. @ 1983 by John Wiley & Sons, Inc.]

it, since limited proteolysis (which degrades the or- and fl-subunits) and dissociation of the holoenzyme into partial complexes (/3V6 and g6) also cause activation. Activation by covalent modification increases the affinity of the kinase for phosphorylase b. The kinase also is activated by the binding of a 6'-subunit (Ca2+/calmodulin or Ca2+/troponin C) to a site formed by the or- and fl-subunits. There are sites for four 6'-subunits on the holoenzyme. Activation by Ca2+/troponin C is 20-30 times greater than by Ca2+/calmodulin. This route may provide a means of coordinating muscle contraction, initiated by binding of Ca 2+ to troponin C (Chapter 21), with glycogenolysis. Finally, phosphorylase kinase is allosterically activated by the binding of up to 8 mol of ADP. Only 20-30% of isolated phosphorylase kinase is associated with glycogen particles, whereas all glycogen phosphorylase activity is associated with glycogen particles. This fact, together with the subcellular localization of phosphorylase kinase in intact cells and its biochemical properties, suggests that phosphorylase kinase may control other metabolic pathways as well. Suggested phosphorylase kinase targets include the Na +, K +and Ca2+-ATPases in the sacrolemma and sarcoplasmic reticulum, respectively.

SECTION 15.2 Glycogen Metabolism

289 Epinephrine

Nervous stimulus

Outside Plasmamembrane Inside

CaZ+/troponinC (thin filaments)~

Outside Plasma membrane Inside

I/creased

[Ca=+] in cytosol

cAMP + PP~

t

usc,

coo, raO,oo

f

li

r

j

Activationof phosphorylase kinase Activation of

Activationof

Activation of calmodulin-

t

inhibitor-1

dependentmultiprotein kinase

-.~

--

..9

. ~

glycogenphosphorylase

t Increased glycogen

breakdown

~176 ~176

ATP

Activationof cAMPdependent proteinkinase

......

Activationof ~ k kate'~cla~e

.....

t

Inhibitionof

Activation of Inhibition of phosphorylase kinase ........ protein phosphatase-1 .........

%

"'glycogen synthase

l

Reduced

glycogen synthesis

F I G U R E 15-11 Possible mechanism for regulation of glycogen metabolism in skeletal muscle by changes in cytosolic calcium. Increased glycogen breakdown may be coordinated with muscle contractions, as indicated here. The actual control scheme is probably more complicated, since phosphoprotein phosphatases are also involved. Interactions with cAMP-activated reactions, which also may complicate regulation, are not included. Whether glycogen synthase is a substrate for phosphorylase kinase in vivo is unclear. [Modified and reproduced with permission from E Cohen, Protein phosphorylation and the control of glycogen metabolism in skeletal muscle. Philos. Trans. R. Soc. Lond. (Biol.) 302, 13 (1983).]

Integrated Regulation of Muscle Glycogen Metabolism Figures 15-11 and 15-12 provide a model for the regulation of glycogen metabolism in muscle. This model is consistent with many of the known facts, but alternative interpretations are equally plausible. Only two of the external stimuli that affect muscle glycogen metabolism are considered in these figures. 1. Initiation of muscle contraction by action potential from an o~ motor neuron depolarizes the muscle-cell membrane and causes an increase in the myoplasmic [Ca2+] (Chapter 21). The increase in calcium activates phosphorylase kinase and CaZ+/calmodulin-dependent kinase, which inactivate glycogen synthase and active glycogen phosphorylase (Figure 15-11). This step coordinates muscle contraction with glycogenolysis. These steps are reversed by one or more phosphatases at the end of contraction. 2. Binding of epinephrine to an adrenergic receptor away from a motor end plate on the muscle membrane causes glycogenolysis to increase and glycogenesis to decrease. This activates adenylate cyclase, increasing the cytosolic concentration of cAMP and activating

Activation of

o~176176

,,X

glycogenphosphorylase

L

Increased glycogen breakdown

Inhibition of

glycogensynthase

,*****9

? ....... ................................................................................ Reduced

glycogen synthesis

F I G U R E 15-12 Possible mechanism for regulation of glycogen metabolism in skeletal muscle by changes in cyclic AMP. The actual control scheme is probably more complicated, since changes in cytoplasmic calcium concentration are likely also to be important. Whether glycogen synthase is a substrate for phosphorylase ldnase in vivo is unclear (indicated by ?). Dashed arrows from protein phosphatase-1 to glycogen synthase and phosphorylase and phosphorylase kinase indicate that inhibition of dephosphorylation may be less important than activation of phosphorylation in changing the activities of these enzymes. [Modified and reproduced with permission from P. Cohen, Protein phosphorylation and the control of glycogen metabolism in skeletal muscle. Philos. Trans. R. Soc. Lond. (Biol.) 302 (1983).]

the cAMP-dependent protein kinase (Figure 15-12). The net effect in vivo is an increase in phosphorylation of glycogen synthase and phosphorylase. This may occur in several ways, and the relative importance of increased phosphorylation compared to decreased dephosphorylation remains unclear. Insulin also regulates skeletal muscle glycogen metabolism. Skeletal muscle is the major site of insulinstimulated glucose uptake from the bloodstream, most of which is converted to glycogen. The insulin-induced glucose uptake is mediated by the glucose transporter GLUT4 (Chapter 13). Binding of insulin to its cell surface receptor initiates a cascade of phosphorylation reactions followed by dephosphorylation reactions (Chapter 22). Activation of glycogen synthase occurs by dephosphorylation at sites phosphorylated by cAMP-dependent kinase and GSK-3. Protein phosphatase-1 is a key component of the insulin signaling pathway and it activates glycogen synthase; it simultaneously inactivates phosphorylase a and phosphorylase kinase, promoting glycogen synthesis. Inactivation of GSK-3 via the protein kinase B pathway has also

~0

CHAPTER15 Carbohydrate Metabolism I1: Gluconeogenesis, Glycogen Synthesis and Breakdown, and Alternative Pathways

been implicated in insulin-mediated activation of glycogen synthase. Liver In liver, glycogen serves as the immediate glucose reserve for other tissues, and regulation of its metabolism is closely linked to maintenance of blood glucose concentration. An important feature of glucose transport in hepatocytes is its bidirectional flux across the plasma membrane that is mediated by glucose transporter GLUT2. Insulin does not regulate GLUT2 and its function is not rate limiting. Therefore, the concentrations of glucose in the blood and hepatocytes are equal. During energy deprivation for short periods of time such as overnight fasting, moderate exercise, and stress, which cause hypoglycemia (e.g., hypovolemic shock), glycogenolysis is stimulated. During sustained periods of starvation, gluconeogenesis maintains blood glucose homeostasis. During refeeding, hepatocytes replenish their stores of glycogen by using glucose-6-phosphate obtained from either plasma glucose or from the gluconeogenic pathway using amino acids, glycerol, and propionate as precursors. Regulatory factors include glucagon, vasopressin, angiotensin II, and ot-adrenergic agonists (including epinephrine). Glycogen synthesis is stimulated by a rise in insulin or blood glucose concentration.

Control of Glycogen Synthase Hepatic glycogen synthase is similar to the muscle enzyme, although it is encoded by different genes. It is inactivated by phosphorylation and activated by dephosphorylation and may contain 12 mol of alkali-labile phosphate per mole of subunit. The phosphorylation sites have not been mapped, and the specificities of hepatic glycogen synthase kinases are not known. Several enzymes that phosphorylate glycogen synthase (muscle or liver) in vitro have been identified in liver. cAMP-dependent protein kinase, thought to mediate the inhibition of glycogen synthesis by glucagon, is similar to the muscle enzyme. A Ca2+/calmodulin-dependent protein kinase may mediate Ca2+-dependent inactivation of the synthase by vasopressin, angiotensin II, and o~-adrenergic agonists. Casein kinase I and GSK-3 have also been found in liver, and there are undoubtedly other hepatic glycogen synthase kinases. As in muscle, hepatic phosphorylase kinase is probably not important as a glycogen synthase kinase in vivo, despite its ability to readily phosphorylate the synthase in vitro. Muscle and liver thus have major regulatory pathways in common but differ in more subtle aspects of control.

Protein phosphatases-1, 2A, 2B, and 2C occur in mammalian liver and, as in skeletal muscle, possess essentially all of the phosphatase activity toward enzymes and regulatory proteins of glycogen metabolism. In liver, however, the ratios of the activities of phosphatase-2A and 2C to that of phosphatase-1 are seven-fold higher than in muscle. Although protein phosphatase-I sediments with glycogen particles in both tissues, a much smaller fraction is glycogen-associated in liver than in muscle. The specific activity of phosphatase-2B is lower in liver than in muscle. Protein phosphatase inhibitors-1 and 2 have been identified in liver, where they appear to function as they do in muscle. A disinhibitor protein (M.W. ~9,000) of liver can block the effects of inhibitors-1 and 2 on phosphatase-1.

Control of Glycogen Phosphorylase Liver glycogen phosphorylase exists in an inactive, dephosphorylated form and in at least one active, phosphorylated form. Conversion of phosphorylase b to phosphorylase a is catalyzed by phosphorylase kinase, which is activated by vasopressin, angiotensin II, and c~-adrenergic agonists (mediated by Ca 2+) and by glucagon (which elevates cAMP). Glucagon activation of phosphorylase is in some way antagonized by insulin. Dephosphorylation of glycogen phosphorylase a is probably catalyzed by either protein phosphatase-1 or 2A. Binding of free glucose to phosphorylase a makes it a better substrate for the phosphatase, increasing the rate of inactivation of glycogenolysis. Phosphorylase a is a competitive inhibitor of the reaction between glycogen synthase b and phosphatase, suggesting that the same enzyme dephosphorylates the synthase and kinase, activating one and inhibiting the other.

Integrated Regulation of Liver Glycogen Metabolism Glycogen metabolism in liver is regulated by phosphorylation and dephosphorylation of regulatory and metabolic enzymes. Control of the phosphorylation state is mediated by Ca 2+, cAME cytosolic glucose concentration, and perhaps, in the case of insulin, by another mechanism. When the glucose concentration rises in the hepatocyte, the rate of conversion of glycogen phosphorylase a to phosphorylase b increases. This decreases the rate of glycogenolysis and, initially, causes no change in glycogen synthesis. As the concentration of phosphorylase a falls, however, its ability to competitively inhibit glycogen synthase b dephosphorylation decreases. The rate of activation of glycogen synthase then increases, as does the rate of synthesis of glycogen. Glucose can be an effective regulator because its concentration in the hepatocyte varies

SECTION 15.3

Alternative Pathways of 61ucose Metabolism and Hexose Interconversions

with its concentration in the blood. The postprandial rise in blood glucose contributes to the inhibition of hepatic glycogenolysis and to the activation of glycogen synthesis. Following a meal, the blood level of insulin also rises, while that of glucagon falls. As glucagon concentration decreases, activity of the hepatocyte adenylate cyclase decreases and cytosolic cAMP concentration falls owing to degradation by phosphodiesterase. The result is a decreased phosphorylation of glycogen synthase and phosphorylase, activation of the synthase, and inhibition of the phosphorylase. Insulin, which may accelerate this shift, can also directly antagonize the cAMP-mediated effects of glucagon. The mechanisms of insulin action may include activation of a low-Km cAMP-phosphodiesterase, reduction in basal activity of cAMP-dependent protein kinase, or increase in basal activity of glycogen synthase phosphatase. Since the effects on the kinase and phosphatase are on basal activities, insulin may affect the metabolism of glycogen that has not been previously stimulated by glucagon. The net effect of blood glucose, insulin, and glucagon, then, is to coordinate the relative rates of hepatic glycogen synthesis and breakdown (see Chapter 22). In shock, inadequate perfusion of the heart, brain, and other organs is due to a variety of causes. The body's responses include release of epinephrine from the adrenal medulla, release of vasopressin from the neurohypophysis, and activation of the renin-angiotensin system with elevation of angiotensin II. In the liver, epinephrine (acting as an ot-adrenergic agonist), vasopressin, and angiotensin II increase the intrahepatocytic [Ca 2+ ], stimulating glycogenolysis. The consequent rise in blood glucose provides a rapid energy source in times of stress. Since shock is often caused by an absolute (hemorrhage) or relative (vasodilation) decrease in intravascular volume, release of the large amount of water of hydration stored with glycogen may contribute to restoration of blood volume.

Glycogen Storage Diseases This group of diseases is characterized by accumulation of normal or abnormal glycogen due to deficiency of one of the enzymes of glycogen metabolism. Although all are rare (overall incidence of ~1:25,000 births), they have contributed greatly to the understanding of glycogen metabolism. They are summarized in Table 15-1 and Figure 15-13. Infants with type I disease (subtypes Ia, Ib, Ic, and Id) develop hypoglycemia even after feeding because of inability to convert glucose-6-phosphate to glucose. Lactate is produced at a high rate in extrahepatic tissues and is transported to the liver for gluconeogenesis. The low insulin and high glucagon levels caused by hypoglycemia

291

promote glycogenolysis, leading to an unusual metabolic substrate cycle in these infants (Figure 15-14). The small amount of glucose released by the action of debranching enzyme is the only endogenous source of glucose available to these patients. Many characteristics of type I disease are attributed to the attendant hypoglycemia, and patients have been treated with frequent daytime feedings and continuous nocturnal intragastric feeding with a high-glucose formula. This regimen produces substantial improvement in growth, reduction in hepatomegaly, and normalization of other biochemical parameters. Feeding uncooked cornstarch every 6 hours resulted in normoglycemia, resumption of normal growth, and reduction in substrate cycling and liver size. The success of this simple nutritional therapy is thought to depend on slow hydrolysis of uncooked starch in the small intestine by pancreatic amylase, with continuous release and absorption of glucose. (Cornstarch that is cooked or altered in other ways is ineffective, presumably because of too rapid hydrolysis.) The therapy is ineffective in patients with low pancreatic amylase activity due, for example, to prematurity.

15.3 Alternative Pathways of Glucose Metabolism and Hexose Interconversions Glucuronic Acid Pathway The glucuronic acidpathway (Figure 15-15) is a quantitatively minor route of glucose metabolism. Like the pentose phosphate pathway, it provides biosynthetic precursors and interconverts some less common sugars to ones that can be metabolized. The first steps are identical to those of glycogen synthesis, i.e., formation of glucose-6-phosphate, its isomerization to glucose-l-phosphate, and activation of glucose-1-phosphate to form UDP-glucose. UDP-glucose is then oxidized to UDP-glucuronic acid by NAD + and UDP-glucose dehydrogenase. An aldehyde intermediate is formed that remains bound to an amino group on the enzyme as a Schiff base. Since this is a four-electron oxidation, two molecules ofNAD + are used for each molecule of UDP-glucuronic acid formed. UDP-glucuronic acid is bound to the enzyme as a thioester and is released by hydrolysis. UDP-glucuronic acid is utilized in biosynthetic reactions that involve condensation of glucuronic acid with a variety of molecules to form an ether (glycoside), an ester, or an amide, depending on the nature of the acceptor molecule. As in condensation reactions that use nucleotide sugars as substrates, the high-energy bond between UDP and glucuronic acid provides the energy to form the new

292

C

,mm

o~

9

Z

9

9

Z

s:::

=:-: r,.3

r.~

~.~~ 0 ;>

r,J

9

9

Z

r.~

r.~

~'" ~

~

r.~

~~

o

:~~ ~

~-~

9

i

-._~

:~ o

.,=

,.~

r,.)


Orotate )

D~H.~ NADH + H§ Orotate PRPP OPRT PPi

~

AIIopurinol

XO

PRT Oxipurinol ~ ~ OxipurinOlpRppf ~p~ Ribonucleotide 9

Orotidine 5"-Monophosphate (OMP) . . . . -~ Orotidine OMP Decarboxylase

Uridine 5"-Monophosphate

F I G U R E 17-8 The metabolic interrelationship between mitochondrial carbamoyl phosphate synthesis to urea formation and to cytosolic carbamoyl phosphate channeled into pyrimidine biosynthesis. In ornithine transcarbamoylase (OTC) deficiency, mitochondrial carbamoyl phosphate diffuses into the cytosol and stimulates pyrimidine biosynthesis, leading to orotidinuria. Administration of ailopurinol augments orotidinuria by increasing the flux in the pyrimidine biosynthetic pathway. CPS = Carbamoyl phosphate synthase, AT = aspartate transcarbamoylase, D = dihydroorotase, DH -- dihydroorotate dehydrogenase, OPRT = orotate phosphoribosyltransferase, XO = xanthine oxidase, PRT = phosphoribosyltransferase, PRPP = 5-phosphoribosyl-l-pyrophosphate.

of nitrogen to products other than urea is achieved by administration of sodium benzoate or sodium (or calcium) phenylacetate. Administration of benzoate leads to elimination of hippurate (benzoylglycine):

phosphate, catalyzed by mitochondrial glycine synthase (glycine cleavage enzyme). Phenylacetate or phenylbutyrate administration increases excretion of phenylacetylglutamine: O

O ~~'-~nO-

)--CH3--COO- + ATP4- + CoASHActivatingenzyme

CH3MC~SCoA + AMPz- + PPi3-

+ ATP 4- + CoASH Acfivalingenzyme

PhenylacetyI-CoA

Benzoate

CONH2 CONH2 I

O

~~--~mSmCoA

+ AMp2- + PPi 3-

BenzoyI-CoA

Hippurate is rapidly secreted since its clearance is five times greater than its glomerular filtration rate. The glycine nitrogen is derived from ammonia in a complex reaction that uses CO2, NADH, NS,Nl~ (a source of a single carbon unit; Chapter 27) and pyridoxal

O (0H2)2 II I CH3BC~SCoA + H3+N- CH I COOGlutamine

l

0 (CH~)~ II I Conjugatmgenzyme,/g~=.~CH3MC__NBCH

'

H I COO-

+ CoASH+ H +

Phenylacetylglutamine

The excretion of phenylacetylglutamine produces loss of two nitrogen atoms. NAG synthase deficiency cannot be treated by administration of NAG, since NAG undergoes cytosolic inactivation by deacylation and is not readily permeable across the inner mitochondrial membrane. An analogue of NAG,

SECTION 17.3

345

Metabolism of Some Individual Amino Acids

N-carbamoylglutamate, activates CPSI, does not share the undesirable properties of NAG, and has been effective in management of this deficiency. The most common cause of hyperammonemia in adults is disease of the liver (e.g., due to ethanol abuse, infection, or cancer). The ability to detoxify ammonia is decreased in proportion to the severity of the damage. In advanced disease (e.g., cirrhosis), hyperammonemia is augmented by shunting of portal blood that carries ammonia from the intestinal tract and other splanchnic organs to the systemic blood circulation (bypassing the liver) and leads to portalsystemic encephalopathy. In addition to dietary protein restriction, colonic growth of bacteria must be suppressed by antibiotics (e.g., neomycin) and administration of lactulose (Chapter 9), a nonassimilable disaccharide. Enteric bacteria catabolize lactulose to organic acids that convert NH3 to NH4+, thereby decreasing absorption of NH3 into the portal circulation. Catabolism of lactulose also leads to formation of osmotically active particles that draw water into the colon, produce loose, acid stools, and permit loss of ammonia as ammonium ions.

17.3 Metabolism of Some Individual Amino Acids Mammalian tissues synthesize the nonessential amino acids from carbon skeletons derived from lipid and carbohydrate sources or from transformations that involve essential amino acids. The nitrogen is obtained from NH4+ or from that of other amino acids. Nonessential amino acids (and their precursors) are glutamic acid (ot-ketoglutaric acid), aspartic acid (oxaloacetic acid), serine (3-phosphoglyceric acid), glycine (serine), tyrosine (phenylalanine), proline (glutamic acid), alanine (pyruvic acid), cysteine (methionine and serine), arginine (glutamate-y-semialdehyde), glutamine (glutamic acid), and asparagine (aspartic acid). Amino acids may be classified as ketogenic, glucogenic, or glucogenic and ketogenic, depending on whether feeding of a single amino acid to starved animals or animals with experimentally induced diabetes increases plasma or urine levels of glucose or ketone bodies (Chapter 18). Leucine and lysine are ketogenic; isoleucine, phenylalanine, tyrosine, and tryptophan are glucogenic and ketogenic; and the remaining amino acids are glucogenic. Points of entry of amino acids into the gluconeogenic pathway are discussed in Chapter 15.

Arginine

Arginine participates in a number of metabolic pathways depending on the cell type. It is synthesized as an inter-

mediate in the urea cycle pathway and is also obtained from dietary proteins. A number of key metabolites such as nitric oxide, phosphocreatine, spermine and ornithine are derived from arginine. During normal growth and development, under certain pathological conditions (e.g., endothelial dysfunction) and if the endogenous production of arginine is insufficient, a dietary supplement of arginine may be required. Thus, arginine is considered a semiessential amino acid.

Metabolism and Synthesis of Nitric Oxide

Nitric oxide (NO) is a reactive diatomic gaseous molecule with an unpaired electron (a free radical). It is lipophilic and can diffuse rapidly across biological membranes. NO mediates a variety of physiological functions such as endothelial derived relaxation of vascular smooth muscle, inhibition of platelet aggregation, neurotransmission, and cytotoxicity. The pathophysiology of NO is a doubleedged sword. Insufficient production of NO has been implicated in the development of hypertension, impotence, susceptibility to infection, and atherogenesis. Excessive NO production is linked to septic shock, inflammatory diseases, transplant rejection, stroke, and carcinogenesis. NO is synthesized from one of the terminal nitrogen atoms or the guanidino group of arginine with the concomitant production of citrulline. Molecular oxygen and NADPH are cosubstrates and the reaction is catalyzed by nitric oxide synthase (NOS). NOS consists of several isoforms and is a complex enzyme containing bound FMN, FAD, tetrahydrobiopterin, heme complex, and nonheme iron. A calmodulin binding site is also present. NO formation from arginine is a two-step process requiring five-electron oxidations. The first step is the formation of NC-hydroxylarginine (N G denotes guanidinium nitrogen atom): Arginine + O2 + NADPH + H + -+ HO-NC-Arg + NADP + + H20 This step is a mixed-function oxidation reaction similar to the one catalyzed by cytochrome P-450 reductase and there is considerable homology between NOS and cytochrome P-450 reductase. In the second step, further oxidation of NG-hydroxyl arginine yields NO and citrulline:

1 HO-NG-Arg + 02 -~- ~(NADPH + H +) --+ 1 citrulline + NO + H20 -k- - N A D P + 2

346

CHAPTER 17 Protein and Amino Acid Metabolism

The overall reaction is" O

H

II

I

?H 2

"Om C n OH m (0H2)3~ N - - C - - N H

I

+ 0 2 + NADPH + H §

NH3+ Arginine Nitric Oxide Synthase

/ 1

FMN, FAD, Tetrahydrobiopterin Fe 2+, Heme complex

o H O II I II -O-- C - - OH - - (OH2) 3 - N -- C - - NH 2 + NO

I

interferon-v), or bacterial lipopolysaccharides can induce and cause expression of NOS in many cell types. Glucocorticoids inhibit the induction of iNOS. In stimulated macrophages and neurophils, NO and superoxide radical (O2) react to generate peroxynitrite, a powerful oxidant, and hydroxyl radicals. These reactive intermediates are involved in the killing of phagocytized bacteria (Chapter 14). Excessive production of NO due to endotoxinemia produces hypotension and vascular hyporeactivity to vasoconstrictor agents, and leads to septic shock. NOS inhibitors have potential therapeutic application in the treatment of hypotensive crisis.

NH3+ Citrulline

The NOS activity is inhibited by NO-substituted analogues of arginine, such as NC-nitroarginine and N cmonomethyl-L-arginine.

Isoforms (Also Known as Isozymes) of Nitric Oxide Synthase There are three major isoforms of Nitric Oxide Synthase (NOS) ranging in molecular size from 130 to 160 kDa. Amino acid similarity between any two isoforms is about 50-60%. Isoforms of NOS exhibit differences in tissue distribution, transcriptional regulation, and activation by intracellular Ca 2+. Two of the three isoforms of NOS are constitutive enzymes (cNOS) and the third isoforin is an inducible enzyme (iNOS). The cNOS isoforms are found in the vascular endothelium (eNOS), neuronal cells (nNOS), and many other cells, and are regulated by Ca 2+ and calmodulin. In the vascular endothelium, agonists such as acetylcholine and bradykinin activate eNOS by enhancing intracellular Ca 2+ concentrations via the production of inositol 1,4,5-trisphosphate, which activates the phosphoinositide second-messenger system (Chapter 30 ). The NO produced in the vascular endothelium maintains basal vascular tone by vasodilation which is mediated by vascular smooth muscle cells. Organic nitrates used in the management of ischemic heart disease act by denitration with the subsequent formation of NO. Sodium nitroprusside, an antihypertensive drug, is an NO donor. Thus, organic nitrates and sodium nitroprusside are prodrugs, and the exact mechanism by which these prodrugs yield NO is not yet understood. Inhaled NO can produce pulmonary vasodilation. This property of NO has been used in the management of hypoxic respiratory failure associated with primary pulmonary hypertension in neonates. NO produced by cNOS in neuronal tissue functions as a neurotransmitter. The inducible class of NOS (iNOS) is found in macrophages and neutrophils and is Ca2+-independent. Bacterial endotoxins, cytokines (e.g., interleukin-1,

Signal Transduction of NO NO is lipophilic and diffuses readily across cell membranes. It interacts with molecules in the target cells producing various biological effects. One mechanism of action of NO is stimulation of guanylate cyclase, which catalyzes the formation of cyclic guanosine monophosphate (cGMP) from GTP, resulting in increased intracellular cGMP levels (Figure 17-9). NO activates guanylate cyclase by binding to heme iron. The elevation of cGMP levels may activate cGMP-dependent protein kinases. O

II ~ "~0\5,2

HN

II

II

O-- I~--0-- P--O--I.P - - O -

H"~H H//~,H F--V-' OH OH

O-

O-

O-

Guanosine Triphosphate (GTP)

J PPi j

GuanylateCyclase ~

~

Derived from Nerve Impulse 7/

NO Derived from a Ligand-cell interaction

O

HN

H2N : o .,> Smoothmuscle .. relaxation O , Cyclic GMP

~

CH3CH20

Cyclic GMP phosphodiesterase O

/CH 3

HN~N\

....

I /;

N~CH2CH2CH3

O2S\ N" ~

HN

N%,

H2N - ~'N /

-Nk/o

O 5" C -- O-- P-- O-

H, H ,/ ~ H

I O-

~.

CH3

Sildenafil

OH OH 5"-GMP

F I G U R E 17-9 NO-mediated synthesis of cGMP from GTP in the corpus cavernosum that leads to smooth muscle relaxation. Sildenafil potentiates the effects of NO by inhibiting cGMP phosphodiesterase.

SECTION17.3 Metabolism of Some Individual Amino Acids --Glucose

Purines Proteins\\

3-PhosphoglyceratJ/ f ~ Pyruvate ~TCA ,_ | ~ Serin e ~ Ethan~ a m i n e . , ~ cycle-----*CO2 oI line

Urea_ Glutathione~\ \ " ~ ~ " + NH~ '+ " ~ ~GlyciEe \~ "One-carbon" J / ~ " / ~ ~ Bileacidconjugates Hippurate and // ~ ~ otheracyl conjugates Porphyrins

347

Glyoxalate l . Glycolate"

" Phospholipids

Creatine Oxalate

1 1 Creatinine

Phosphocreatine

FIGURE 17-11) Overviewof glycineand serinemetabolism.

These kinases phosphorylate specific proteins that may be involved in removal or sequestration of Ca 2+ or other ions, resulting in physiological stimuli. The physiological actions of cGMP are terminated by its conversion to 5'-GMP by cGMP-phosphodiesterase. Inhibitors of cGMP phosphodiesterase promote the actions of NO. Sildenafil is a selective inhibitor of a specific cGMP phosphodiesterase (type 5) present in the corpus cavernosum. This compound (structure shown in Figure 17-9) is used orally in the therapy of some types of erectile dysfunction. NO is the principal transmitter involved in the relaxation of penile smooth muscle. During central or reflex sexual arousal, NO production is enhanced leading to increased production of cGME Smooth muscle relaxation permits the corpus cavernosum to fill with blood. Since the therapeutic effect of sildenafil potentiates the action of cGMP, the drug is ineffective in the absence of sexual arousal. The relaxation of cavernosal smooth muscle caused by cGMP involves inhibition of Ca 2+ uptake. Prostaglandin E1 (alprostadil) inhibits the uptake of Ca 2+ smooth muscle by a separate mechanism and causes erections in the absence of sexual arousal. Blood flow through corpus cavernosum may also be increased by o~-adrenergic blocking agents (e.g., phentolamine mesylate). Coadministration of NO donor drugs with the NO potentiation drug sildenafil may have severe consequences on the cardiovascular system. Signal transduction of NO by cGMP-independent mechanisms include ADP-ribosylation of glyceraldehyde3-phosphate dehydrogenase (GADPH), an enzyme of the glycolytic pathway (Chapter 13), and interactions with many heme-containing and nonheme iron-sulfur containing proteins. NO activates ADP-ribosyltransferase which catalyzes the transfer of ADP-ribose from NAD + to GADPH. This results in the inactivation of GADPH causing inhibition of glycolysis and decreased ATP production.

The antiaggregability of platelets and the neurotoxicity of NO have been attributed to inhibition of glycolysis by NO.

Glycine Glycine participates in a number of synthetic pathways and is oxidized to provide energy (Figure 17-10). The interconversion of glycine and serine by serine hydroxymethyltransferase is shown below: pyridoxalphosphate NH~--CH2-COO- + N 5,N 10-methylene-FH4 + H20 < > glycine H O H z C - C H N H + - C O O - + FH4 serine

The one-carbon carrier NS,Nl~ is derived from reactions of the one-carbon pool (Chapter 27). [The term one-carbon pool refers to all single-carbon-containing metabolites (e.g.,-CH3,-CHO, N H = C - , etc.) that can be utilized in biosynthetic reactions such as formation of purine and pyrimidine.] These reactions include oxidation of glycine by glycine cleavage enzyme complex (glycine synthase): pyridoxal phosphate

NH+-CH2-COO - nt- FH4 + NAD +


glycine

NH + +

C02

+ NADH + NS,Nl~

This reaction favors glycine degradation, but the formation of glycine may also occur. The enzyme complex is mitochondrial and contains a pyridoxal phosphate-dependent glycine decarboxylase, a lipoic acid-containing protein that is a carrier of an aminomethyl moiety, a tetrahydrofolate-requiring enzyme, and lipoamide dehydrogenase. The reactions of glycine cleavage resemble those of oxidative decarboxylation of pyruvate (Chapter 13).

348

CHAPTER 17 Protein and Amino Acid Metabolism

Glycine is also oxidized by D-amino acid oxidase, an FAD protein:

NH~--CH2-COO- -Jr-02 -~- H20 glycine C H O - C O O - + NH + + H202 glyoxalate Glyoxalate can be transaminated to glycine, reduced to glycolate, converted to ot-hydroxy-/~-ketoadipate by reaction with o~-ketoglutarate, or oxidized to oxalate and excreted in urine. The first three reactions require pyridoxal phosphate, NADH, and thiamine pyrophosphate, respectively. In humans, ascorbic acid (vitamin C) is a precursor of urinary oxalate (Chapter 38). Since calcium oxalate is poorly soluble in water, it can cause nephrolithiasis and nephrocalcinosis due to hyperoxaluria.

Creatine and Related Compounds Phosphocreatine serves as a high-energy phosphate donor for ATP formation (e.g., in muscle contraction; see Chapter 21). Synthesis of creatine (methyl guanidinoacetate) requires transamidination, i.e., transfer of a guanidine group from arginine to glycine, to form guanidinoacetate (glycocyamine) by mitochondrial arginine-glycine amidinotransferase NH2 I ~NH2 I

NH3 +

NH3 +

NH2 I ~NH2+

I

I

(CH2)3

+

NH I

+

CH 2

~

'~

I I

NH

(CH2)3

I

I

CHNH3

I CHNH3 +

COO-

CH2 I

CO0 -

I

I

coo-

COO -

Arginine

Disorders of Glycine Catabolism Nonketotic hyperglycinemia is an inborn error due to a defect in the glycine cleavage enzyme complex in which glycine accumulates in body fluids and especially in cerebrospinal fluid. It is characterized by mental retardation and seizures. Glycine is an inhibitory neurotransmitter in the central nervous system, including the spinal cord. Strychnine, which produces convulsions by competitive inhibition of glycine binding to its receptors, gives modest results in treatment but not very effective. Sodium benzoate administration reduces plasma glycine levels but does not appreciably alter the course of the disease. Exchange transfusion may be useful. Ketotic hyperglycinemia also occurs in propionic acidemia but the mechanism has not been established. Primary hyperoxaluria type I is due to a deficiency of cytosolic oe-ketoglutarate-glyoxylate carboligase, which catalyzes the following reaction:

Glycine

II

Glyoxalate

In the next step guanidinoacetate is methylated by Sadenosylmethionine by cytosolic S-adenosylmethionine guanidinoacetate-N-methyltransferase to form creatine. OH 3 I S+.Adenosyl I (0H2)2

I

+

NH2 I C--NH2 + [ NH

I

Carboligase

O

OH

II

CH2

COO-

COO-

COO-

S-Adenosylmethionine

Guanidino-

Creatine

I

The glyoxylate oxalate.

that

accumulates

is

converted

H+

coo -

S-Adenosylhomocysteine

acetate

NH--PO3 2-

I

C=NH2 +

I

N - - C H 3 + ATP 4- ~

N--CH 3

I

I

CH2

CH 2

I

to

I

+

These reactions occur in liver, kidney, and pancreas, from which creatine is transported to organs such as muscle and brain. Creatine synthesis is subject to negative modulation of amidinotransferase by creatine. Phosphocreatine production is catalyzed by creatine kinase:

I

0e-Hydroxy-/Y-ketoadipate

S-Adenosyl I (CH2)2 I CHNH3 § I

+

I

I

C--NH2 +

CO2 + - O O C ~ C H 2 m C H 2 - - C ~ C H ~ C O O -

~

CH2.

I

I

Thiaminepyrophosphate

NH2 I C--NH2 + I N--OH3

CHNH3 +

NH2

0c-Ketoglutarate

Ornithine

(glycocyamine)

O C H O m C O O - + - OOC__CH2~CH2wC..._COO -

Guanidinoacetate

+

ADP 3- + H +

I

COO-

COO-

Creatine

Phosphocreatine

This kinase is a dimer of M and B (M = muscle, B = brain) subunits produced by different structural genes. Three isozymes are possible: BB (CK-1), MB (CK-2), and MM (CK-3). Another isozyme differs immunologically and electrophoretically and is located in the intermembrane space of mitochondria. Tissues rich in CK-1

349

SECTION17.3 Metabolism of Some Individual Amino Acids

are brain, prostate, gut, lung, bladder, uterus, placenta, and thyroid; those rich in CK-3 are skeletal and cardiac muscle. Cardiac muscle contains significant amounts of CK-2 (25-46% of total CK activity, as opposed to less than 5% in skeletal muscle), so that in myocardial infarction the rise in serum total CK activity is accompanied by a parallel rise in that of CK-2 (Chapter 8). Phosphocreatine undergoes a slow and nonenzymatic cyclization to creatinine.

NH~PO3 2I ~NH2, I N--CH3 +H* I CH2

NH H2N---C , \

j

COOPhosphocreatine

/

~~O /

+ Pj~- + H20

N~CH2

HaC Creatinine

Creatinine has no useful function and is eliminated by renal glomerular filtration and to a small extent by renal tubular secretion. Creatinine clearance approximately parallels the glomerularfiltration rate (GFR) and is used as a kidney function test. It is calculated as follows:

regenerates ATP from ADP, thereby maintaining a high level of ATP required during intense exercise. A large pool of phosphocreatine resides in the skeletal muscle. It has been theorized that in order to maximize phosphocreatine stores in the skeletal muscle to replenish ATP during rapid muscle contractions, an exogenous source of creatine may be beneficial. Double-blind placebo-controlled studies of oral supplementation of creatine in human subjects have shown increased performance during short duration, strenuous, high-intensity exercise. Such activities require that ATP be replenished rapidly from phosphocreatine stores during anaerobic metabolism. These studies usually consisted of ingestion of 20 g of creatine per day for 5 days followed by a maintenance dose of 5-10 g/day. Studies on creatine as an ergogenic aid have not been uniformly positive; some have shown no beneficial effect and still others have been equivocal and indicated that creatine supplementation did not enhance athletic activities. The safety issues of long-term creatine supplementation on kidney, liver, nerve, muscle, and other tissues are not known.

Serine

Creatinine concentrations are measured from a precisely timed urine specimen (e.g., 4-hour, 24-hour) and a plasma specimen drawn during the urine collection period. Excretion of creatinine depends on skeletal muscle mass and varies with age and sex. However, day-today variation in a healthy individual is not significant. Creatinuria, the excessive excretion of creatine in urine, may occur during growth, fever, starvation, diabetes mellitus, extensive tissue destruction, muscular dystrophy, and hyperthyroidism.

Synthesis of serine from 3-phosphoglycerate, an intermediate of glycolysis (Chapter 13), requires oxidation of 3-phosphoglycerate to 3-phosphohydroxypyruvate, transamination of 3-phosphohydroxypyruvate by glutamate, and hydrolysis of 3-phosphoserine to serine (Figure 17-11). This cytosolic pathway is regulated by inhibition of phosphoserine phosphatase by serine. Serine is converted to pyruvate by cytosolic serine dehydratase. More importantly, it is converted in mitochondria to 2-phosphoglycerate by way of hydroxypyruvate and D-glycerate; and the enzymes involved are a transaminase, a dehydrogenase, and a kinase. Serine is interconvertible with glycine (Figure 17-10) and is involved in phospholipid (Chapter 19) and in cysteine synthesis.

Use of Creatine as a Dietary Supplement

Proline

The creatine pool in the human body comes from both endogenous synthesis and the diet which provides 1-2 g. Red meat provides large amounts of dietary creatine and vegetables a limited amount. Using glycine, arginine, and methionine, creatine is synthesized in the liver, pancreas, and kidney. Creatine transported in blood crosses muscle and nerve cell membranes by means of a specific creatine transporter system against a concentration gradient of 200:1. Intracellularly, creatine is converted to phosphocreatine by ATE a reaction catalyzed by creatine kinase. Phosphocreatine, with its high phosphoryl transfer potential,

Proline arises from and gives rise to glutamate. Synthesis is by reduction of glutamate to glutamate-v-semialdehyde by way of an enzyme-bound v-glutamyl phosphate. The v-semialdehyde spontaneously cyclizes to A'-pyrroline5-carboxylate, which is then reduced by NAD(P)H to proline (Figure 17-12). Proline is converted to A'-pyrroline-5carboxylate by proline oxidase, which is tightly bound to the inner mitochondrial membrane in liver, kidney, heart, and brain. A'-Pyrroline-5-carboxylate is in equilibrium with glutamate-v-semialdehyde, which can be transaminated to ornithine or reduced to glutamate (Figure 17-12).

Creatinine clearance =

urine creatinine (mg/L) plasma creatinine (mg/L) • urine volume per unit time

350

CHAPTER17 Protein and Amino Acid Metabolism cool

Phosphoglycerate dehydrogenase ,

Glycolysis-"-"-"-~ HC-- OH

coo-

I

H --O--(~ NAD+ NADH+ H2C--O--(~ 3-Phosphoglycerate 3-Phosphohydroxypyruvate t~nGlutamate

osphoserine saminase ec-Ketoglutarate

COO-

COOPhosphoserine -_ phosphatase(,,.~ -'~ H2N~H

, H3N?H I

CH~----OH L-Serine

P~

HO

F I G U R E 17-11 Synthesis of serine from 3-phosphoglycerate. P = PO 2-"

Pi

--

~C--O--(~ 3-Phosphoserine HPO 2-.

Decarboxylation of ornithine to putrescine by ornithine decarboxylase serves as a source of the polyamines spermidine and spermine. Ornithinemia results from deficiency of ornithine aminotransferase or ornithine

decarboxylase. Ornithine aminotransferase deficiency is associated with gyrate atrophy of the choroid and retina (Chapter 38). Proline and hydroxyproline (produced by posttranslational modification) are major constituents

CO01

ATP (?H2)= ADP,~......-------~ ? HNH+ COONADPH+ H+.~/(~" L-Glutamate

NAOP

)

CHO I (C,H,)~ ~ ICHNH+ ~

._. /

(~/---NADH+ H§ NAD+

ICOO- ~ ~ H Glutamate-7-semialdehyde _ l,,-----Glutamate ( ' ~ F er -Ketoglutarate ?(rH2NH+ (H~ CHNH+ I COOOrnithine Via

reactionsof ureacycle Arginine

|

C02 3 ,

2

0

?HF---NH+ (?Hz), CH=NH+ Putrescine

I-I=? H20 H+~

?Hz

HC~N/CHCOOH A -Pyrroline5"carbOxylate

NAD(P)H+

Flav%iotein

NAD(P)*-~ CH2

~C

I H2C~N/CHCOOH H

Proline

FIGURE 17-12 Metabolism of proline (1) A1-pyrroline-5-carboxylate (P5C) synthase; (2) ornithine aminotransferase; (3) P5C reductase; (4) proline oxidase; (5) P5C dehydrogenase; (6) ornithine decarboxylase.

Metabolism of Some Individual Amino Acids

SECTION 17.3

of collagen (Chapter 25). Hydroxyproline released by collagen turnover undergoes degradation similar to that of proline. Hydroxyproline cleavage initiated by hydroxyproline oxidase eventually yields glyoxylate and pyruvate. In hyperprolinemia type I, proline oxidase is deficient, and in type II, A'-pyrroline-5-carboxylate dehydrogenase is deficient. Hydroxyprolinemia results from hydroxyproline oxidase deficiency. All are clinically harmless autosomal recessive traits.

Histidine Histidine is not essential for adults except in persons with uremia. It is essential for growth in children. Histidine is synthesized from 5-phosphoribosyl1-pyrophosphate and ATE forming N'-l'-phosphoribosylATE catalyzed by the allosteric enzyme ATP phosphoribosyltransferase. This reaction is analogous to the initial reaction of purine nucleotide biosynthesis (Chapter 27). Histamine breakdown produces a one-carbon unit (N 5formiminotetrahydrofolate) and glutamate (Figure 17-13) by a nonoxidative deamination to urocanate, cleavage of the imidazole ring to N-formiminoglutamate (Figlu), and transfer of the formimino group (-CH=NH) to tetrahydrofolate (Chapter 27). Folate deficiency leads to accumulation of Figlu, which is excreted in urine. The excretion is very pronounced after a loading dose of histidine, a test used to detect folate deficiency. More sensitive radioisotopic assays use folate binders to the vitamins. High urinary levels of Figlu may coexist with elevated levels of serum folate. Thus, in vitamin B]2 (cobalamin) deficiency, since cobalamin participates in the following reaction, FH4 is trapped as

NH +

I

---------~CHF-CH--COO-

N~.~/NH

Histidineammonia-lyase ,. (histidase)

NH3

L-Histidine

O

Urocanate

H20-~ Urocanate hydratase

H

II I -O--C--C--CH~--C~COOI HN~c./NH

I

H N-Formiminoglutamate(Figlu) Glutamate~ F H , formiminotransferase NLformino.FH ' O

II

-O-- C-- CH--CH.~H2COOL-Glutamate

FIGURE 17-13 Catabolismof histidine.

I]--C"=CHCOO N~,,.,/.NH

Imidazolone=~oropionase ~0

0~]~ 1

CHF"CH2COO-

N~NH 4-1midazolone-5-propionate

351

NS-methyltetrahydrofolate and is unavailable as a carrier for the formimino group of Figlu. NS-Methy I-FH 4

Homocysteine methyltransferase

(methylcobalamin) /~

Homocysteine

~FH4

Methionine

Similarly, a deficiency of glutamate formiminotransferase leads to accumulation of Figlu and high levels of serum folate. Histidinemia results from deficiency of histidine ammonia-lyase. With a normal diet, histidine (and the products imidazole-pyruvate, imidazole-lactate, imidazoleacetate) accumulates in plasma, cerebrospinal fluid, and urine. This rare autosomal recessive disease may be benign or may manifest with mental retardation and speech defects. Histidine and #-alanine yield the dipeptide carnosine (present in muscle), and histidine and 7-aminobutyrate yield homocarnosine (found in brain). Methylhistidyl residues are found in some proteins (e.g., actin; Chapter 21) as a result of posttranslational modification. Histamine is decarboxylated histidine. H I N~.~,,,,, NH Histidine

COO-

Histidine decarboxylase

(pyridoxal phosphate)

CO2

)[

] CH2--CH2--NH3+

N~,,~,,,/N H Histamine

Histamine occurs in blood basophils, tissue mast cells, and certain cells of the gastric mucosa and other parts of the body (e.g., anterior and posterior lobes of the pituitary, some areas of the brain). Histamine is a neurotransmitter in certain nerves ("histaminergic") in the brain. In mast cells found in loose connective tissue and capsules, especially around blood vessels, and in basophils, histamine is stored in granules bound by ionic interactions to a heparin-protein complex and is released (by degranulation, vacuolization, and depletion) in immediate hypersensitivity reactions, trauma, and nonspecific injuries (infection, burns). Degranulation is affected by oxygen, temperature, and metabolic inhibitors. Release of histamine from gastric mucosal cells is mediated by acetylcholine (released by parasympathetic nerve stimulation) and gastrin and stimulates secretion of hydrochloric acid (Chapter 12). Histamine causes contraction of smooth muscle in various organs (gut, bronchi) by binding to H1 receptors. The conventional antihistaminic drugs (e.g., diphenhydramine and pyrilamine) are Hi-receptor antagonists and are useful in the management of various allergic manifestations. However, in acute anaphylaxis, bronchiolar constriction is rapidly relieved by epinephrine (a physiological antagonist

352

CHAPTER 17

Protein and Amino Acid Metabolism

of histamine). Its effect on secretion of hydrochloric acid is mediated by H2 receptors. Hz-receptor antagonists are cimetidine and ranitidine (Chapter 12), which are useful in treatment of gastric ulcers. Histamine is rapidly inactivated by methylation from S-adenosylmethionine of one of the nitrogen atoms of the imidazole ring (catalyzed by N-methyltransferase) or of the terminal amine group (catalyzed by methyltransferase). Ring-methylated histamine is deaminated by monoamine oxidase to methyl imidazole acetic acid, which is readily excreted. Inactivation also results from deamination of histamine by diamine oxidase. The imidazole acetic acid formed is then excreted as 1ribosylimidazole-4-acetic acid. This reaction is the only known reaction in which ribose is used for conjugation. Branched-Chain Amino Acids

Leucine, isoleucine, and valine are essential amino acids but can be derived from their respective ot-keto acids. A single enzyme may catalyze transamination of all three. The ot-keto acids, by oxidative decarboxylation, yield the acyl-CoA thioesters, which, by ot,/3-dehydrogenation, yield the corresponding ot,fl-unsaturated acyl-CoA thioesters. The catabolism of these thioesters then diverges. Catabolism of leucine yields acetoacetate and acetyl-CoA via fl-hydroxy-fl-methylglutaryl-coenzyme A (HMG-CoA)malso an intermediate in the biosynthesis of cholesterol and other isoprenoids (Chapter 19). Catabolism of isoleucine yields propionyl-CoA (a glucogenic precursor) and acetyl-CoA. Catabolism of valine yields succinyl-CoA (Figure 17-14). Thus, leucine is ketogenic and isoleucine and valine are ketogenic and glucogenic. Oxidative decarboxylation of the c~-keto acids is catalyzed by a branched-chain keto acid dehydrogenase (BCKADH) complex analogous to that of the pyruvate dehydrogenase and c~-ketoglutarate dehydrogenases complexes. BCKADH is widely distributed in mammalian tissue mitochondria (especially in liver and kidney). It requires Mg 2+, thiamine pyrophosphate, CoA-SH, lipoamide, FAD, and NAD + and contains activities of otketo acid decarboxylase, dihydrolipoyl transacylase, and dihydrolipoyl dehydrogenase. Like the pyruvate dehydrogenase complex, BCKADH is regulated by product inhibition and by phosphorylation (which inactivates) and dephosphorylation (which activates). The o~,fl-dehydrogenation is catalyzed by an FAD protein and is analogous to the dehydrogenation of straightchain acyl-CoA thioesters in fl-oxidation of fatty acids (Chapter 18). Methylenecyclopropylacetyl-CoA derived from the plant toxin hypoglycin (Chapters 15 and 18), which inhibits this step in fl-oxidation, also inhibits it in the catabolism of branched-chain amino acids.

FIGURE 17-14 Overviewof the catabolismof branched-chainaminoacids. TPP = thiamin pyrophosphate.

Hypoglycin produces hypoglycemia and metabolic acidosis, which frequently are fatal. Branched-chain ketoaciduria (maple syrup urine disease), an autosomal recessive disorder characterized by ketoacidosis starts early in infancy and is due to a defect in the oxidative decarboxylation step of branchedchain amino acid metabolism. The name derives from the characteristic odor (reminiscent of maple syrup) of the urine of these patients. Five different variants (classic, intermittent, intermediate, thiamine-responsive, and dihydrolipoyl dehydrogenase deficiency) are known, of which the first, which is due to deficiency of branchedchain ot-keto acid decarboxylase, is the most severe. The incidence of maple syrup urine disease in the U.S. population is 1 in 250,000-400,000 live births. In Mennonite populations the incidence is extremely high (1 in 760). Neonatal screening programs consist of measuring leucine levels in dried blood spots using a bacterial inhibition assay. Neonatal screening programs usually include testing for a number of other treatable metabolic disorders such as hypothyroidism, phenylketonuria, galactosemia, and others. If the screening test is positive for a given metabolic disease, a confirmatory test is performed. For maple syrup urine disease, the confirmation requires quantitation of the serum levels of branched-chain amino acids and urine levels of both the branched-chain amino acids and their ketoacids. Long-term management includes dietary restriction of the branched-chain amino acids. Frequent measurement of plasma concentrations of these amino

SECTION 17.3

Metabolism of Some Individual Amino Acids

acids is necessary to monitor the degree of dietary restriction and patient compliance. Many aminoacidurias and their metabolites give rise to abnormal odors, maple syrup urine disease is one example. Some of the others are phenylketonuria (musty odor), tyrosinemia type I (boiled cabbage), glutaric aciduria (sweaty feet), 3-methylcrotonylglycinuria (cat's urine), and trimethylaminuria (fish). In patients with trimethylaminuria the compound responsible for the fish odor is trimethylamine which is a byproduct of protein catabolism by the large intestinal bacterial flora. Normally, trimethylamine is inactivated by hepatic flavin monooxygenases. Several different mutations in the gene for flavin monooxygenases have been identified in trimethylaminuric patients. An inhibitor of flavin monooxygenases is indole-3-carbinol found in dark green vegetables (e.g., broccoli). The amelioration of symptoms of bad odor in trimethylaminuria may be achieved by limiting intake of dark green vegetables and protein, and by administering low doses of antibiotics to reduce intestinal bacterial flora.

353 NH2

NH2

I

I

+

Adenosine ~ S ~ (CH2);--CHmCOOH

Adenosine ~ S ~ (CH=)z~CH~ COOH /

CH3 S-Adenosylhomocysteine S-Adenosylmethionine . Specificmethyltransferase Acceptor (examples)

Acceptor ~ CH, (examples)

1. Guanidinoacetic acid 2. Nicotinamide 3. Norepinephrine 4. Phosphatidylethanolamine 5. N-Acetyl-serotonin

1. Creatine 2. N-Methylnicotinamide 3. Epinephrine 4. Phosphatidylcholine (three cycles of methylation) 5. Melatonin

FIGURE 17-15 Selected methyl transfer reactions involving S-adenosylmethionine.

The methyl group is transferred to appropriate acceptors by specific methyltransferases with production of S-adenosylhomocysteine (Figure 17-15), which is hydrolyzed to homocysteine and adenosine by adenosylhomocysteinase: NH3 +

NH3 +

I AdenosyI--S--(CH2)2--CHmCOO-

H20 Adenosine I ~ J : HS~(CH2)2~ CHmCOO-

S-Adenosylhomocysteine

Homocysteine

Sulfur-Containing Amino Acids

Methionine and cysteine are the principal sources of organic sulfur in humans. Methionine is essential (unless adequate homocysteine and a source of methyl groups are available), but cysteine is not, since it can be synthesized from methionine.

Homocysteine can be recycled back to methionine either by transfer of a methyl group from betaine catalyzed by betaine-homocysteine methyltransferase, or from N 5methyltetrahydrofolate (NS-methyl-FH4) catalyzed by N 5methyl-FH4-methyltransferase, which requires methyl cobalamin: (:)

NH3 +

!

Methionine

HS~(CH2)2~H~COO-

Methionine is utilized primarily in protein synthesis, providing sulfur for cysteine synthesis, and is the body's principal methyl donor. In methylation reactions, S-adenosylmethionine (SAM) is the methyl group donor. SAM is a sulfonium compound whose adenosyl moiety is derived from ATP as follows:

+ (CH3)3+N- CH2~COO-

Homocysteine

Betaine NH3 +

I

H3C__S__(Ch2)2mCH~COO- + (CH3)2N--CH2--COOMethionine

(2)

Dimethylglycine

NH 3+ I H S ~ ( C H 2 ) 2 ~ C H ~ C O O - + NS~MethyI~FH4

NH3 + I

ATP.H~

CH3~S~(CH2)2--CH~COO

Betaine-homocysteine methyltransferase ,

NS_Methyt_FH4.homocysteine methyltransferase (methylcobalamin)

~.spp, + p.

NH3 +

Methionine adenosyltransferase

I

Methionine

H3CmSm(CH2)2--CHmCOO - + FH4

NH2

Betaine (an acid) is obtained from oxidation of choline (an alcohol) in two steps"

NH3 +

I

FAD

- O O C - - C H - - ( C H 2 ) 2 - - ~;

CH2

I

I

(CH3)3+N__CH2--CH2OH Choline

CH3

~

FADH2

J

Cholineoxidase

,(CH3)3+N__CH2mCHO Betaine aldehyde NAD*~I '~

HO OH S-Adenosylmethionine (active methionine)

NADH +

Belaine aldehyde

9.,/1dehydr~ H+Wl

(CH3)3N__CH2mCOO Betaine

CHAPTER17 Protein and Amino Acid Metabolism

354

Cysteine In the biosynthesis of cysteine, the sulfur comes from methionine by transsulfuration, and the carbon skeleton and the amino group are provided by serine (Figure 17-16). Cysteine regulates its own formation by functioning as an allosteric inhibitor of cystathionine v-lyase, otKetobutyrate is metabolized to succinyl-CoA by way of propionyl-CoA and methylmalonyl-CoA. Cysteine is required for the biosynthesis of glutathione and of CoA-SH. A synthetic derivative, Nacetylcysteine, is used to replenish hepatic levels of glutathione and prevent hepatotoxicity due to overdosage with acetaminophen. When high concentrations of acetoaminophen are present in the liver, the drug undergoes N-hydroxylation to form N-acetyl-benzoquinoneimine, which is highly reactive with sulfhydryl groups of proteins and glutathione and causes hepatic necrosis. NAcetylcysteine is used as a mucolytic agent (e.g., in cystic fibrosis) because it cleaves disulfide linkages of mucoproteins. Cysteine and cystine are interconverted by NADdependent cystine reductase and nonenzymatically by an appropriate redox agent (e.g., GSH). The major end products of cysteine catabolism in humans are inorganic sulfate, taurine, and pyruvate. Taurine is a/3-amino acid that has a sulfonic acid instead of a L-Methionine M~tATP hionine adenosyltransferase PPI + P0 S-Adenosylmethionine [~,~.( n CH3)in methyltransferase reaction

S-Adenosylhomocysteine nosylhomocysteinase denosine HS~CH~-'CHi--~H-- COONH~

.

L -Hornocysteine

NH+

I/,-.- HOH,C----*CH--COO- L-Serine ~Cystathionine [~-synthase (pyridoxal phosphate)

L"--H,O

~H* *

[.

.

OOC--CHmCl-l=w S--CHi---Cl-.l=m C H ~ C OO-

I

NH+ L-Cystathionine H,O ~Cystathionine ~'-Iyase (pyridoxal phosphate) ["---. NH+ (cystathionase) NH+ OOCmCH--CH/-'-SH L-Cysteine

0

II

+ CH3----CH~---~_,mCOO---.,-~--~ Succinyl-OoA or

F I G U R E 17-16 Biosynthesis of cysteine. The sulfur is derived from methionine, and the carbon skeleton and amino group are derived from serine.

carboxylic acid group. Taurine is conjugated with bile acids in the liver (Chapter 19) and is readily excreted by the kidney. It is a major free amino acid of the central nervous system (where it may be an excitatory neurotransmitter) and the most abundant in the retina; it also occurs in other tissues (e.g., muscle, lung). Sulfate can be converted to the sulfate donor compound 3'-phosphoadenosine-5'-phosphosulfate (PAPS) in a twostep reaction (Figure 17-17). PAPS participates in the sulfate esterification of alcoholic and phenolic functional groups (e.g., in synthesis of sulfolipids and glycosaminoglycans).

Abnormalities Involving Sulfur-Containing Amino Acids Deficiencies of methionine adenosyltransferase, cystathionine fl-synthase, and cystathionine y-lyase have been described. The first leads to hypermethioninemia but no other clinical abnormality. The second leads to hypermethioninemia, hyperhomocysteinemia, and homocystinuria. The disorder is transmitted as an autosomal recessive trait. Its clinical manifestations may include skeletal abnormalities, mental retardation, ectopia lentis (lens dislocation), malar flush, and susceptibility to arterial and venous thromboembolism. Some patients show reduction in plasma methionine and homocysteine concentrations and in urinary homocysteine excretion after large doses of pyridoxine. Homocystinuria can also result from a deficiency of cobalamin (vitamin BI2) or folate metabolism. The third, an autosomal recessive trait, leads to cystathioninuria and no other characteristic clinical abnormality. Hereditary sulfite oxidase deficiency can occur alone or with xanthine oxidase deficiency. Both enzymes contain molybdenum (Chapter 27). Patients with sulfite oxidase deficiency exhibit mental retardation, major motor seizures, cerebral atrophy, and lens dislocation. Dietary deficiency of molybdenum (Chapter 37) can cause deficient activity of sulfite and xanthine oxidases. Cystinuria is a disorder of renal and gastrointestinal tract amino acid transport that also affects lysine, ornithine, and arginine. The four amino acids share a common transport mechanism (discussed above). Clinically, it presents as urinary stone disease because of the insolubility of cystine. In cystinosis, cystine crystals are deposited in tissues because of a transport defect in ATP-dependent cystine efflux from lysosomes (discussed above).

Homocysteine Homocysteine is an amino acid not found in proteins. Its metabolism involves two pathways; one is

355

Metabolism of Some Individual Amino Acids

SECTION 17.3

NH2

~ -

O

Sulfate

O---S--O

adenyl~transferase.-

''"~';'~"g~---'"

-

oli

ATP

-

N

O

II

II

O-- IIS'--O-- P - - O - - C H , / O

PP,

O

O]-

I~ I HO

I OH

Adenosine5"-phosphosulfate

Adenylylsulfate~. ATP

kinase~Mg''~ z/ "----*ADP NH=

O

II

\N~N

O

J

II

-O--S--O--P--O--CH2~O II I_ P~ I o

I O--P--OI

OH

O-

PAPS

FIGURE 17-17 Formationof 3'-phosphoadenosine-5'-phosphosulfate(PAPS).

the methylation of homocysteine to methionine using NS-methyltetrahydrofolate (NS-methyl-FH4) catalyzed by a vitamin B12-dependent enzyme. The second is the transsulfuration pathway where homocysteine condenses with serine to form cystathionine; this is catalyzed by cystathionine #-synthase (CBS) which is a pyridoxal5'-phosphate enzyme. End products of the transsulfuration pathway are cysteine, taurine, and sulfate (Figure 17-18). The methyl donor NS-methyl-FH4 is synthesized from NS,Nl~ and the reaction is catalyzed by NS,Nl~ reductase (MTHFR). MTHFR is a FAD-dependent enzyme. Thus, the metabolism of homocysteine involves four water soluble vitamins, folate, vitamin B12, pyridoxine, and riboflavin. Any deficiencies or impairment in the conversion of the four vitamins to their active coenzyme forms will affect homocysteine levels. Severe cases of hyperhomocysteinemia occur due to deficiencies of enzymes in the homocysteine remethylation or transsulfuration pathways. Individuals with a homozygous defect in cystathionine

#-synthase have severe hyperhomocysteinemia (plasma concentrations >50 #M/L) and their clinical manifestations are premature atherosclerosis, thromboembolic complications, skeletal abnormalities, ectopia lentis and mental retardation. In plasma, homocysteine is present as both free (< 1%) and oxidized forms (>99%). The oxidized forms include protein (primarily albumin)-bound homocysteine mixed disulfide (80-90%), homocysteine-cysteine mixed disulfide (5-10%), and homocystine (5-10%). Several studies have shown the relationship between homocysteine and altered endothelial cell function leading to thrombosis. Thus, hyperhomocysteinemia appears to be an independent risk factor for occlusive vascular disease. Five to ten percent of the general population have mild hyperhomocysteinemia. It has been shown that a thermolabile form of MTHFR is a major cause of mildly elevated plasma homocysteine levels, which have been associated with coronary heart disease. The thermolabile MTHFR gene has a

3,~

CHAPTER17 Protein and Amino Acid Metabolism

Folate (F) /i--2 NADPH+2H+ ihydrofolate reductase (DR)

~

~ "~ 2 NADP+ Tetrahydrofolate(FH4) ~ n f e Serine e hydroxymethyl rase oxal phosphate)

~

Glycine

Methylene FH4

/~

PP/+ Pi

//( Methionine / \Adenosyltransferase ( \ATP

I

I\ Methylated . i / \ accept~ 4~'- 1

Methylation

Methionine

MTHFR (FAD)

Methyl (acceptors

S-adenosylmethionine ~

~(~t

S-adenosylhomocysteine

Cycle

ethyl transferase

Methyl FH4 Adenosine ~

y l c o ~ a l a m i Hh i m o c y s t e i n e

enosylhomocysteinase

r gerine

fy

sti~tOhi xO~ipnheSspy~tathea)se

Cystathionine

Transulfuration Pathway

~ c~-Ketobutyrate

Cystathionine3'-lyase (pyridoxal phosphate)

Cysteine

/ / Glutathione

\ \ Taurine

,v_

-~ Sulfate

FIGURE 17-18 Homocysteine metabolism.

mutation of C to T at nucleotide position 677 which causes an alanine-to-valine amino acid substitution in the protein. The mechanism by which homocysteine mediates vascular pathology remains to be understood. The targets for homocysteine damage are connective tissue, endothelial cells, smooth muscle cells, coagulation factors, nitric oxide metabolism, plasma lipids and their oxidized forms (Chapter 20). Vitamin supplementation with B12, folate, and B6 has reduced total plasma homocysteine levels. Vitamin supplementation may decrease the morbidity and mortality from atherosclerotic vascular disease due to hyperhomocysteinemia. However,

further studies are required to assess the utility of vitamin supplementation.

Phenylalanine and Tyrosine Phenylalanine is an essential amino acid. Tyrosine is synthesized by hydroxylation of phenylalanine and therefore is not essential. However, if the hydroxylase system is deficient or absent, the tyrosine requirement must be met from the diet. These amino acids are involved in synthesis of a variety of important compounds, including thyroxine, melanin, norepinephrine, and epinephrine

357

SECTION17.3 Metabolism of Some Individual Amino Acids DIET




I

Dihydr0pteridine

Jl

Tyrosine

[f~(z -Ketoglutarate

I

[Tyrosinetransaminase

i I L_..7,8.Dihydrobiopterin~__~ pNADP* ~"NADPH+ H+

CH2-'---CH-- COO-

HO~

o

II

CH2--.-C--COO-

t

* p-Hydroxyphenylpyruvate

Hydroxylation,shiftp~p-HyC~roxyphenylpyruvic of the side chain I acid oxidase;ascorbic

decarboxylation~.~d~CoU'+

H O ~ O H COO

CH=

m

I CH II HC I

I I CH= I

CH2CO0

C--O

CO0

Homogentisicacid fO,

Homogentisicacid oxidase; ascorbic acid; GSH

CO0

Fumarate Acetoacetate H20[ Fumarylacetoacetase m

OOC

\c /

H

COO-

i

C ?H2 C H . . H/ \C \C / \CO0II 11

0

MaleylacetoacetateHC CHz .CH, isomerase \C / \C / \CO0-

0

Fumarylacetoacetate

II

0

It

0

Maleylacetoacetate

F I G U R E 17-20 Conversion of phenylalanine to tyrosine and the oxidative pathway of tyrosine.

6-pyruvoyl tetrahydrobiopterin synthase leads to hyper-

phenylalaninemia. Phenylketonuria (PKU) Deficiency of phenylalanine hydroxylase, tetrahydrobiopterin, or dihydropteridine reductase results in phenylketonuria (PKU), an autosomal recessive trait. Because phenylalanine accumulates in tissues and plasma (hyperphenylalaninemia), it is metabolized by alternative pathways and abnormal amounts of phenylpyruvate appear in urine (Figure 17-22). Phenylalanine hydroxylase deficiency may be complete (classic PKU, type I) or partial

(types II and III). Many mutations of the phenylalanine hydroxylase gene have been identified (missense, nonsense, insertions, deletions, and duplications) leading to PKU or non-PKU hyperphenylalaninemia. The incidence of classic PKU is about 1 in 10,00020,000 live births and exhibits considerable geographic variation (the incidence in Ireland is 1 in 4000, whereas the condition is rare among blacks and Asians). About 2% of hyperphenylalaninemic infants have a deficiency of biopterin or biopterin reductase. The most important clinical presentation is mental impairment. Diagnosis can be made early in the neonatal period by measurement of phenylalanine concentration in blood collected from

SECTION17.3 Metabolism of Some Individual Amino Acids H

I

H~ j N ~ jN~,../NH2 H/~]'~ 8I T "T H .~ II H3C~ ? ~ C /

"N"

/

OH OH

"hl"

H

!

R-group

5,6,7,8-Tetrahydrobiopterin H

I H~,,-N".-,,~ N"-,~NH

H~N~..-N~I,./NH2

O

O

Ortho-quinonoid dihydropteridine

Para-quinonoid dihydropteridine

"Quinonoid"dihydrobiopterins H

I

!~i I~'~NHN~NH2 O 7.8-Dihydrobiopterin F I G U R E 17-21 Structures of biopterin derivatives.

a heel prick onto filter paper. Treatment of phenylalanine hydroxylase deficiency consists of a diet low in phenylalanine but which maintains normal nutrition. This diet is effective in preventing mental retardation, and its con~H+ Transaminase @ CHu--CH-- COO~ (pyridoxal phosphate) y,.-~ " -Ketoglutarate Glutamate Phenylalanine ~ NAD+ ~

~'~

CH2COO-

0 II COOCH;---C--

~

Phenylpyruvate NADH+ H+

NAD+

C02

Phenylacetate

OH Phenyllactate

/

Inthei"'-- Glutamine liver~...--H20 /!

\\

0 H COOII , I

~k__/~---OH2---C--N--(~H

CONH2 Phenylaeelyk:jlutamine F I G U R E 17-22 Formation of metabolites of phenylalanine that accumulate in abnormal amounts and are excreted in phenylketonuria.

359

tinuation throughout the first decade, or for life, may be necessary. Treatment of biopterin and biopterin reductase deficiency consists not only of regulating the blood levels of phenylalanine but of supplying the missing form of coenzyme and the precursors of neurotransmitters, namely, dihydroxyphenylalanine and 5-hydroxytryptophan, along with a compound that inhibits peripheral aromatic decarboxylation. This compound is necessary because the amine products do not cross the blood-brain barrier. Successfully treated females who have reached reproductive age may expose their offspring (who are obligate heterozygotes) to abnormal embryonic and fetal development. These effects include spontaneous abortion, microcephaly, congenital heart disease, and intrauterine growth retardation, and they correlate with the plasma level of phenylalanine of the pregnant mother. Thus, reinstitution of a low-phenylalanine diet during preand postconception periods may be necessary. The diet should also restrict intake of phenylalanine-containing substances, such as the synthetic sweetener aspartame (Laspartyl-L-phenylalanyl methyl ester). Because defective myelination occurs in the brain in PKU, there is an increased incidence of epileptic seizures and abnormal electroencephalograms are common. The biochemical basis for the severe mental impairment is not understood. One factor may be inhibition of glutamate decarboxylase by phenylpyruvate and phenylacetate: NH3 +

I

- OOC--(CH2)2--CH--COO-

Glutamate decarboxylase

CO2

L-Glutamate -

+

OOC--(CH2)3--NH 3 7-Aminobutyrate

The substrate is an excitatory neurotransmitter and the product an inhibitory one in the central nervous system." Abnormal indole derivatives in the urine and low levels of serotonin (a product of tryptophan metabolism) in blood and brain point to a defect in tryptophan metabolism in PKU. 5-Hydroxytryptophan decarboxylase, which catalyzes the conversion of 5-hydroxytryptophan to serotonin, is inhibited in vitro by some of the metabolites of phenylalanine. Phenylalanine hydroxylase is similar to the enzyme that catalyzes the hydroxylation of tryptophan to 5-hydroxytryptophan, a precursor of serotonin. In vitro, phenylalanine is also found to inhibit the hydroxylation of tryptophan. The mental defects associated with PKU may be caused by decreased production of serotonin. High phenylalanine levels may disturb the transport of amino

360 acids into cells. Variations in the clinical manifestations of PKU may reflect differences in this disturbance. Defects in pigmentation of skin and hair (light skin and hair) may be caused by interference of melanin formation by phenylalanine and its metabolites and also by lack of tyrosine.

CHAPTER17 Proteinand AminoAcid Metabolism ~'~--C--COO-

Tyrosinase

--CO0-

[O1 H,O H Tyrosine

N/

3,4-Dihydroxyphenylalanine (dopa) [O]-'~ Tyrosinase

H.O~ 1 Melanin Melanin is an insoluble, high-molecular-weight polymer of 5,6-dihydroxyindole, which is synthesized from tyrosine (Figure 17-23). It is produced by pigment cells (melanocytes) in cytoplasmic organelles (melanosomes). In the epidermis, melanocytes are associated with keratinocytes, which contain melanosomes supplied by melanocytes via dendritic processes. Color variation in human skin reflects the amount of melanin synthesized in melanosomes. Melanin synthesis is apparently under hormonal and neural regulation. The first two steps in the synthesis of melanin are catalyzed by tyrosinase, a copper-containing oxidase, which converts tyrosine to dopaquinone. All subsequent reactions presumably occur through nonenzymatic auto-oxidation, in the presence of zinc, with formation of the black to brown pigment eumelanin. The yellow to reddish brown, high-molecular-weight polymer known as pheomelanin and the low-molecular-weight trichromes result from addition of cysteine to dopaquinone and further modification of the products. Pheomelanins and trichromes are primarily present in hair and feathers.

CO0_---.,---,,--O.,j,~~.,j+I~N/I IH--C--CO0H Dopachrome

co2J zn'+

H Indole-5,6-quinone Cysteine,,......,,I.[

H

5,6-Dihydroxyindole Trichromes

/

/

Pheomelanins

Abnormalities of Tyrosine Metabolism Hepatic cytosolic tyrosine aminotransferase (tyrosine transaminase) deficiency produces tyrosinemia type II, an autosomal recessive trait marked by hypertyrosinemia and tyrosinuria. Clinical manifestations may include corneal erosions and plaques, inflammation (from intracellular crystallization of tyrosine), and mental retardation. Low-tyrosine and low-phenylalanine diets are beneficial. Tyrosinosis is presumably due to fumarylacetoacetate hydrolase deficiency and has a high prevalence in the French-Canadian population of Qu6bec. It is associated with abnormal liver function, renal tubular dysfunction, anemia, and vitamin D-resistant rickets. Transient tyrosinemia of the newborn, particularly in premature infants, is the most common form of tyrosinemia in infancy. Alcaptonuria is a rare metabolic hereditary disease in which homogentisic acid is eliminated in urine, which darkens upon exposure to air owing to oxidation of

Dopaquinone

1

Polymerization [

H N

O

N~c

0

Eumelanins F I ( ; U R E 17-23 Biosynthesis of melanins.

homogentisic acid. The biochemical lesion is homogentisic acid oxidase deficiency. Clinical features include pigmentation of cartilage and other connective tissues (ochronosis) later in life from deposition of oxidized homogentisic acid. Patients nearly always develop arthritis in later years, but the relationship between pigment deposition and arthritis is not understood. Lack of melanin production (hypomelanosis) gives rise to several hereditary disorders collectively known as albinism. Some forms result from deficiency of tyrosinase. The inheritance pattern of albinism varies with type. Affected individuals have increased susceptibility to various

SECTION17.3 Metabolismof Some Individual Amino Acids

361 O

pyrrolase, CH;---CHmCOO- Tryptophan f

E ~

NIH:

02

II

E~IH~I~--i7

I

COO

N-Formylkynurenine

H

f-H,O

Formamidase Formate~ One-carbonpool

_CmCH2--'-CH--COO"~ I'Y ] ~. NH.~ ' 'I , k,.~ ~NH= i;

Z"

3-Hydroxykynurenine

o2~ N A D P H + H +

""~eeb~, ~

Kynureninase H20 (pyridoxal~f'ph~ ~'~Alanine

~

Kynurenine hydroxylase

'

,~"~C--

I-

II

~_,

I~

~

CHs ICHmCOO

".§

....3

"NH2 Kynurenine

o/,. ~J"Ob'o ~e/~ ~O~,o/, -" ~

OH

CO0 ....

NAD+

~NH+ OH

3-Hydroxyanthranilate

I~/,~CO0OH

Xanthurenate (excretedin urine)

FIGURE 17-24 Pathway for the synthesis of NAD from tryptophan.

types of carcinoma (from the effect of solar radiation on DNA). When the eyes are involved, photophobia, subnormal visual acuity, strabismus, and nystagmus may be present.

Tryptophan

Tryptophan is an essential amino acid involved in synthesis of several important compounds. Nicotinic acid (amide), a vitamin required in the synthesis of NAD + and NADP +, can be synthesized from tryptophan (Figure 17-24). About 60 mg of tryptophan can give rise to 1 mg of nicotinamide. The synthesis begins with conversion of tryptophan to N-formylkynurenine by tryptophan pyrrolase, an inducible iron-porphyrin enzyme of liver. N-Formylkynurenine is converted to kynurenine by removal of formate, which enters the one-carbon pool. Kynurenine is hydroxylated to 3-hydroxykynurenine, which is converted to 3-hydroxyanthranilate, catalyzed by kynureninase, a pyridoxal phosphate-dependent enzyme. 3-Hydroxyanthranilate is then converted by a series of reactions to nicotinamide ribotide, the immedi-

ate precursor of NAD. In deficiency of pyridoxal phosphate, 3-hydroxykynurenine accumulates and is converted to xanthurenate, which is excreted in urine. Thus, vitamin B6 deficiency can be diagnosed by measurement of urinary xanthurenate after administration of a standard dose of tryptophan (tryptophan load test). 5-Hydroxytryptamine (serotonin) is found in enterochromaffin cells, brain, and platelets. In the former two, it is produced from tryptophan, whereas in platelets, serotonin is taken up from plasma. Synthesis involves hydroxylation of tryptophan by tryptophan 5-hydroxylase and decarboxylation by aromatic L-amino acid decarboxylase (Figure 17-25). Hydroxylation is the rate-limiting reaction, is analogous to that of phenylalanine, and requires molecular oxygen and tetrahydrobiopterin. Serotonin is a powerful vasoconstrictor and stimulator of smooth muscle contraction. In the brain it is a neurotransmitter, and in the pineal gland it serves as a precursor of melatonin. Synthesis of melatonin requires N-acetylation of serotonin, followed by methylation (Figure 17-26). The role of melatonin in humans is not understood; in frogs, it lightens the color of skin melanocytes and blocks the action of melanocyte-stimulating hormone (MSH) and

362

CHAPTER17 Protein and Amino Acid Metabolism

(.CHCH--COO~H;~

I

H

Tryptophan 02, tetrahydrobiopterin~1 H20, dihydrobiopterin..~Tryptophan-5-hydroxylase

HO~,~CHr--CH--COO

-

I

N

HO.

5-Hydrox~ryptophan Aromatic L-aminoacid r,~ .,.,~decarboxylase "'=t(pydd~ phosphate)

I

I

N

5-Hydroxytryptamine (serotonin) FIGURE 17-25 Biosynthesis of serotonin from tryptophan.

H O . ~

II--CHE--CHs NH+ jlJ Serotonin

?

H AcelyI'C~ 4 5-Hydrox~ryptamine-

HO~

CoASH~'~N-acetyltransferase

O

II

CHT'-CHu--NH~C~CH 3 N-Acetyl-5-hydroxytryptamine

9 H S-Adenosylmethionine-~

S.Adenosylhomocysteine.~,.[Hydroxyindolemethyltransferase O

j

CHE---CHs

N

I

H

Melatonin (N-acetyl-5-methoxytryptamine) FIGURE 17-26 Biosynthesis of melatonin from serotonin.

"

adrenocorticotropic hormone (ACTH). In rats, melat0nin regulates the breeding cycle, and its secretion is increased by exposure to light via adrenergic stimulation of the pineal gland. N-Acetyltransferase is activated by increased concentrations of cytosolic cyclic AMP and Ca 2+ that is mediated by the activation of adrenergic receptors of the pineal gland. In humans, melatonin synthesis and its release follows a circadian rhythm which is stimulated by darkness and inhibited by light. The blood levels of melatonin increase by passive diffusion from the central nervous system after the onset of darkness, reaching a peak value during the middle of the night and declining during the second half of the night. Melatonin secretion is also regulated endogenously by signals from the suprachiasmatic nucleus. Since melatonin's peak concentration in plasma coincides with sleep, exogenous administration of the hormone can affect the circadian rhythm. Melatonin supplementation has been used to ameliorate subjective and objective symptoms of jet lag caused by travel across time zones. At high levels, melatonin promotes sleep. Short-term and longterm biological effects of melatonin supplementation have yet to be determined. Serotonin is degraded to 5-hydroxyindoleacetic acid (5-HIAA) by monoamine oxidase and aldehyde dehydrogenase acting in sequence. 5-HIAA is excreted in the urine. Its excretion is markedly increased in subjects with carcinoid tumor (found most frequently in the gastrointestinal tract and lungs). Carcinoid tumors are frequently indolent and asymptomatic; however, a significant number of these tumors can manifest as metabolic problems. Although increased production of serotonin is a characteristic feature of the carcinoid tumor, cells also synthesize other substances. These include kinins, prostaglandins, substance P, gastrin, somatostatin, corticotropin, and neuron-specific enolase. Carcinoid tumor cells are known as enterochromaffin cells because they stain with potassium dichromate and also are known as argentaffin cells because they take up and reduce silver salts. The symptoms of the carcinoid tumor are due to synergistic biochemical interactions between serotonin and the above-mentioned active metabolites. Characteristics of the carcinoid syndrome are flushing, diarrhea, wheezing, heart valve dysfunction, and pellagra. Lifestyle conditions that precipitate symptoms include intake of alcohol or spicy foods, and strenuous exercise. The treatment options for the carcinoid syndrome are multidisciplinary, including lifestyle changes, inhibitors of serotonin release and serotonin receptor antagonists, somatostatin analogues, hepatic artery embolization, chemotherapy, and surgery.

Supplemental Readings and References

Hartnup disease is a disorder of renal tubular and intestinal absorption of tryptophan and other neutral amino acids.

Supplemental Readings and References J. Abrains: The role of nitrates in coronary heart disease. Archives oflnternal Medicine 155, 357 (1995). M. L. Batshaw: Inborn errors of urea synthesis. Annals of Neurology 35, 133 (1994). D. Bohn: Nitric oxide in acute hypoxic respiratory failure: From the bench to the bedside and back again. Journal of Pediatrics 134, 387 (1999). A. Brezezinski: Melatonin in Humans. The New England Journal of Medicine 331, 186 (1997). M. E. Caplin, J. R. Buscombe, A. J. Hilson, et al.: Carcinoid tumour. Lancet 352, 799 (1998). K. J. Carpenter: Protein requirements of adults from an evolutionary perspective. American.Journal of Clinical Nutrition 55, 913 (1992). D. Christensen: What's that smell? Science News 155, 316 (1999). D. T. Chuang: Maple syrup urine disease: It has come a long way. Journal of Pediatrics 132, S 17 (1998). J. E Cooke and E S. Tasao: Arginine: A new therapy for artherosclerosis? Circulation 97, 311 (1997). B. R. Crane, A. S. Arvai, R. Gachhui, et al.: The structure of nitric oxide synthase oxygenase domain and inhibitor complexes. Science 278, 425 (1997). E. L. Dobyns, D. N. Cornfield, N. G. Anas, et al.: Multicenter randomized control trial of the effects of inhaled nitric oxide therapy on gas exchange in children with acute hypoxic respiratory failure. Journal of Pediatrics 134, 406 (1999). E. B. Feldman: Creatine: A Dietary supplement and ergogenic aid. Nutrition Reviews 57, 45 (1999). I. Goldstein, T. E Lue, H. Padma-Nathan, et al.: Oral sildenafil in the treatment of erectile dysfunction. The New England Journal of Medicine 338, 1397 (1998).

363 R. Jalan and E C. Hayes: Hepatic encephalopathy and ascites. Lancet 350, 1309 (1997). S. G. Korenman: New insights into erectile dysfunction. American Journal of Medicine 105, 135 (1998). J. Loscalzo and G. Welch: Nitric oxide and its role in the cardiovascular system. Progress in cardiovascular diseases 38, 87 (1995). N. E. Maestri, D. Clisold, and S. W. Brusilow: Neonatal onset ornithine transcarbamylase deficiency: A retrospective analysis. Journal of Pediatrics 134, 268 (1999). M. M. Malinow, E B. Duell, D. L. Hess, et al: Reduction of plasma homocysteine levels by breakfast cereal fortified with folic acid in patients with coronary heart disease. The New England Journal of Medicine 338, 1009 (1998). A. J. Maxwell and J. E Cooke: Cardiovascular effects of L-arginine. Current Opinion in Nephrology and Hypertension 7, 63 (1998). D. J. Millward: Aging, protein requirements, and protein turnover. American Journal of Clinical Nutrition 66, 774 (1997). J. D. Parker and J. O. Parker: Nitrate therapy for stable angina pectoris. The New England Journal of Medicine 338, 520 (1998). S. M. Riordan and R. Williams: Treatment of hepatic encephalopathy. The New England Journal of Medicine 337, 473 (1997). A. M. Spiekerman: Proteins used in nutritional assessment. Clinical Laboratory Medicine 13, 353 (1993). J. H. Stein and E E. McBride: Hyperhomocysteinemia and atherosclerotic vascular disease. Archives of Internal Medicine 158, 1301 (1998). J. G. Theone: Treatment of urea cycle disorders. Journal of Pediatrics 134, 255 (1999). R. D. Utiger: A pill for impotence. The New England Journal of Medicine 338, 1458 (1998). G. Wagner and I. S. de Tejada: Update on male erectile dysfunction. British Medical Journal 376, 678 (1998). N. J. Wald, H. C. Watt, M. R. Law, et al.: Homocysteine and ischemic heart disease. Archives of Internal Medicine 158, 862 (1998). G. N. Welch and J. Loscalzo: Homocysteine and atherothrombosis. The New England Journal of Medicine 338, 1042 (1998). M. Wyss, R. Kaddurah-Daouk: Creatine and creatine metabolism. Physiological Reviews 80, 1107 (2000).

CHAPTER

18

Lipids I: Fatty Acids and Eicosanoids

Lipids (or fats) are a heterogeneous group of organic compounds defined by their solubility in nonpolar solvents such as chloroform, ether, and benzene and by their poor solubility in water. Lipids may be polar or nonpolar (amphipathic). Polar lipids have limited solubility in water because they are amphipathic, i.e., they possess hydrophilic and hydrophobic regions in the same molecule. Major polar lipids include fatty acids, cholesterol, glycerophosphatides, and glycosphingolipids. Very short chain fatty acids and ketone bodies are readily soluble in water. Nonpolar lipids serve principally as storage and transport forms of lipid and include triacylglycerols (also called triglycerides) and cholesteryl esters. Lipids have numerous functions including the following: thermal insulation, energy storage (as triacylglycerol), metabolic fuels, membrane components (phospholipids and cholesterol; Chapter 10), hormones (steroids and vitamin D metabolites; Chapters 32 and 37, respectively), precursors of prostanoids (discussed on p. 391) and leukotrienes, (vitamins A, C, D, E, and K; Chapters 36-38), emulsifying agents in the digestion and absorption of lipids (bile acids; Chapters 12 and 19), and surfactants in the alveolar membrane (phosphatidylcholine; Chapter 19). The metabolism of fatty acids (saturated and unsaturated) is discussed in this chapter. The metabolism of phospholipids, glycosphingolipids, and cholesterol is considered in Chapter 19.

Fatty acids that contain no carbon-carbon double bonds are known as saturated and those with carbon-carbon double bonds as unsaturated. Fatty acids that contain an even number of carbon atoms and are acyclic, unbranched, nonhydroxylated, and monocarboxylic make up the largest group. The most abundant saturated fatty acids in,animals are palmitic and stearic acids (Table 18-1). The melting point of fatty acids rises with increase in chain length, the even-numbered saturated fatty acids having higher melting points than the odd-numbered. Among the evennumbered, the presence of cis double bonds lowers the melting point significantly. Free fatty acids at physiological pH are ionized (pK ~4.85) and exist only in small quantities; in plasma, they typically are bound to albumin. They are usually present as esters or amides. Digestion and absorption of lipids are discussed in Chapter 12. The Western diet contains about 40% fat, mostly as triacylglycerol (100-150 g/day). Triacylglycerols packaged as chylomicrons in the intestinal epithelial cell are delivered to the blood circulation via the lymphatic system and are hydrolyzed to glycerol and fatty acids by endothelial lipoprotein lipase. Fatty acids are taken up by the cells of the tissue where the hydrolysis occurs, whereas glycerol is metabolized in the liver and kidney (Chapter 15). Another means of triacylglycerol transport is very-low-density lipoprotein (VLDL), which is synthesized in the liver. Its triacylglycerol is also hydrolyzed by endothelial lipoprotein lipase. The metabolism of plasma

365

366

CHAPTER 18

Lipids I: Fatty Acids and Eicosanoids

TABLE 18-1

Naturally Occurring Saturated Fatty Acids Common Name Capric Lauric Myristic Palmitic t Stearic t Arachidic Behenic Lignoceric C erotic Montanic

Systematic Name*

Molecular Formula

n-Decanoic n-Dodecanoic n-Tetradecanoic n-Hexadecanoic n-Octadecanoic n-Eicosanoic n-Docosanoic n-Tetracosanoic n- Hexaco s anoic n-Octacosanoic

C 10H2002 C 12H2402 C 14H2802 C 16H3202 C 18H3602 C20H400 2 C22H440 2 C24H4802 C26H5202 C28H560 2

Structural Formula CH3[CH2]8COOH CH3[CH2] loCOOH CH3[CH2] 12COOH CH3[CH2] 14COOH CH3[CH2] 16COOH CH3[CH2] 18COOH CH3[CH2]20COOH CH3[CH2122COOH CH3[CHz]z4COOH CH3[CH2126COOH

Melting Point (~ 31 44 58 63 70 76 80 84 88 92

*Systematic name is based on replacingthe final letter "e" of the parenthydrocarbonwith"oic." tMost abundantfattyacidspresentin animallipids. lipoproteins is discussed in Chapter 20. Fatty acids are released by hydrolysis of triacylglycerol in adipocytes by hormone-sensitive lipase, particularly during starvation, stress, and prolonged exercise (Chapter 22). Interrelationships of tissues in lipid metabolism are discussed in Chapter 22.

fully assembled, mature enzymes. During/~-oxidation of acyl-CoA, the chain length of the substrate is shortened by two carbon atoms (acetyl-CoA) each cycle. Thus, ,6oxidation requires a group of enzymes with chain length specificity.

Activation of Fatty Acids

18.1 Oxidation of Fatty Acids The overall fatty acid oxidation process in mitochondria consists of uptake of fatty acids, their activation to acylCoA, then to thioesters, and finally translocation into mitochondria which involves a carnitine transesterification shuttle and/3-oxidation. Fatty acids released from chylomicrons and VLDLs are transferred across cell membranes by passive diffusion, which depends on the concentration gradient. Fatty acids are also obtained from the hydrolysis of triacylglycerol stored in adipose tissue which are bound to albumin and transported in blood. Fatty acids serve as substrates for energy production in liver, skeletal and cardiac muscle during periods of fasting. Although the brain does not utilize fatty acids for generating energy directly, brain cells can utilize ketone bodies synthesized from acetyl-CoA and acetoacetyl-CoA. The latter two are obtained from/3-oxidation of fatty acids in the liver. All of the enzymes involved in mitochondrial fatty acid/3oxidation are encoded by nuclear genes. After their synthesis in the cytosolic endoplasmic reticulum, the enzymes are transported to mitochondria. The transport of the enzymes into mitochondria, in many instances, requires the presence of N-terminal extensions to guide the protein across the mitochondrial membrane, receptor-mediated ATP-dependent uptake, and proteolytic processing to form

At least three acyl-CoA synthases, each specific for a particular size of fatty acid, exist: acetyl-CoA synthase acts on acetate and other low-molecular-weight carboxylic acids, medium-chain acyl-CoA synthase on fatty acids with 4-11 carbon atoms, and acyl-CoA synthase on fatty acids with 6-20 carbon atoms. The activity of acetyl-CoA synthase in muscle is restricted to the mitochondrial matrix. Medium-chain acyl-CoA synthase occurs only in liver mitochondria, where medium-chain fatty acids obtained from digestion of dietary triacylglycerols and transported by the portal blood are metabolized. Acyl-CoA synthase, the major activating enzyme, occurs on the outer mitochondrial membrane surface and in endoplasmic reticulum. The overall reaction of activation is as follows: RCOO- + A TP 4- + CoASH ~

O II

R C - - S C o A + AMP 2- + PPi 3AcyI-CoA (a thioester)

The reaction favors the formation of fatty acyl-CoA, since the pyrophosphate formed is hydrolyzed by pyrophosphatase: PPi 4-H20--+ 2Pi. Thus, activation of a fatty

367

SECTION18.1 Oxidation of Fatty Acids

acid molecule requires expenditure of two high-energy phosphate bonds. The reaction occurs in two steps (E -enzyme)"

The overall translocation reaction is as follows: O

OH3

II

I

RBCmSCoA + H3C u N +~CH2~CHmCH2--COOH

I

RCOO- + ATP + E AcyI-CoA

Step 1

OH Carnitine

[Carnitpal inemitoyltransferase

O

II

E~AMPmCmR

CoASH~ ~

I

CH3

+

PPi OH3

I

H

H3C--N +--CH2--CmCH2mCOOH + CoASH

Step 2

I

OH3

O

I

O

I

C--O }

II

E + RmC~SCoA + AMP

I R

A mitochondrial acyl-CoA synthase, which utilizes GTP, has also been identified:

Acyl group

The standard free-energy change of this reaction is about zero, and therefore the O-ester bond of acylcarnitine may be considered as a high-energy linkage. Malonyl-CoA, a precursor in the synthesis of fatty acids, is an allosteric inhibitor of CPTI in liver and thus prevents a futile cycle of simultaneous fatty acid oxidation and synthesis. Carnitine is synthesized from two essential amino acids, lysine and methionine. S-Adenosylmethionine donates three methyl groups to a lysyl residue of a protein with the formation of a protein-bound trimethyllysyl. Proteolysis yields trimethyllysine, which is converted to carnitine (Figure 18-2). In humans, liver and kidney are major sites of carnitine production; from there it is transported to skeletal and cardiac muscle, where it cannot be synthesized. Four inherited defects of carnitine metabolism lead to impaired utilization of long-chain fatty acids for energy production. These include defects of plasma

R. COO- + CoASH + GTP 4- --+ RCO. SCoA + GDP 3- + p2Its role is not known.

Transport of AcyI-CoA to Mitochondrial Matrix This transport is accomplished by carnitine (L-/3-hydroxyg-trimethylammonium butyrate), which is required in catalytic amounts for the oxidation of fatty acids (Figure 18-1). Carnitine also participates in the transport of acetylCoA for cytosolic fatty acid synthesis. Two carnitine acyltransferases are involved in acyl-CoA transport: carnitine palmitoyltransferase I (CPTI), located on the outer surface of the inner mitochondrial membrane, and carnitine palmitoyltransferase II (CPTII), located on the inner surface.

Mitochondria

,,k.

f

Cytosol

Outer membrane

O

II

C~

RBC--OH

+ATP

Intermembranal space / /

AcyI-CoA

synthetase

H

AMP + PP~~J~

d

~)

9

I I

Outer surface

1"-

Carnitine palmitoyl

acyltransferaseI

9R--C--SCoA - - ~ (acyI-CoA)

Inner membrane 9Acyl-carnitine [acylcarniti-

~

Inner surface

Matrix

CoASH Carnitine palmitoyl transferase II

,,---..AcyliCoA 13-Oxidation

F I G U R E 18-1 Role of carnitine in transport of fatty acids into the mitochondrial matrix. Entry of acyl-carnitine is linked to the exit of carnitine, both mediated by a translocase.

368

CHAPTER 18

ec-Ketoglutarate

NH2

I (C~)3~N--[CHJ;-"CH--COOH

Succinate

+ O2~/+

CO2

NH2

I

"~-"Ascorbate, " (CH3~N--[CH2];--~H--CH--COOH Fe2+

Succinate

OH

13-Hydroxy-r

-N-Trimethyllysine

H

ot-Ketoglutarate

+ COz

+ 02

glycine

NADH + H+

NAD+

(C..).---~--CH~~--CHr-COO.-k,,~~ k,,j/ Ascorbate, (CH.).~tC.,],---COOH 3 OH Carnitine

Lipids I: Fatty Acids and Eicosanoids

Fe=+

, Pyridoxal

~phosphate

%

1C~:~--~--[CHz]~ - CHO

" 7-Butyrobetaine

Y-Butyrobetainealdehyde

FIGURE 18-2 Carnitine biosynthesisin humans.A lysyl residueis trimethylatedby S-adenosylmethionine,with subsequent proteolytic releaseof trimethyllysine,the starting material.The reactionsare catalyzedby (1) trimethyllysine /~-hydroxylase,(2)/3-hydroxy-trimethyllysinealdolase(pyridoxalphosphate),(3) y-trimethylaminobutyraldehyde dehydrogenase,and (4) y-butyrobetainehydroxylase.

membrane carnitine transport, carnitine palmitoyltransferase I (CPTI), carnitine palmitoyltransferase II (CPTII), and carnitine-acylcarnitine translocase. Clinical manifestations of disorders of carnitine metabolism and fatty acid oxidation disorders (discussed later) span a wide spectrum and can be affected by the severity and site of the defect (e.g., muscle, liver, and kidney). The disorders may be characterized by hypoketotic hypoglycemia, hyperammonemia, liver disease, skeletal muscle weakness, and cardiomyopathy. In some instances, dietary intervention brings about marked improvement in the clinical manifestations; for example, patients with carnitine transport defect respond well to carnitine therapy. /3-Oxidation The major pathway for fatty acid oxidation,/3-oxidation (Figure 18-3), involves oxidation of acyl-CoA at the/3carbon and removal of two carbon fragments as acetylCoA and takes place entirely in the mitochondrial matrix. Oxidation of a saturated acyl-CoA with an even number of carbon atoms to acetyl-CoA requires repeated sequential action of four enzymes. 1. Acyl-CoA dehydrogenase dehydrogenates acyl-CoA at the oe- and/3-carbon atoms to yield the ot,/3-unsaturated acyl-CoA (or AZ-unsaturated acyl-CoA). Each one of four distinct dehydrogenases is specific for a given range of fatty acid chain length. All four are flavoproteins and contain a tightly bound molecule of flavin adenine dinucleotide (FAD). The electrons from the acyl-CoA dehydrogenase are

transferred to the main respiratory chain (Chapter 14) through mitochondrial electron transfer flavoprotein (ETF) and ETF-ubiquinone oxidoreductase (ETF-QO) (Figure 18-4). Both ETF and ETF-ubiquinone oxidoreductase are nuclear encoded proteins. They also mediate transfer of electrons from dimethylglycine dehydrogenase and sarcosine dehydrogenase. Inherited defects in ETF and ETF-QO cause accumulation of organic acids (acidemia) and their excretion in the urine (acidurias) Examples of these disorders are glutaric acidemia type I and type II which are inherited as autosomal recessive traits. Glutaric acid is an intermediate in the metabolism of lysine, hydroxy lysine, and tryptophan. Glutaric acidemia type I is caused by deficiency of glutaryl-CoA dehydrogenase which catalyzes the conversion of glutaryl-CoA to crotonyl-CoA. Glutaric acidemia type H is caused by defects in the ETF/ETF-QO proteins. The clinical manifestations of these disorders are similar to medium-chain acyl-CoA dehydrogenase deficiency (discussed later). The A 2 double bond formed by the acyl-CoA dehydrogenase has a trans configuration. The double bonds in naturally occurring fatty acids are generally in the cis configuration. The oxidation of unsaturated cis-fatty acids requires two auxiliary enzymes, enoyl-CoA isomerase and 2,4-dienoyl-CoA reductase. Acyl-CoA dehydrogenase (especially butyryl-CoA dehydrogenase) is irreversibly inactivated by methylene cyclopropylacetyl-CoA through the formation of covalent adduct with the FAD of the enzyme. The inhibitor is derived by transamination

SECTION18.1 Oxidation of Fatty Acids

369

O

, II R~CHi---CH2--C--OH

(A fatty acid)

CoASH-----~ / ATP ~ A c y I - C o A ..... M~ ~synthetase ~ M ~ + r'~ ~---[

t~'tE t - t . O~ . O u)

>,__

Many thiokinases are known. They differ in their substrate specificity and occur in the outer membrane of the mitochondria and microsomes.

O R~CH2---CHs

II

ITranslocation to mit(x~hondrial [matri x involving carnitine Mitochondrial matrix [

O

II (AcyI-CoA) _- R R CH2---- CH~----C ~ S ~ C o A A I "" A t ' ~ - - F P ~ ..---* Red. carriers ~

( /

(~

~ - 1 / 2 02, 2ADP, 2Pa

d,~h,,droCYen~a~ ~ (respiratory chain) ~ ~ Y ~ ~"--* FPH2"~" "~ Ox. carriers = 1 .,..._.31-1=O,2ATP trans

R~ CH--CH~C~S~CoA

(0r ~-Unsaturated acyI-CoA or z~?.-unsaturated acyI-CoA)

EnoyI-CoA~- H20 @

hydratase[

OH

II

R--CHmCH~---C~SmCoA (L (+)-13-hydroxy acyI-CoA)

= O "-,~

-o ~ 0

O

I

,-I-

9

" I " A II r( NAD = --Y t - -.- Red L-13-Hyaroxyacy-L;o . " .carriers . . . ~ . . . .v- - - - 1 / 2 02 ' 3ADP, 3P~ dehydrogenase~ . . . . . . . . . + . j , ~ i,resplralory, c n a l n ) ~ ~ . ~ ~ ----N,,~url + N ux. carriers 4H20, 3ATP

(~

cCL

R--C--

@

CH~--- C ~ S m C o A

i

3-Ketoacyl-CoA

TCA cycle;

L ""*CH~---C~SmCoA

thi~

~

respiratory chai n.... 2CO2 + 12ATP

(acetyI-CoA) ~

)

'-

R-- C~S~CoA Fatty acyi-CoA, two carbons shorter

Ketone bodies (in liver)

"

To cytosoi, for cholesterol synthesis

FIGURE 18-3 Fatty acid activation, transport, and/~-oxidation. The shortened fatty acyl-CoA from one cycle is further oxidized in successive passes until it is entirely converted to acetyl-CoA. Odd-chain fatty acids produce one molecule of propionyl-CoA. Ox. = Oxidized; Red. = reduced; respiratory chain = oxidative phosphorylation and electron transport; = high-energy bond; FP = flavoprotein.

and oxidative decarboxylation of the amino acid hypoglycin (Chapter 15). Ingestion of hypoglycin causes severe hypoglycemia due to the inhibition of /~-oxidation and corresponding decrease in ATP synthesis. Gluconeogenesis, which is important in maintaining fasting glucose levels, is dependent on adequate supplies of ATE The action of hypoglycin thus serves to emphasize the importance of /~-oxidation in gluconeogenesis under normal circumstances.

Among the fatty acid oxidation disorders,

medium-chain acyl-CoA dehydrogenase deficiency (MCAD) is the most common and its frequency is similar to that of phenylketonuria. The disorder can be identified by mutant alleles and some key abnormal metabolites. An A --+ G transition mutation occurs at position 985 of MCAD-cDNA in about 90% of cases. This mutation leads to replacement of lysine with glutamate at position 329 (K329E) of the polypeptide.

370

CHAPTER 18

Lipids I: Fatty Acids and Eicosanoids

Fatty acids I Activati~ t~ C~ derivatives } ~ ~ . . Carnitine mediated translocation into mitochondria

ISites of Defects]

Fatty acids CoA Mitochondrial electron transfer flavoprotein (ETF) and ETF-Ubiquinone oxidoreductase ~ - - NADH

I /

]Main mitochondrial respiratory chain I ATP

Turns in } [3-oxidation Spiral

Acetyl CoA

[ In Liver

~ADH2,NADd

/

Acetoacetate, ~ ~- ~-hydroxybutyrate ; ~ (Ketone bodies)

Oxidationin extrahepatic tissues (including brain)

|

f

Energy utilized in: \\ Citrate 1. liver for conversion \ \ / of ammonia into \\ / urea and gluconeogenesis N,x" 8CH3COSCoA + AMP + PPi + 7FADH2 + 7NADH + 7H + Each molecule of acetyl-CoA yields 12 ATP (12 x 8 = 96); FADH2 yields 2 ATP (7 x 2 = 14); NADH yields 3ATP (7 x 3 = 21); and two high-energy bonds are consumed ( - 2 ; ATP --+ AMP + PPi). Thus, net ATP production is 129. The energy yield from total combustion of palmitic acid in a bomb calorimeter (Chapter 5) is

372

CHAPTER18 Lipids I: Fatty Acids and Eicosanoids

TABLE 18-2 Reaction

Direct Consequences of the Reaction

Activation reaction First dehydrogenation Hydration Second dehydrogenation Oxidation of acetyl-CoA

Hexanoic acid --->Hexanoyl-CoA Dehydrogenation of acyl-CoA; 2 (FAD ---->FAD H 2) Hydration of a,/]-unsaturated fatty acyl-CoA Dehydrogenation of/3-hydroxy-acyl-CoA; 2 (NAD +--->NAD H + H +) Formation of 3 mol of acetyl-CoA, and their oxidation in the TCA cycle and electron transport system

Moles of ATP Gained or Lost per Mole of Hexanoic Acid

-2 +4 0 +6 +(3 x 12) = +36

Total

- 2 3 8 0 kcal/mol:

C16H3202 Jr- 2302

--+ 16CO2 + 16H20 at 20~

AH ~ = - 2 3 8 0 kcal/mol In biological oxidation, the energy conserved as ATP is about 942 kcal/mol (129 x 7.3). Thus, the percentage of standard free energy of oxidation of palmitic acid conserved as high-energy phosphate is about 40% (942/2380 x 100). Regulation of Fatty Acid Oxidation Regulation of fatty acid oxidation involves diet, cofactors, competing substrates and hormones of fatty acid mobilization. Adipose tissue triacylglycerol lipolysis is one of the major sites of regulation. The other site is CPTI. The latter is inhibited by malonyl-CoA which is involved in fatty acid biosynthesis (discussed later). Thus, fatty acid oxidation and synthesis do not occur simultaneously. Insulin inhibits fatty acid oxidation by blocking lipolysis in adipose tissue, and it stimulates lipogenesis and synthesis of malonyl-CoA. Glucagon stimulates fatty acid oxidation by inhibiting synthesis of acetyl-CoA carboxylase which leads to decreased synthesis of malonyl-CoA. This causes enhanced activity of CPTI, and promotion of fatty acid oxidation. In the fed state, the glucagon/insulin ratio is low, and fatty acid synthesis is promoted in the liver. In the fasting state, the glucagon/insulin ratio is high and mobilization of free fatty acids from adipose tissue and mitochondrial fatty acid oxidation are augmented. Peroxisomal Fatty Acid Oxidation

Peroxisomes have a single membrane and contain a fairly homogeneous, moderately electron-dense matrix. They

ATP = +44

are present in many mammalian cells and are particularly abundant in liver and kidney. A normal hepatocyte contains about a thousand peroxisomes, whose proliferation is inducible by hypolipidemic drugs such as clofibrate (Chapter 20). Peroxisomes contain HzOz-producing oxidases and also HzOz-inactivating catalase. Oxidation of very long-chain, saturated, unbranched fatty acids (C24-C26) appears to take place mainly, if not exclusively, in peroxisomes after the acyl-CoA derivatives are transported across the membrane without involvement of carnitine. Oxidation is mediated by flavoprotein dehydrogenases that yield H202 and acetyl-CoA and terminates with octanoyl-CoA. Octanoyl- and acetyl-CoA are transferred to mitochondria for further oxidation. Phytinic acid monooxygenase, which initiates the catabolism of phytinic acid (a 20-carbon branched chain fatty acid of dietary origin), is probably a peroxisomal enzyme. Peroxisomal oxidation does not yield ATE All the energy produced appears as heat. Three genetic disorders (Zellweger's syndrome, neonatal adrenoleukodystrophy, and childhood adrenoleukodystrophy) exhibit defective formation of peroxisomes (in Zellweger's syndrome no morphologically detectable peroxisomes are present) or deficiency of one or more constituent enzymes. All three disorders are characterized by a marked accumulation of very long chain, saturated, unbranched fatty acids (tetracosanoic and hexacosanoic acids) in liver and central nervous system tissues, severe neurological symptoms, and early death. Peroxisomes contain dihydroxyacetone phosphate acyltransferase and alkyldihydroxyacetone phosphate synthase, which are involved in synthesis of the plasmalogens (Chapter 19). Peroxisomes may also participate in the biosynthesis of bile acids. The conversion of trihydroxycholestanoic acid to cholic acid (Chapter 19) has been localized to peroxisomes.

373

SECTION18.1 Oxidation of Fatty Acids

Other Pathways of Fatty Acid Oxidation

Propionyl-CoA Oxidation /~-Oxidation of fatty acids with an odd number of carbon atoms yields propionyl-CoA. Since the concentration of such fatty acids in the diet is small, little propionyl-CoA is produced. Important sources of propionyl-CoA are the catabolism of isoleucine, valine, methionine, and threonine (Chapter 17). Cholesterol side chain oxidation also yields propionyl-CoA. Thus, propionyl-CoA is derived from the catabolism of lipids and proteins. In ruminants, propionate is largely derived from bacterial fermentation in the rumen. Propionyl-CoA is converted to succinyl-CoA, which is oxidized or converted to glucose by way of oxaloacetate and pyruvate (gluconeogenesis; Chapter 15). SuccinylCoA may also form 3-aminolevulinate, a precursor of porphyrin biosynthesis (Chapter 29). Formation of succinylCoA from propionyl-CoA requires three mitochondrial enzymes and two vitamins (Figure 18-5). 1. Propionyl-CoA carboxylase is a tetramer of nonidentical subunits, ot and/~. The native enzyme (M.W. ~540,000) appears to have the structure (ol/~)4. Biotin is bound through an amide linkage to an e-amino group of a lysyl residue in the o~-subunit. Carboxylation is a two-step reaction similar to that of acetyl-CoA carboxylase (see below). The first step requires ATP and Mg 2+ and fixes CO2 with the

Sources

H2~CH3

Valine, isoleucine ] methionine, threonine, cholesterol side chain, odd-chain fatty acids

O---C~SCoA PropionyI-CoA Cq----ATP___~ PropionyI-CoA ADP + P~~---" carboxylase (biotin) COOH

COOH

I HC3--~H

*MethylmalonyI-COAracemase

I I

HC--CH3

O---C--SCoA

O~---C-- SCoA L-MethymalonyI-CoA

formation of an apoenzyme-biotin-CO2 complex. In the second step, the carboxyl group from the biotinyl complex is transferred to propionyl-CoA to form D-methylmalonyl-CoA. 2. Methylmalonyl-CoA racemase converts o-methylmalonyl-CoA to the L-isomer by labilization of an or-hydrogen atom, followed by uptake of a proton from the medium. 3. Methylmalonyl-CoA mutase utilizes 5'-deoxyadenosylcobalamin (Chapter 38) to catalyze intramolecular isomerization by the migration of the -COSCoA group. The only other cobalamindependent reaction in the mammalian system is methylation of homocysteine to methionine (Chapters 17, 27, and 38). Inborn errors of metabolism may be due to propionylCoA carboxylase deficiency, defects in biotin transport or metabolism, methylmalonyl-CoA mutase deficiency, or defects in adenosylcobalamin synthesis. The former two defects result in propionic acidemia, the latter two in methylmalonic acidemia. All cause metabolic acidosis and developmental retardation. Organic acidemias often exhibit hyperammonemia, mimicking ureagenesis disorders, because they inhibit the formation of N-acetylglutamate, an obligatory cofactor for carbamoyl phosphate synthase (Chapter 17). Some of these disorders can be partly corrected by administration of pharmacological doses of the vitamin involved (Chapter 38). Dietary protein restriction is therapeutically useful (since propionate is primarily derived from amino acids). Propionic and methylmalonyl acidemia (and aciduria) results from vitamin B12 deficiency (e.g., pernicious anemia; Chapter 38). c~-Oxidation

u-Oxidation is important in the catabolism of branchedchain fatty acids. The general reaction, catalyzed by a monooxygenase, requires O2, Fe z+, and either ascorbate or tetrahydropteridine. It has been demonstrated in plants and in microsomes from brain and other tissues.

D-Methylmalonyl-CoA

Methylmalonyi-CoA mutase (5"-deoxyadenosyl-cobalamin)

R--CH2--CH2--COOH + Reduced cofactor + 02 Monooxygenase

COOH

I I CH2 I

CH2

O~--~-C - - SCoA

Metabolized in TCA cycle or used in other metabolic pathways (porphyrin synthesis, ketone body utilization, etc.)

OH

I

R--CH2mCH--COOH

+ Oxidized cofactor + H20

~-Hydroxy fatty acid

SuccinyI-CoA

FIGURE 18-5 Metabolism of propionyl-CoA.

This reaction is also a route for the synthesis of hydroxy fatty acids. The o~-hydroxy fatty acid can be further

374

CHAPTER 18

oxidized and decarboxylated to a fatty acid one carbon shorter than the original. Thus, if an odd-chain-length compound is used initially, an even-chain-length acid is produced that can be further oxidized by fl-oxidation. OH I

R--CH2--CHmCOOH -

NAD '

Oxidation of unsaturated fatty acids requires A3-cis-,A 2trans-enoyl-CoA isomerase and NADPH-dependent 2,4-

In Refsum's disease, an autosomal recessive disorder, the defect is probably in the ot-hydroxylation of phytanic acid. Phytanic acid is a 20-carbon, branched-chain fatty acid derived from the plant alcohol phytol, which is present as an ester in chlorophyll. Thus, its origin in the body is from dietary sources. The oxidation of phytanic acid is shown in Figure 18-6. The clinical characteristics of Refsum's disease include peripheral neuropathy and ataxia, retinitis pigmentosa, and abnormalities of skin and bones. Significant improvement has been observed when patients are kept on low-phytanic acid diets for prolonged periods (e.g., diets that exclude dairy and ruminant fat).

Phytol CH~

/

CH~

~ " { CH,)

dienoyl-CoA reductase, in addition to the enzymes of fioxidation. The enoyl-CoA isomerase produces the substrate for the hydration step. The reductase catalyzes the reduction of a2-trans-,a4-cis-decadienoyl-CoA to A 3trans-decenoyl-CoA. The latter is isomerized to AZ-trans decenoyl isomerase, which is a normal/3-oxidation intermediate. These reactions are illustrated for oxidation of oleic and linoleic acids in Figures 18-7 and 18-8.

18.2 M e t a b o l i s m

~-Oxidation blocked by the methyl group OOH

Phytanicacid(3,7,11,15-tetramethylhexadecanoic acid) CO2-,~ a-Oxidation (block in Refsum's disease)

CH3 ~

~ C O O H

Pristanic acid Degradation by 13-oxidation as shown byarrowmarks

H=~CHmCOO H _ ( 3 H,O/

+ 3CH3CH=COOH+3CI-~COOH

FIGURE 18-6 Oxidation of phytol and phytanicacid.

of K e t o n e B o d i e s

Ketone bodies consist of acetoacetate, D-fl-hydroxybutyrate (D-3-hydroxybutyrate), and acetone. They are

1

H3C

>

Oxidation of Mono- and Polyunsaturated Fatty Acids

o II R~CH2~C--OH

CH3

w-oxidation

Further oxidation of the co-hydroxy acids produces dicarboxylic acids, which can be fl-oxidized from either end.

C02

, / ~

co-Oxidation is oxidation of the carbon atom most remote from the carboxyl group in a fatty acid. The basic reaction, catalyzed by a monooxygenase that requires NADPH, O2, and cytochrome P-450, is shown below. It has been observed in liver microsomes and some bacteria.

HO-CH2-(CH2)n-COOH -t- NADP + -t- H20

o II R--CH2mC~COOH decarboxylation~~

w-Oxidation

H3C-(CH2)n-COOH + 02 + NADPH + H +

DehydrogenaseL ~ ""-,----~ NADH+ H '

OxidativLe

LipidsI: FattyAcidsand Eicosanoids

SECTION18.2 Metabolism of Ketone Bodies

375

9 10 OleyI-CoA

mSC~

~-Oxidations

3 AcetyI-CoA4

o

4

II

3

C--SCoA

As-cis-EnoyI-CoA

(inactive substrate in ~-oxidation)

[ l~.cis-~.trans-

EnoyI-CoAisomerase o

3

II

C~SCoA

A=.trans-EnoyI-CoA (normal substrate in 13-oxidation)

[

~-Oxidation

6 AcetyI-CoA F I G U R E 18-7 Oxidation of oleic acid.

synthesized in liver mitochondria. The overall steps involved in the formation of ketone bodies include the mobilization of fatty acids by lipolysis from adipose tissue triacylglycerol by hormone-sensitive triacylglycerol lipase, plasma fatty acid transport, fatty acid activation, fatty acid transport into mitochondria (with acylcarnitine as an intermediate), and /3-oxidation. The regulatory reactions are those of lipolysis and of acyl-CoA transport across the mitochondrial membrane (CPTI). Synthesis of ketone bodies from acetyl-CoA consists of three steps: formation of acetoacetyl-CoA, formation of acetoacetate, and reduction of acetoacetate to/3hydroxybutyrate. Nonenzymatic decarboxylation of acetoacetate yields acetone, which is eliminated via the lungs. The pathways of formation of ketone bodies are shown in Figure 18-9. The major pathway of production of acetoacetate is from /3-hydroxy-/3-methylglutaryl-CoA (HMG-CoA). Hydrolysis of acetoacetyl-CoA to acetoacetate by acetoacetyl-CoA hydrolase is of minor importance because the enzyme has a high Km for acetoacetylCoA. HMG-CoA is also produced in the cytosol, where it is essential for the synthesis of several isoprenoid compounds and cholesterol (Chapter 19). The reduction of acetoacetyl-CoA to /3-hydroxybutyrate depends on the mitochondrial [NAD +]/[NADH] ratio. Ketone bodies are oxidized primarily in extrahepatic tissues (e.g., skeletal muscle, heart, kidney, intestines, brain) within mitochondria. /3-Hydroxybutyrate is oxidized

to acetoacetate by NAD+-dependent/3-hydroxybutyrate dehydrogenase by reversal of the reaction that occurred during ketogenesis: CH3CH(OH)CH2CO- + NAD + --+ D-/3-Hydroxybutyrate

CH3COCHzCOO- + NADH + H + acetoacetate

Activation of acetoacetate requires transfer of coenzyme A from succinyl-CoA, derived from the TCA cycle, by succinyl-CoA-acetoacetate-CoA transferase (thiophorase): CH2COSCoA

I

CH2COOSuccinyiCoA

+1

CH2COO- CH2COO- CH2COSCoA ----' I

+1

COCH3

CH2COO- COCH3

Acetoacetate

Succinate AcetoacetylCoA

The activation occurs at the expense of conversion of succinyl-CoA to succinate in the TCA cycle and formation of GTP (Chapter 13). Acetoacetyl-CoA is cleaved to two molecules of acetyl-CoA by acetyl-CoA acetyltransferase, the same enzyme involved in the synthesis of acetoacetylCoA (Figure 18-9). Acetyl-CoA is oxidized in the TCA cycle. Thus, formation of ketone bodies in the liver and their oxidation in extrahepatic tissues are dictated by the ratio [substrates]/[products].

376

CHAPTER18 Lipids I" Fatty Acids and Eicosanoids O

13

12

10

9

~SCoA

i

3 Acetyl CoAql3-Oxidations

o

7 . . . .6 . . . 4. .

C---SCoA

3

Z~-cis-Ae-cis-DienoyI-CoA (inactive substrate in 13-0xidation) [ A3-cis-A 2-trans-

EnoyI-CoA isomerase O

II

3 7

C~

SCoA

6

~2. trans.A6.cis.DienoyI.CoA (normal substrate in 13-oxidation)

Acetyl CoAq 13-Oxidation

~

,

cis ~ C - - S C o A 5

4

-cis-enoyI-CoA AcyI-CoA dehydrogenase O

~C~SCoA

cis 5

II

4 ...... 2

z~ - trans-A" -cis DienoyI-CoA 2,4-Dienoyl-CoA redu~ase 4

~

C

~

S

C

o

A

A3.trans.EnoyI.CoA

II 0

EnoyI-CoA isomerase

~

C

~

S

C

o

AL trans'En~176

A

0II

13-Oxidations

5 AcetyI-CoA F I G U R E 18-8 Oxidation of linoleic acid.

Physiological and Pathological Aspects of Metabolism of Ketone Bodies Acetoacetate and fl-hydroxybutyrate are products of normal metabolism of fatty acid oxidation and serve as metabolic fuels in extrahepatic tissues. Their level in blood depends on the rates of production and utilization. Oxidation increases as their plasma level increases. Some extrahepatic tissues (e.g., muscle) oxidize them in preference to glucose and fatty acid. Normally, the serum concentration of ketone bodies is less than 0.3 mM/L. The rate of formation of ketone bodies depends on the concentration of fatty acids derived from hydrolysis of

adipose tissue triacylglycerol by hormone-sensitive lipase. Insulin depresses lipolysis and promotes triacylglycerol synthesis and storage, while glucagon has the opposite effects. Thus, insulin is antiketogenic and glucagon is ketogenic (Chapter 22). Uncontrolled insulin-dependent diabetes may result in fatal ketoacidosis (Chapter 39). Although ketonemia and ketonuria are generally assumed to be due to increased production of ketone bodies in the liver, studies with depancreatized rats have shown that ketosis may also arise from their diminished oxidation. Ketosis can occur in starvation, in ethanol abuse, and following exercise, the last because of a switch in blood

377

SECTION18.3 Metabolism of Ethanol AcetyI-CoA

2CH3COSCoA AcetyI-CoA

acetyltransferase

-

(~,, .................CoASH

9CH~COCI-IzCOSCoA

................

9

..(Acet~ %,

.... "(~ -- C~COSCoA AcetyI-CoA

HMG-CoA synthase* ~---*CoASH CH3

I

-OOCCH2~CH2COSCoA OH

J3-Hydroxy- 13-methylglutaryI-CoA (H MG-CoA) HMG-CoA

lyase NADH + H+

CH3COSCoA + CHsCOCH=COO Acetoacetate

CO2~ymati

c

NAD+

~-Hydroxy-

, C~CH(OH)CH=COO-

butyrate dehydrogenase

O-.6-Hydroxybutyrate

CH3COCHs

Acetone F I G U R E 18-9 Ketogenesis in the liver. All reactions occur in mitochondria; the rate-controlling reactions (not shown) are release of fatty acids from adipose tissue and uptake of acyl-CoA into mitochondria, in particular, the CPTI reaction (see Figure 18-1). Acetoacetyl-CoA may regulate ketogenesis by inhibiting the transferase and the synthase. *This enzyme is similar to citrate synthase (Chapter 13) which catalyzes an analogous reaction.

flow. During sustained exercise, the blood flow to the liver, intestines, and kidneys is substantially decreased, with a corresponding increase in blood flow to working muscles, so that more fatty acids mobilized from adipose tissue are delivered to the muscle. Thus, the formation of ketone bodies is severely curtailed. But during the postexercise period, with the resumption of normal blood flow to liver, ketone bodies are generated as a result of increased mobilization of fatty acids. Reduced ketone body utilization in the extrahepatic tissues can occur due to deficiency of either succinyl-CoAacetoacetate-CoA transferase or acetyl-CoA acetyltransferase. These patients are susceptible to attacks of ketoacidosis and the presence of persistent ketone bodies in the urine.

18.3 Metabolism of Ethanol Ethanol is consumed widely. Microbial fermentation in the large intestine of humans can produce about 3 g of ethanol per day. Ethanol is rapidly absorbed throughout the gastrointestinal tract or, when inhaled, through the lungs. It is metabolized in the liver by a process having zeroorder kinetics; i.e., the rate of oxidation is constant with time. The amount metabolized per unit time depends on

liver size (or body weight); the average rate in an adult is about 30 mL in 3 hours. The energy content of alcohol is about 7 kcal/g. Ethanol oxidation begins with conversion to acetaldehyde by alcohol dehydrogenase (M.W. ~85,000), a zinccontaining, NAD+-dependent enzyme that is a relatively nonspecific cytoplasmic enzyme with a Km of about 1 mM/L: CH3CH2OH + NAD + --+ CH3CHO 4- NADH + H + Acetaldehyde is rapidly converted to acetate by NAD +dependent aldehyde dehydrogenase: CH3CHO + NAD + + H 2 0 - + CH3COOH + NADH + H + Ethanol is also oxidized by the mixed-function oxidase of smooth endoplasmic reticulum, which requires NADPH, oxygen, and a cytochrome P-450 electron transport system (Chapter 14): CH3CHzOH + NADPH + H + + 202 --+ CH3CHO + 2H202 -Jr-NADP + Many drugs are metabolized by this enzyme, hence the competition between ethanol and other drugs (e.g.,

378

CHAPTER18

barbiturates). Peroxisomal catalase catalyzes the reaction: CH3CH2OH -+- H202 --+ CH3CHO --F 2H20 The Km for this catalase and for the mixed-function oxidase is about 10 mM/L. The extent to which these two enzymes metabolize ethanol is not known. Acetaldehyde is converted to acetate in the liver by NAD+-linked aldehyde dehydrogenases, one in the cytosol (Km = 1 mM/L) and another in mitochondria ( K m - - 0 . 0 1 mM/L):

CH3COO- + NADH + 2H + Disulfiram (tetraethylthiuram disulfide)

H5C~

/ 02H5

N--CwS--S--CmN

H5C2

H

S

for acetaldehyde. Thus, the cause of ethanol sensitivity may be impaired rate of removal of acetaldehyde rather than its excessive formation. Individuals who are predisposed to ethanol sensitivity should avoid ethanol intake in any form. Acetate produced from ethanol is converted to acetylCoA by acetyl-CoA synthase in hepatic and extrahepatic tissues. CH3COO- + ATP 4- + CoASH ---+ CH3COSCoA + AMP 2- + PP~-

CH3CHO + NAD + + H20--+

/

Lipids I: Fatty Acids and Eicosanoids

ii

S

\

C2H5

causes irreversible inactivation of these aldehyde dehydrogenases by reacting with sulfhydryl groups, with a buildup of acetaldehyde that produces the acetaldehyde syndrome (vasodilation, intense throbbing, pulsating headache, respiratory difficulties, copious vomiting, sweating, thirst, hypotension, vertigo, blurred vision, and confusion). Disulfiram by itself is relatively nontoxic. It is used in the treatment of chronic alcoholism but does not cure it. Disulfiram provides a willing patient with a deterrent to consumption of alcohol. A shorter acting reversible inhibitor of aldehyde dehydrogenase is calcium carbimide, which causes accumulation of acetaldehyde and unpleasant symptoms. Thus, calcium carbimide can also be used as a deterrent to alcohol consumption. Symptoms similar to the disulfiram-ethanol reaction occur in high proportion in certain ethnic groups (e.g., Asians and Native Americans) who are extremely sensitive to ethanol consumption. The ethanol sensitivity in these populations is accompanied by a higher acetaldehyde steady-state concentration in the blood, which may be due to a rapid rate of formation of acetaldehyde by alcohol dehydrogenase or to a decreased rate of its removal by aldehyde dehydrogenase. Both of these dehydrogenases are present in several isozyme forms and exhibit extensive polymorphism among racial groups. An alcohol dehydrogenase variant found in the ethanol-sensitive populations has a relatively higher rate of activity at physiological pH and may account for more rapid oxidation of ethanol to acetaldehyde. However, a more important cause of acetaldehyde accumulation appears to be deficiency of an isozyme of aldehyde dehydrogenase, which has a low Km

Acetyl-CoA is oxidized in the TCA cycle and is used in liver and adipose tissue for biosynthesis of fatty acids and triacylglycerol. Alcoholism affects about 10% of the drinking population and alcohol (ethanol) abuse has been implicated in at least 20% of admissions to general hospitals. This chronic disease exhibits high mortality due to a wide variety of factors. Ethanol produces effects in virtually every organ system. The biochemical effects of ethanol are due to increased production of NADH that decreases the [NAD+]/[NADH] ratio in the cytoplasm of liver cells at least tenfold from the normal value of about 1000. Increased production of lactate and inhibition of gluconeogenesis (Chapter 15) result. The hyperuricemia associated with ethanol consumption has been attributed to accelerated turnover of adenine nucleotides and their catabolism to uric acid (Chapter 27). Alcohol increases hepatic fatty acid and triacylglycerol synthesis and mobilization of fat from adipose tissue, which can lead to fatty liver, hepatitis, and cirrhosis. These effects are complicated by a deficiency of B vitamins and protein. Alcohol increases the plasma level of VLDL and of HDL cholesterol (Chapter 20). Many actions of ethanol may be attributed to a membrane-disordering effect. Changes in membrane fluidity can affect membrane-bound enzymes (e.g., Na +, K +ATPase, adenylate cyclase) and phospholipid architecture. Alcohol also affects several neurotransmitter systems in the brain. These include dopamine (mediates pleasurable effects), v-aminobutyric acid (GABA), glutamate, serotonin, adenosine, norepinephrine, and opioid peptides. Potential drug therapy for alcohol dependence consists of the use of antagonists and agonists of alcohol affected neurotransmitter systems. For example, naltrexone, a/z-opioid antagonist, inhibits alcohol-induced dopamine release, thus minimizing the pleasurable effect of alcohol and reducing the desire to consume alcohol. Another drug, acamprosate, reduces the craving for alcohol presumably by an agonist activity at GABA receptors

SECTION 18.4

Synthesis of Long-Chain Saturated Fatty Acids

and an inhibitory activity at N-methyl-D-aspartate receptors. A selective antagonist of serotonin receptor 5-HT3, ondansetron, reduces alcohol consumption in patients with early onset alcoholism. The 5-HT3 receptors are densely distributed in mesocorticolimbic neuronal terminals and regulate dopamine release. Attenuation of dopamine release reduces alcohol consumption. In chronic alcoholics, heavy drinking and decreased food intake lead to ketoacidosis. Accelerated lipolysis arising from reduced insulin and increased glucagon secretion caused by hypoglycemia leads to ketosis with a high [flhydroxybutyrate]/[acetoacetate] ratio. Treatment requires normalization of fluid and electrolyte balance (Chapter 39) and of glucose level. Administration of glucose provokes insulin release and depresses glucagon release, thus suppressing the stimuli for ketogenesis. The distinction between diabetic ketoacidosis and alcoholic ketoacidosis may be difficult to determine, and in some patients plasma glucose levels may not discriminate between the two entities (although in diabetic ketoacidosis plasma glucose levels are usually high, whereas in alcoholic ketoacidosis these levels may be low, normal, or marginally elevated). Fluid and electrolyte replacement and glucose administration in ketoacidosis are essential regardless of etiology. Ethanol is a teratogen partly because it inhibits embryonic cellular proliferation. Maternal alcoholism causes fetal alcohol syndrome, which is characterized by abnormal function of the central nervous system, microcephaly, cleft palate, and micrognathia.

18.4 Synthesis of Long-Chain Saturated Fatty Acids The reactions of d e n o v o fatty acid biosynthesis are shown in Figure 18-10. They are carried out by two multienzyme systems functioning in sequence. The first is acetyl-CoA carboxylase, which converts acetyl-CoA to malonyl-CoA. The second is fatty acid synthase, which sequentially joins two-carbon units of malonyl-CoA, eventually producing palmitic acid. Both complexes consist of multifunctional subunits. The various catalytic functions can be readily separated in plant cells and prokaryotes, but in yeasts, birds, and mammals, attempts to subdivide catalytic functions lead to loss of activity. Important features of this system are as follows: synthesis takes place in the cytosol (whereas fatty acid oxidation occurs in mitochondria). 2. All carbon atoms are derived from acetyl-CoA (obtained from carbohydrates or amino acids), and 1. D e n o v o

:379

palmitate (C16) is the predominant fatty acid

3.

4.

5.

6.

produced. Fatty acids longer than 16 carbons, those that are unsaturated, and hydroxy fatty acids are obtained by separate processes of chain elongation, desaturation, or ot-hydroxylation, respectively. The committed (rate-controlling) step is the biotin-dependent carboxylation of acetyl-CoA by acetyl-CoA carboxylase. Important allosteric effectors are citrate (positive) and long-chain acyl-CoA derivatives (negative). Although the initial step requires CO2 fixation, CO2 is not incorporated into fatty acids. The labeled carbon in 14C02 (as H14CO3) is not incorporated into the carbons of fatty acids synthesized. Synthesis is initiated by a molecule of acetyl-CoA that functions as a primer. Its two carbons become C15 and C16 of palmitate. The acetyl group is extended by successive addition of the two carbons of malonate originally derived from acetyl-CoA, the unesterified carboxylic acid group being removed as CO2. In mammalian liver and mammary gland, butyryl-CoA is a more active primer than acetyl-CoA. Odd-chain-length fatty acids found in some organisms are synthesized by priming the reaction with propionyl-CoA instead of acetyl-CoA. Release of the finished fatty acid occurs when the chain length reaches C16 by action of thioester hydrolase, which is specific for long-chain acyl-CoA derivatives. A thioester hydrolase of mammary gland is specific for acyl residues of C8, C10, or C12.

The overall reaction for palmitate synthesis from acetylCoA is 8Acetyl-CoA + 14NADPH 4- 14H + 4- 7ATP 4- H20 -+ palmitate 4- 8CoASH 4- 14NADP + 4- 7ADP 4- 7Pi The reducing equivalents of NADPH are derived largely from the pentose phosphate pathway. Acetyl-CoA carboxylase is a biotin-dependent enzyme. It has been purified from microorganisms, yeast, plants, and animals. In animal cells, it exists as an inactive protomer (M.W. ~400,000) and as an active polymer (M.W. 4-8 million). The protomer contains the activity of biotin carboxylase, biotin carboxyl carrier protein (BCCP), transcarboxylase, and a regulatory allosteric site. Each protomer contains a biotinyl group bound in amide linkage to the s-amino group of a lysyl residue. Citrate shifts the equilibrium from inactive protomer to active polymer. The polymeric form appears as long filaments in electron micrographs.

380

CHAPTER18 Lipids I" Fatty Acids and Eicosanoids O

AcetyI-CoA O carboxylase system * Ii ..e"-" ~ - -1:'-- " HOOC-- CH2--C--SCoA ~ S H - ~ ~,~ / Mg" "~ '-'2 ATP ADP MalonylCoA CoASH~ Acetyl + H20 +P~ L , - S H ~ ,transacylase O transacylasel . . . . . reactions S--C--CH3 CysteinyI-SH group [ O "O

II

4"-ehosphopantetheine ~"'.~

CH3Cm SCoA

NO_ Icl.__C

H ;II - - - C ~ S ~ MalonyI-ACP l

II O

13-Ketoacyl synthase

~

~-Ketoacylsynthase~_..C02 0 0 II II ..... H3C--C~CH2---'C~S" v v v v AcetoacyI-ACP (in successive cycles 13-ketoacyI-ACP)

(~ Condensing reaction (~) First reduction

13.KetoacylF NADPH + H+ reductase~,-.NADP+ I-~C~ CHm C H 2 C - - S ~ D(-) 13-HydroxybutryI-ACP (in successive cycles 13-hydroxyacyI-ACP)

(~) Dehydration

13-Hydroxyacyl dehydratase "-H20 H3C--C-- C - - C ~ S ~

I

H

trans-2-ButenyI-ACP (in successivecycles ct.13-unsaturatedACP) o~.13-UnsaturatedJ~ NADPH + H+ acyI-ACP reductase (enoyl reductase) NADP+ O

(~) Second reduction

II

I--~Cm CH;--- CHz----C--S~ ButyryI-ACP (in successive cycles acyI-ACP) Is tMe acyl group16carbons long yet?

If ,es

If no

~

SH

~-~O

13-Ketoacyl synthase Acyl-group transfer

|

O

Thioester hydrolase

II

(~SmC--CH2C~CH3 Substrate for reaction 2 Palmitic acid (Accepts another malonylgroup, reaction 1, and the malonylACP initiatesthe next cycle of two-carbon extension.) F I G U R E 18-10 Synthesis of fatty acid. ACP = Functional unit of acyl-carrier-protein segment of fatty acid synthase. The cysteinyl-SH group of/3-ketoacyl synthase accepts the acetyl group or the acyl group, and its catalytic site, which is adjacent to the -SH group, catalyzes the condensation reaction.

SECTION 18.4

Synthesis of Long-Chain Saturated Fatty Acids

381 O

a(-)

/L.

~--(i~)wi~)--Ou(~)--O--~+OH

:NH 9

~".....

HO

ATP

\

................ ""O- ..""

/

NH

O

II

(CH2),--C-- NH

I

HO~C---OaBicarbonate

BCCP

Mg carboxylase a(-)

R

0

-OuCmN

'~')~

I

2'~Biotin

ADP + H2PO,-+H+

NH

Carboxylated-BCCP

.................................................................. 9 '" ~.CmCwSCoA

*>...

Lysyl

5+

R

H"lH OIJ 6-

Transcarboxylase

AcetyI-CoA

O

[ I

o H O

_o lel [

//~

II

C--C--SCoA

I

H

NH

NH

+

R

MalonyI-CoA

Regenerated BCCP

FIGURE 18-11 Mechanismof carboxylationof acetyl-CoA.BCCP= Biotincarboxylcarrierprotein; Ad = adenosine.

The mechanism of the carboxylation reaction consists of two half-reactions: biotincarboxylase ATP + HCO~- + BCCP < > Mg2+ ADP + Pi -Jr-B C C P - C O O and BCCP-COO- + acetyl-CoA Z BCCP + malonyl-CoA The overall reaction is ATP 4- HCO~- 4- acetyl-CoA ~ malonyl-CoA 4- ADP 4- Pi The presumed reaction mechanisms are shown in Figure 18-11. Other biotin-dependent enzymes include propionylCoA carboxylase and pyruvate carboxylase (Chapter 15). The latter, like acetyl-CoA carboxylase, is subject to allosteric regulation. Pyruvate carboxylase, a mitochondrial enzyme, is activated by acetyl-CoA and converts pyruvate to oxaloacetate which, in turn, is converted to glucose via

the gluconeogenic pathway or combines with acetyl-CoA to form citrate. Some of the citrate is transported to the cytosol, where it activates the first step of fatty acid synthesis and provides acetyl-CoA as substrate (see below). Other carboxylation reactions use bicarbonate but are dependent on vitamin K, the acceptor being glutamyl residues of glycoprotein clotting factors II, VI, IX, and X and anticlotting factors Protein C and Protein S (Chapter 36). Acetyl-CoA carboxylase is under short- and long-term control. Allosteric modulation functions as a short-term regulator. Positive modulators are citrate and isocitrate; negative modulators are long-chain acyl-CoA derivatives. The binding of citrate increases the activity by polymerization of the protomers, whereas negative modulators favor dissociation of active polymers to inactive monomers. Acetyl-CoA carboxylase is also regulated by covalent modification by phosphorylation, which inhibits activity, and by dephosphorylation, which restores activity. Phosphorylation can occur by action of cAMP-dependent protein kinase through/~-adrenergic agonists and glucagon or by action of calcium-dependent protein kinase through c~-adrenergic agonists. It is not known whether the

382

CHAPTER18 Lipids I" Fatty Acids and Eicosanoids

kinases act at different sites. Insulin suppresses cAMP levels and promotes activity of acetyl-CoA carboxylase. Insulin may also increase the activity of acetyl-CoA carboxylase phosphatase, which is complexed with the carboxylase. This phosphatase also dephosphorylates glycogen synthase, phosphorylase a, and HMG-CoA reductase. Thus, common mediators (e.g., insulin, glucagon, and catecholamines) regulate fatty acid synthesis and carbohydrate metabolism. Long-term regulation of acetyl-CoA carboxylase involves nutritional, hormonal (e.g., insulin, thyroxine), and other factors. In animals on high-carbohydrate diets, fatfree diets, choline deprivation, or vitamin B 12 deprivation, the activity is enhanced. However, fasting, high intake of fat or of polyunsaturated fatty acids, and prolonged biotin deficiency leads to decreased activity. In diabetes, the enzyme activity is low, but insulin administration raises it to normal levels. Fatty acid synthesis is also carried out by a multienzyme complex and leads from acetyl-CoA, malonyl-CoA, and NADPH to palmitic acid. The overall process is as follows: o

o

II

/3-Ketoacyl-ACP synthase (condensing enzyme) has an active cysteine-SH group that forms an acetyl-S (or acylS) intermediate (E -- enzyme). O

II CH3C~S~ACP + E~SH O

II CH3C~S~E + ACP~SH

The condensation occurs with release of CO2" O

O

o

II

II

II

CH3C~S~E + HO~C---CH2~C~S~ACP

O

O

II

II

.~

C H 3 C C H 2 C ~ S ~ A C P + CO2 + E ~ S H

/3-Ketoacyl-ACP reductase catalyzes the first reduction reaction: O

O

II

II

C H 3 C ~ C H 2 C ~ S ~ A C P + NADPH * + H +

II

.GHa.G--SCoA + 7HOOC--CH2G--SGoA + 14NADPH + 14H +

Acetyl-CoA

OH

MalonyI-CoA

I

C;H3(~H2(CH2CH2)6CH2COOH+ 7CO2 + 14NADP+ + 8CoASH + 6H20

0 II

C H 3 ~ C H C H 2 C ~ S - - - A C P + NADP +

Palmitic acid

The fatty acid synthesis complex contains seven catalytic sites. Acetyl transacylase catalyzes the reaction O

II CH3C~SCoA + A C P ~ S H

;

It is stereospecific, and the product formed is D(--)-flhydroxybutyryl-ACP (or D(-)-fl-hydroxyacyl-ACP, in subsequent reactions). fl-Hydroxyacyl-ACP dehydratase catalyzes the reaction: OH

O

I

II

CH3CHCH2C~S~ACP : O

II

O

C H 3 C ~ S ~ A C P + CoASH

II

trans-CH3CH'--CHC~S~ACP + H20

and malonyl transacylase catalyzes the reaction O

O

II

II

HO~C~CH2~C~SCoA

+ ACP - SH --,

O

O

II

II

HO~C--CH2--C~S--ACP

In this stereospecific reaction, the D(--) isomer is converted to a trans-oe,fl-unsaturated acyl-ACP derivative. Enoyl-ACP reductase catalyzes the second reduction with NADPH. O

Ii CH3CH=CH C ~ S ~ A C P

(ACP-SH is the 4'-phosphopantetheine-SH of the acyl carrier domain.) These two priming reactions load a cysteinyl-SH of one subunit with acetyl-CoA, and t h e - S H of the 4'phosphopantetheine arm of the acyl carrier site on the other subunit with malonyl-CoA.

+ NADPH + H + .-

O

II

C H 3 C H 2 C H 2 C ~ S ~ A C P + NADP +

The product formed is a saturated acyl-thioester of ACE

SECTION18.4 Synthesis of Long-Chain Saturated Fatty Acids

383

F I G U R E 18-12 Diagram of a fatty acid synthase dimer with its head-to-tail association of the two multifunctional polypeptides. KS =/5-Ketoacyl synthase; MT = malonyl transacylase; AT = acetyl transacylase; DH = dehydratase; ER = enoyl reductase; KR =/3-ketoacyl reductase; ACP = acyl carrier site; TE -- thioesterase. [Reproduced with permission from S. J. Wakil, J. K. Stoops, and V. C. Joshi, Fatty acid synthesis and its regulation. Annu. Rev. Biochem. 52, 537 (1983). 9 1983 by Annual Reviews Inc.]

Thioester hydrolase catalyzes the removal of palmitate from the 4'-phosphopantetheine arm of the acyl carrier site. O

II CH3(CH2)14C--S--ACP + H20

CH3(CH2)14COOH + ACP--SH

The enzyme has an active serine residue and is specific for long-chain acyl derivatives.

Functional Organization of Fatty Acid Synthase In animal cells, fatty acid synthase (FAS) consists of two subunits, each having a molecular weight of 250,000. Evidence from negative-stain electron microscopy indicates that FAS is a linear polypeptide with a series of globular domains representing areas of catalytic activity. Fatty acid synthase mRNA is large enough to code for the ~2300 amino acids required. Animal FAS is the largest

known multifunctional protein. Although each subunit contains all of the catalytic activities required to synthesize palmitate, the monomer lacks fl-ketoacyl synthase activity. The two subunits must be juxtaposed head to tail to bring the cysteine-SH of the/3-ketoacyl synthase of one close to the 4'-phosphopantetheine-SH of the acyl carrier site of the other to obtain a fully functional dimer. Figure 18-12 presents schematically the active FAS homodimer complex. Synthesis may proceed from either end of the active complex. The assembly of rat liver FAS involves three stages: synthesis of the multifunctional polypeptide chains, formation of the dimer, and attachment of a 4'-phosphopantetheine group by an enzyme-catalyzed reaction. This assembly process is influenced by changes in developmental, hormonal, and nutritional states. The FAS complex provides considerable catalytic efficiency, since free intermediates do not accumulate and the individual activities are present in equal amounts. The central role of the acyl carrier domain is to carry acyl groups from one catalytic site to the next. The

384

CHAPTER 18

4'-phosphopantetheine (20 nm long) derived from coenzyme A is bound as a phosphodiester through the hydroxyl group of a specific seryl residue. The acyl intermediates are in thioester linkage with t h e - S H of the prosthetic group, which serves as a swinging arm to carry acyl groups from one catalytic site to the next. The structure of 4'-phosphopantetheine attached to the serine residue of ACP is 0

Seryl ) residue

OH 3

0

O

II ] II II --O--P--OnCH2mCmCH--C--NmCH2mCHzmC--N--CH2--CH2uSH I I I H H OH I OH CH3

Sources of N A D P H for Fatty Acid Synthesis The reducing agent for fatty acid synthesis is NADPH. It is mostly supplied by the pentose phosphate pathway (Chapter 15) in the reactions Glucose-6-phosphate

dehydrogenase

Glucose 6-phosphate

f--,~ NADP §

6-Phosphate-gluconolactone

NADPH + H *

and 6-Phosphogluconate

dehydrogenase

/%--.,,

6-Phosphogluconate

NADP *

CO2

Ribulose 5-phosphate

NADPH+ H

Lipids I Fatty Acids and Eicosanoids

mitochondrial membrane to the cytosol, its carbon atoms are transferred by two transport mechanisms. 1. Transport dependent upon carnitine: Carnitine participates in the transport of long-chain acyl-CoA into the mitochondria and plays a similar role in the transport of acetyl-CoA out of mitochondria. However, carnitine acetyl transferases have a minor role in acetyl-CoA transport. 2. Cytosolic generation of acetyl-CoA ("citrate shuttle"): This pathway is shown in Figure 18-13. Citrate synthesized from oxaloacetate and acetyl-CoA is transported to the cytosol via the tricarboxylate anion carrier system and cleaved to yield acetyl-CoA and oxaloacetate. Citrate 3- + ATP 4- + CoA

ATP citrate-lyase

>

(or citrate cleavage enzyme)

acetyl-CoA + oxaloacetate 2- -I- ADP 3- + p2Thus, citrate not only modulates the rate of fatty acid synthesis but also provides carbon atoms for the synthesis. The oxaloacetate formed from pyruvate may eventually be converted (via malate) to glucose by the gluconeogenic pathway. The glucose oxidized via the pentose phosphate pathway augments fatty acid synthesis by providing NADPH. Pyruvate generated from oxaloacetate can enter mitochondria and be converted to oxaloacetate, which is required for the formation of citrate.

Oxidation of malate also provides NADPH: M a late

Malic enzyme

, NADP *

CO2

, Pyruvate

NADPH + H *

These three enzymes, like the fatty acid synthase complex, are located in the cytosol. Active lipogenesis occurs in liver, adipose tissue, and lactating mammary glands, which contain a correspondingly high activity of the pentose phosphate pathway. Thus, lipogenesis is closely linked to carbohydrate oxidation. The rate of lipogenesis is high in humans on carbohydrate-rich diets. Restricted energy intake, a high-fat diet, and insulin deficiency decrease fatty acid synthesis. Source and Transport of Acetyl-CoA Acetyl-CoA is synthesized in mitochondria by a number of reactions: oxidative decarboxylation of pyruvate, catabolism of some amino acids (e.g., phenylalanine, tyrosine, leucine, lysine, and tryptophan; see Chapter 17), and fl-oxidation of fatty acids (see above). Since acetylCoA cannot be transported directly across the inner

Regulation of Fatty Acid Synthase Like acetyl-CoA carboxylase, FAS is under short- and long-term control. The former is due to negative or positive allosteric modulation or to changes in the concentrations of substrate, cofactor, and product. The latter usually consists of changes in enzyme content as a result of protein synthesis or decreased protein degradation. Variation in levels of hormones (e.g., insulin, glucagon, epinephrine, thyroid hormone, and prolactin) and in the nutritional state affect fatty acid synthesis through short- and long-term mechanisms. In the diabetic state, hepatic fatty acid synthesis is severely impaired but is corrected by administration of insulin. The impairment may be due to defects in glucose metabolism that lead to a reduced level of an inducer or increased level of a repressor of transcription of the FAS gene, or both. Glucagon and epinephrine raise intracellular levels of cAME and their inhibitory effect on fatty acid synthesis may be due to phosphorylation or dephosphorylation of acetyl-CoA carboxylase. They also stimulate the action of hormone-sensitive triacylglycerol lipase and raise intracellular levels of long-chain acyl-CoA.

SECTION18.4 Synthesis of Long-Chain Saturated Fatty Acids

385

FIGURE 18-13 Cytoplasmic generationof acetyl-CoAvia citrate transport and related reactions. PPP -- Pentose phosphate pathway; FAS = fatty acid synthase.~ Negativeallosteric modifier.~ Positiveallosteric modifier.

As a result, acetyl-CoA carboxylase and citrate synthase are inhibited. The stimulatory effect of prolactin is confined to the mammary gland and may involve synthesis of the enzyme.

Mitochondrial fatty acid elongation occurs primarily when the [NADH]/[NAD +] ratio is high (e.g., anaerobiosis, excessive ethanol oxidation).

Linoleic acid, C18:2(9,12) (from the diet)

Fatty Acid Elongation Cytoplasmic fatty acid synthase yields palmitate. Human triacylglycerol contains fatty acids with 18, 20, 22, and 24 carbon atoms, which are synthesized by elongation of palmitate in endoplasmic reticulum or mitochondria. Elongation in the endoplasmic reticulum occurs mainly in liver and involves C]0_16-saturated and Cl8-unsaturated fatty acids by successive addition of two-carbon groups derived from malonyl-CoA (Figures 18-14 and 18-15). The reductant is NADPH. The intermediates, however, are CoA thioesters. The mitochondrial system uses acetyl-CoA, not malonyl-CoA, by a slightly modified reversal of /3oxidation. The substrates are saturated and unsaturated C12, C14, and C16 fatty acids, and the products are C18, C20, C22, and C24 fatty acids. The first reduction step utilizes NADH, and the enzyme is/3-hydroxyacyl dehydrogenase (a/3-oxidation enzyme); the second reduction step utilizes NADPH, and the enzyme is enoyl reductase.

(octadecadienoic acid) Activation

LinoleoyI-CoA (octadecadienoyI-CoA) H++ NAD(P)H + O2--..I ,+ ] C s desaturase NAD(P) + 2H20.~"~ ~'-LinolenoyI-CoA, C18:3(6,9,12) (octadecatrienoyI-CoA) q~ (MalonyI-CoA, Chain-elongationsystem 2NADPH) Dihomo- ~,-linolenoyI-CoA, C20:3(8,11,14) (eicosatrienoyI-CoA) H++ NAD(P)H + 02--.~ NAD(P~'+ 2H20~ !Cs desaturase ArachidonoyI-CoA, C20:4(5,8,11,14) (eicosatetraenoyI-CoA) FIGURE 18-14 Synthesis of arachidonic acid from linoleic acid. The desaturation and chain elongation occur in microsomes.

386

CHAPTER18 Lipids I: FattyAcids and Eicosanoids

(z-Linolenic acid, C18:3(9,12,15)(from the diet) (octadecatrienoic acid) Activation

o~-LinolenoyI-CoA,C18:3(9,12,15)

I

Desaturation

OctadecatetraenoyI-CoA, C18:4(6,9,12,15)

C2qchain-elongation

system

EicosatetraenoyI-CoA, C20:4(8,11,14,17) Desaturation EicosapentaenoyI-CoA, C20:5(5,8,11,14,17)

c,~

Chain-elongation system

DocosapentaenoyI-CoA, C22:5(7,10,13,16,19)

carbon chain length, number of double bonds, and doublebond positions (in parentheses). Thus, palmitoleic acid is designated C16:1(9) and linoleic acid is C18:2(9,12). The location of the double bond is sometimes indicated by A; for example, a 9 signifies that the double bond is between carbon 9 and carbon 10. In both methods, the carboxyl carbon is carbon 1. The double-bond position can also be related to the co-end of the fatty acid molecule (i.e., the methyl carbon farthest from the carboxyl end); oleic acid is an co-9 acid; linoleic acid has double bonds at o)-6 and co-9 carbons. The structures and names of some naturally occurring unsaturated fatty acids are given in Table 18-3. The presence of a double bond in the hydrocarbon chain gives rise to geometrical isomerism, which is due to restricted rotation around carbon-carbon double bonds and is exemplified by fumaric and maleic acids.

HOOC

Desaturation

DocosahexaenoyI-CoA, C22:6(4,7,10,13,16,19)

H

\/

/\

Synthesis of docosahexaenoic acid from c~-linolenic acid. Desaturation and chain elongation are similar to the use described in Figure 18-13.

18.5 Metabolism of Unsaturated Fatty Acids S t r u c t u r e and N o m e n c l a t u r e of U n s a t u r a t e d Fatty Acids Unsaturated fatty acids contain one or more double bonds. A common method for designating fatty acids gives the

Saturated chain

H

I

C~C ~ I H

trans-Monounsaturatedchain (uncommonin naturally occurringfattyacids)

H H

I I

C-~-C~

cis-Monounsaturated chain

(commonin naturally

occurring fatty acids)

H

COOH

\/

c II c

HOOC

FIGURE 18-15

H

/\

c II c

HOOC

H

Maleic acid

Fumaric acid

( cis-form )

( trans-form )

Almost all naturally occurring, unsaturated, long-chain fatty acids exist as the cis isomers, which are less stable than the trans isomers. The cis configuration introduces a bend (of about 30 ~ in the molecule, whereas the trans isomer resembles the extended form of the saturated chain (Figure 18-16). Arachidonic acid with four cis double bonds is a U-shaped molecule. Some cis isomers are biologically active as essential fatty acids. The trans isomers cannot substitute for them but are metabolized like the saturated fatty acids. F u n c t i o n s of U n s a t u r a t e d Fatty Acids

The cis unsaturated fatty acids provide fluidity of triacylglycerol reserves and phospholipid membranes and many serve as precursors of eicosanoids (prostaglandins, prostacyclins, thromboxanes, and leukotrienes). The importance of membrane fluidity and its relationship to the membrane constituent phospholipids are discussed in Chapter 10. Eicosanoids have numerous functions (see below).

18.6 Nonessential Fatty Acids F I G U R E 18-16 Geometry of saturated, trans monounsaturated, and cis monounsaturated chains.

Palmitoleic and oleic acids, the two most abundant monounsaturated fatty acids of animal lipids, can be

m

m

~Z

o

!

4..om

m

m

Z

i

i

000

o o o

C'.I

i

C'.I

i

9

II

t"t

~0~0~0~0 C'.I

i

c-,I

II

t",l

~

0 Or~ 0 ~-

~

~

i

"~

~.1

~,1

L.) r,.)

r,.)L.)L.)

N N N N ~r~.)N N N

r,.)r,.)r,.)~ r,.) ~

NNNNNNNN

L.) L.) ~

0 0 0 0 0 0 0 0

~&~~~~-

0

0 9

C~

4-- 4+

388

CHAPTER18 Lipids I" Fatty Acids and Eicosanoids

synthesized from the respective saturated fatty acid coenzyme-A esters. Desaturase is a monooxygenase system present in endoplasmic reticulum of liver and adipose tissue. The overall reaction for palmitoleic acid synthesis is

a saturated-fat diet. A diet rich in oleic acid has a lower ratio of LDL cholesterol to HDL cholesterol compared to either of the other diets. The metabolism of lipoproteins and their role in atherosclerosis are discussed in Chapter 20.

Palmitoyl-CoA + NAD(P)H + H + + O2 --+ palmitoleyl-CoA + NAD(P) + + 2H20 One molecule of oxygen accepts two pairs of electrons, one from palmitoyl-CoA and the other from NADPH or NADH. The electrons NAD(P)H are transported via cytochrome-b5 reductase to cytochrome b5 (microsomal electron transport; Chapter 14). An enzyme-bound superoxide radical is responsible for the oxidation of acylCoA. Four desaturases specific for introducing cis double bonds at C9, C6, C5, and C4, respectively, are known. If the substrate is saturated, the first double bond introduced is C9. With an unsaturated substrate, other double bonds are introduced between the carboxyl group and the double bond nearest the carboxyl group. Desaturation yields a divinylmethane arrangement of double bonds (--CH=CH--CHz--CH=CH--). Usually desaturation alternates with chain elongation. Desaturation is inhibited by fasting and diabetes. The oxidation of unsaturated fatty acids occurs in mitochondria.

18.7

trans-Fatty Acids

trans-Fatty acid metabolism is similar to that of saturated fatty acids. During the partial dehydrogenation of vegetable oils (e.g., in the manufacture of margarine), the cis fatty acids are isomerized to trans-fatty acid forms. The "hydrogenated" margarines contain 15-40% of trans-fatty acids. The hypercholesterolemic effect of trans-fatty acids may be due to impairment of the first step in the formation of bile acids from cholesterol. Since the steadystate level of cholesterol depends on its conversion to bile acid (Chapter 19), any perturbation in this process affects cholesterol levels. Both metabolic and epidemiological studies have shown that the consumption of transfatty acids increases the risk of coronary heart disease. This risk appears to be even higher when compared on a per-gram basis with saturated fatty acids. The adverse effects of trans-fatty acids are attributed to the elevation of atherogenic low-density lipoprotein (LDL) cholesterol and a decrease in the antiatherogenic (or cardioprotective) high-density lipoprotein (HDL) cholesterol level. Thus, the ratio of LDL cholesterol to HDL cholesterol is significantly higher with the trans-fatty acid diet compared to

18.8 Essential Fatty Acids Polyunsaturated fatty acids not synthesized in the body but required for normal metabolism are essential fatty acids (EFAs). EFAs are linoleic acid, linolenic acids (or and X), and arachidonic acid. All contain at least one double bond located beyond C-9 or within the terminal seven carbon atoms (Table 18-3). ~-Carbon '~1 2 3 4 5 6 7 8 H3C--CH2--CH2--CH2--CH2mCH2--CH2--CH2m(CH2)n--COOH Introduction of double bonds does not occur in humans

Double bonds can be introduced in this region

A double bond within the terminal seven carbon atoms can be present at ~o-3 or ~-6. y-Linolenic acid is an oJ-6 EFA and ot-linolenic acid an o>-3 EFA. Other co-3 EFA are eicosapentaenoic acid (EPA) and docosahexaenoic acid (DCHA), both abundant in edible fish tissues. Vegetable oils are rich in oJ-6 EFA (Table 18-4). Plants contain otlinolenic acid, which can be converted in the body to EPA and DCHA, but it is found within chloroplast membranes and not in seed oils; hence, it may not be available in significant quantities in the diet. The o~-3 and o>-6 EFA have different metabolic effects (see below). Particularly rich sources of EPA are fishes (e.g., salmon, mackerel, blue fish, herring, menhaden) that live in deep, cold waters. These fishes have fat in their muscles and their skin. In contrast, codfish, which have a similar habitat, store fat in liver rather than muscle. Thus, cod liver oil is a good source of EPA, but it also contains high amounts of vitamins A and D, which can be toxic in large quantities (Chapters 38 and 37, respectively). Shellfish also contain EPA. Plankton are the ultimate source of EPA. Linoleic acid can be converted in mammalian liver to y-linolenic acid and arachidonic acid by the microsomal desaturation and chain elongation process (Figure 18-14). Thus, the requirement for arachidonic acid may be dispensed with when the diet contains adequate amounts of linoleic acid. Similarly, ot-linolenic acid is converted by desaturation and chain elongation to EPA and DCHA (Figure 18-15).

SECTION 18.9

Metabolism of Eicosanoids

Deficiency of Essential Fatty Acids The clinical manifestations of EFA deficiency in humans closely resemble those seen in animals. They include dry, scaly skin, usually erythematous eruptions (generalized or localized and affecting the trunk, legs, and intertriginous areas), diffuse hair loss (seen frequently in infants), poor wound healing, failure of growth, and increased metabolic rate. Abnormalities in ECG patterns may be due to membrane alterations, which may also account for structural and functional abnormalities observed in mitochondria. Surgical patients maintained on glucose-amino acid solutions for prolonged periods develop EFA deficiency, manifested as anemia, thrombocytopenia, hair loss and sparse hair growth, increased capillary permeability, dry scaly skin, desquamating dermatitis, and a shift in the oxygendissociation curve of hemoglobin to the left. Oral or intravenous administration of linoleic acid is necessary to correct these problems. Fat emulsions containing linoleic acid are commercially available for intravenous use. An adult requires 10 g of linoleic acid per day. The recommended dietary allowance for EFA is 1-2% of the total energy intake. EFA deficiency can also occur in infants with highly restricted diets (e.g., primarily skim milk intake), in patients receiving total parenteral hyperalimentation without supplements of unsaturated lipids, and in those with severe malabsorptive defects. In EFA deficiency, oleic acid can be dehydrogenated to yield polyunsaturated fatty acids (PUFAs) that are nonessential and do not substitute for the essential fatty acids. One such PUFA is 5,8,11-eicosatrienoic acid, which occurs in significant amounts in heart, liver, adipose tissue, and erythrocytes of animals fed diets deficient in EFAs but decreases after supplementation with linoleic or linolenic acids. Its appearance in tissues and plasma has been used in the assessment of EFA deficiency. Most vegetable oils are relatively rich in EFAs (coconut oil is an exception), low in saturated fatty acids, and lack cholesterol. Animal fats (except those in fish), on the other hand, are generally low in EFAs, high in saturated fats, and contain cholesterol. The EFA content of body and milk fat of ruminants can be increased by the feeding of EFA encased in formalin-treated casein. The EFA is released at the site of absorption by dissolution of the capsules. These dietary manipulations in ruminants are accompanied by an increase in carcass EFA, a decrease in saturated fatty acids, and no change or an increase in cholesterol content. Table 18-4 summarizes the fatty acid composition of some fats of animal and plant origin. The recommended daily diet does not exceed 30-35% of the total energy intake as fat (current average consumption in North America is

389

40-45%), with equal amounts of saturated, monounsaturated, and polyunsaturated fats, and a cholesterol intake of no more than 300 mg/day (current average consumption in North America is about 600 mg/day). Substitution of co-6 polyunsaturated for saturated fats in the diet lowers plasma cholesterol levels through reduction in levels of VLDL and LDL. Diets rich in polyunsaturated fats lead to higher biliary excretion of sterols, although this effect may not be directly related to reduced levels of plasma lipoproteins. Diets low in EFA (linoleic acid) have been associated with high rates of coronary heart disease. A significantly lower proportion of EFA in the adipose tissue of people dying from coronary heart disease has been reported, and an inverse relationship has been found between the percentage composition of EFA in serum cholesteryl esters and mortality rates from coronary heat disease. Consumption of o9-3 polyunsaturated fatty acids markedly decreases plasma triacylglycerol and, to a lesser extent, cholesterol levels in some hyperlipoproteinemic patients (Chapter 20). Consumption of fish-oil fatty acids decreases the biosynthesis of fatty acids and of VLDL by the liver and also decrease the platelet and monocyte function. These effects of o9-3 fatty acids appear to prevent or delay atherogenesis. Low death rates from coronary heart disease are found among populations with high intake of fish (e.g., Greenland Eskimos, people of fishing villages of Japan, people of Okinawa). Metabolic and functional differences exist between o9-3 and o9-6 fatty acids. They have opposing physiological effects and their balance in the diet is important for homeostasis and normal development.

18.9 Metabolism of Eicosanoids The eicosanoids--prostaglandins (PGs), thromboxanes (TXs), prostacyclins (PGIs), and leukotrienes (LTs)Iare derived from essential fatty acids and act similarly to hormones (Chapter 30). However, they are synthesized in almost all tissues (unlike hormones, which are synthesized in selected tissues) and are not stored to any significant extent; their physiological effects on tissues occur near sites of synthesis rather than at a distance. They function as paracrine messengers and are sometimes referred to as autacoids. The four groups of eicosanoids are derived, respectively, from a 20-carbon fatty acid with three, four, or five double bonds: 8,11,14-eicosatrienoic acid (dihomov-linolenic acid), 5,8,11,14-eicosatetraenoic acid (arachidonic acid), and 5,8,11,14,17-eicosapentaenoic acid (Figure 18-17). In humans, the most abundant precursor is arachidonic acid. Secretion of eicosanoids in response

390

CHAPTER18 Lipids I: Fatty Acids and Eicosanoids

T A B L E 18-4

Fatty Acid Composition of Some Fats of Animal and Plant Origin

Saturated*

C14, C16, C18 (predominantly C16 and C18)

Polyunsaturated*, mostly as linoleic acid (C18:2)

Monounsaturated* C16:1; C18:1 (predominantly C18:1)

Animal Fats Butter Beef Chicken Pork Fish (salmon and tuna)

59 54 40 40 28

37 44 38 46 29

4 2 22 14 23 + 20 t

11 14 14 15 20 29 92 53 16 12

11 26 43 27 45 19 6 37 69 18

78 60 43 58 35 52 2 10 15 70

Vegetable Oils Safflower Corn Sesame Soybean Peanut Cottonseed Coconut Palm Olive Sunflower

*All values expressed as weight percentages of total fatty acids. tOther polyenoic acids, e.g., eicosapentaenoic acid, docosahexaenoic acid.

~

COOH

< . . . t PGE,, PGF,, TXA, LTA~, LTC3, LTD3

8,11,14-Eicosatrienoic acid (dihomo- ~,-linolenic acid)

COOH

PDG2, PGE~, PGF2, PGI2, TXA2 10 are type I; those that are dark are called type II. This staining pattern is reversed at pH Pyruvate

Glycerol

'

I

Glycerol 3-phosphate

>..Oxaloacetate x.

~

Valine / . ~ 9 . . ] / / S u c c , nate iso~euclne ~=::=~/--1-1-,^ \ Th,~u.,.~ ~ - . . . . ] Methionine J ~ CYCLE _ \ / Glutamate ~ k , ~ - K e t o g l u t a r a t e / / Proline I " ~ J Histidine J

~

~ . Glucose /

Rate-limitingsteps Oxaloacetate -" Phosphoenolpyruvate FDP--* Fructose 6-phosphate Glucose 6-phosphate --*Glucose

FIGURE 22-15 Entry of precursors into the pathway of gluconeogenesis.

to the status of gluconeogenesis include glucagon (an acute modulator), glucocorticoid (a chronic modulator), and absence of insulin. The general effects of these hormones are shown in Figure 22-16. Glucagon is directed toward glycogen degradation, gluconeogenesis, triacylglycerol hydrolysis, and fatty acid oxidation. In the liver, it stimulates glycogen degradation (Figure 22-13) and gluconeogenesis, possibly by inactivation of pyruvate kinase. Glucagon also modulates the levels of fructose-2,6-bisphosphate (Chapter 15). Insulin opposes these effects by stimulating glycogen synthesis, glycolysis, and fatty acid synthesis. The stimulation of glycolysis by insulin is not meant to increase energy production, since glycolysis is amphibolic and provides metabolites for biosynthesis, e.g., acetyl-CoA for fatty

Figure 22-15 shows the points of entry of precursors into hepatic gluconeogenesis. Lactate and alanine enter the gluconeogenic pathway at the pyruvate carboxylase reaction. Glycerol is phosphorylated and then oxidized to dihydroxyacetone phosphate. A number of amino acids are converted to intermediates of the TCA cycle. Conversion of gluconeogenic substrates to glucose is not a direct reversal of glycolysis, since the pathway differs at three rate-limiting steps (Chapter 15).

Regulation of Gluconeogenesis Tissues involved in glucose conservation are liver, skeletal muscle, and adipose. Signals that attune the body

FIGURE 22-16 Effects of glucagon,glucocorticoids,and insulin on the pathwaysof carbohydrateand lipid metabolism.G-6-P,Glucose-6-phosphate; PEP, phosphoenolpyruvate;OA, oxaloacetate;FA, fatty acid; TG, triacylglycerol.

SECTION22.5 CarbohydrateHomeostasis

503 muscle, amino acids from muscle protein, and glycerol from adipocytes. The kidney reabsorbs the glucose in the glomerular filtrate and is a site of gluconeogenesis. In mammals, there is no net conversion of fatty acids to glucose because during oxidation of acetyl-CoA to CO2 via the TCA cycle there is no net production of oxaloacetate. However, fatty acids, play an important role in glucose homeostasis (Figure 22-17). Since brain and red blood cells use glucose, the heart functions well on fatty acids without depleting glucose levels. When glucose levels are low, only tissues that have a requirement for glucose and are insulin-independent use glucose because, with low insulin levels, there is essentially no glucose uptake by insulin-dependent tissues. After fasting for a 24-hour period, 180 g of glucose is produced from glycogen or gluconeogenesis, of which 75% is used by the brain and the remainder by red and white blood cells, etc. From the latter, about 36 g of lactate return to the liver for gluconeogenesis,

acid synthesis. Glucocorticoids stimulate fatty acid breakdown and gluconeogenesis and increase the rate of glycogen synthesis. This action appears counterproductive, but glucocorticoids are a major signal for the degradation of muscle proteins and thus for gluconeogenesis. Glucocorticoid effects are long term (whereas those of glucagon are short term), but they are synergistic and act through induction of enzymes of gluconeogenesis. Regulation by glucocorticoids may involve increases in the amounts of regulatory proteins that are involved in the actions of glucagon. Many allosteric regulators in addition to hormones fine-tune the gluconeogenic pathway at the steps catalyzed by pyruvate carboxylase, phosphoenolpyruvate carboxykinase, fructose-bisphosphatase, and glucose-6phosphatase. A summary of glucose homeostasis during fasting is shown in Figure 22-17. The sources of carbon for hepatic glucose formation are hepatic glycogen, lactate from red blood cells or skeletal

~ R B C

BRAIN

Glucose~

ATP ~'"J" Lactate

KIDNEY LIVER

Glycogen

Glutarni

(systemic blood)

Glucose ~

Lactate.

AA Glycerol

TG

Glycerol FA AA

ADIPOSE TISSUE

~

Protein

Lactate Glycogen1~

MUSCLE

A

co 2

ATP

HEART

FIGURE 22-17 Glucosehomeostasis in the fasting state. TG, Triacylglycerol;RBC,red blood cells; FA, fatty acid; AA, aminoacid.

504

CHAPTER22 MetabolicHomeostasis High-fat diet

High-carbohydratediet Glucose

TG

Glucose ~

/

INTESTINE

e

FA q

( Z O,',o,es,or,.,,,s,ers, . / Chylomicrons (lymph) "~

INTESTINE , ~ Phospholipid J VLDL d/ ~ "Apoprotein/

Chylomicrons ~ (blood)

~

~

J

LIVER

(systemic blood)

TG

Jr Lipoprotein lipase

FA

Glucose

,. Glucose--~ Glycerol~ 3-phosphate

(stor

ADIPOSE TISSUE

FIGURE 22-18 Triacylglycerol storage in high-fat and high-carbohydratediets. FA, Fatty acid; TG, triacylglycerol.

supported by amino acids from 75 g of protein and 16 g of glycerol.

The two principal types of lipid in fuel homeostasis are triacylglycerols and fatty acids. Other lipids serve roles in cellular membrane structure and other functions. Sufficient triacylglycerol normally is stored in humans to support many weeks of fasting (Table 22-2). Triacylglycerols constitute a very compact form of energy and their complete oxidation yields a high amount of energy. They are transported between organs as components of lipoproteins. Free fatty acids are amphiphilic and act as a detergent. For interorgan transport, fatty acids are bound to albumin.

small proportion of the free fatty acids, notably the shortchain acids in milk and milk products, is absorbed directly into the portal blood and transported to the liver. The bulk of the fatty acids are esterified in intestinal cells into triacylglycerol and packaged within the Golgi apparatus into chylomicrons that contain triacylglycerol, phospholipids, cholesterol and cholesterol esters, and apoproteins. Chylomicrons are secreted into lymphatic capillaries that surround the intestinal cells and fuse into larger vessels to form the thoracic duct, which opens into the systemic circulation by a one-way valve at the junction of the left jugular and subclavian veins. Chylomicrons appear in the plasma shortly after a fatty meal and reach a maximum concentration within 4-5 hours. Their large size results in light scattering and gives plasma a turbid appearance termed postalimentary lipemia.

Lipid Digestion and Absorption

Disposition of Absorbed Triacylglycerol

Dietary lipid consists mostly of triacylglycerol from plant and animal sources. It supplies about 45% of the energy in a typical Western diet. Except for the essential polyene fatty acids (Chapter 18), the dietary requirement for lipid can be met by carbohydrate or protein. However, the Eskimo has a satisfactory diet consisting of 80-90% lipid. Digestion of triacylglycerol is discussed in Chapter 12. A

In various tissues of the body, chylomicrons are acted upon by lipoprotein lipase that hydrolyzes the associated triacylglycerol. This lipase is secreted by adipocytes, hepatocytes, and cardiac and mammary tissue, and it becomes associated with the external surface of endothelial cells of the vasculature in response to the need of these tissues for fatty acids. Its half-life is a matter of hours. Its synthesis

22.6 Lipid Homeostasis

505

SECTION22.6 Lipid Homeostasis

and export are stimulated by insulin and glucose and inhibited by catecholamines. Mammary gland lipoprotein lipase activity is elevated maximally at parturition when lipid is needed for milk formation. Cardiac lipoprotein lipase is elevated during starvation. The lipoproteins transport triacylglycerol and cholesterol (Chapter 20).

Production of Triacylglycerol from Carbohydrate Processes involved in triacylglycerol storage are presented in Figure 22-18. When dietary carbohydrate exceeds requirements, it is converted into triacylglycerol within adipocytes or hepatocytes. The liver daily synthesizes 40-100 g of triacylglycerols, partly from glucose and partly from free fatty acids present in excess of its energy needs. Their transport to adipocytes occurs via VLDLs, whose synthesis, secretion, and fate are analogous to those of chylomicrons.

Release of Lipid from Adipose Tissue Stores During fasting or exercise and at times of stress, lipid is used as a source of energy. Hormone-sensitive lipase catalyzes the sequential hydrolysis of triacylglycerol to

,,,. Chylomicron-

Insulin / \ triacylglycerol ~ ~ [ Lipoprotein ~ k,~ / li a / ~

Epinephrine Glucagon (~ .~ ~

TABLE 22-3

Hormonal Regulation of Adipocyte Lipolysis Rapid Stimulators Epinephrine Norepinephrine Glucagon ACTH Secretin Vasopressin

Slow Stimulators Glucocorticoids Growth hormone

lnhibitors Insulin Prostaglandin E 1

yield ultimately 3 mol of fatty acids and 1 mol of glycerol. Hormone-sensitive lipase is regulated by phosphorylation by cAMP-dependent protein kinase. Listed in Table 22-3 are hormones that affect lipase. Epinephrine, glucagon, and the other rapid stimulators release free fatty acids within minutes. The actions of growth hormone and of glucocorticoids require hours, but their sites of action are unknown. Insulin and prostaglandin E1 suppress lipolysis by depressing cAMP levels. The major activities of adipocytes are summarized in Figure 22-19. Glucagon, an indicator of the need for

Glucocorticoid urowth hormone

Glucose

FA-all)umin corn )lex

FIGURE 22-19 Regulationof adipocytetriacylglycerolsynthesisand degradation.FA, Fattyacid; PDE,phosphodiesterase.

506

carbohydrate, increases hepatic glycogenolysis, gluconeogenesis, and ketogenesis at the expense of fatty acid oxidation and enhances adipocyte lipolysis. Epinephrine and glucocorticoids are, respectively, acute and chronic indicators of stress. They increase hepatic glucose output and peripheral tissue fatty acid utilization. Growth hormone regulates many processes associated with growth of the organism. The energy for much of this growth derives from fatty acid oxidation. As fatty acids leave the adipocyte, they form complexes with albumin. The availability of separate pathways for fat deposition and release in these cells permits appropriate regulation under anabolic or catabolic circumstances. The absence of glycerol kinase in the adipocyte decreases the potential for resynthesis of the triacylglycerol and promotes hepatic gluconeogenesis. Triacylglycerols are deposited in the adipocyte when glucose and insulin are readily available.

Tissue Utilization of Fatty Acids The level of plasma albumin-bound fatty acids increases when lipolysis rates are high. The tissue uptake of fatty acids is proportional to their concentration in plasma and is therefore largely dependent on blood flow. During intense exercise, the flow of blood through the splanchnic bed is reduced and more fatty acids are available to skeletal muscle. With the exception of nerve tissue and blood cells, tissues can use fatty acids by/3-oxidation and by the TCA cycle. Fatty acid uptake is not regulated by hormones or intracellular effectors. Free fatty acids readily diffuse across the plasma membrane of cells where they are used strictly in response to supply and demand. This is illustrated for cardiac muscle in Figure 22-20. Fatty acid oxidation requires adequate amounts of NAD and FAD. Oxidation of NADH and FADH2 occurs in mitochondria via electron transport and the use of molecular oxygen. However, the electron transport chain can only function if ADP is present, so that fatty acid breakdown occurs at the rate necessary to maintain cellular levels of ATP which decrease if metabolic or other work is performed. Tight coupling of electron transport to oxidative phosphorylation (Chapter 14) is of considerable importance in fuel economy. Without it, fatty acids would be utilized in an unproductive manner. In the liver, fatty acids in excess of requirements are converted to triacylglycerols and secreted as VLDLs. Many tissues preferentially utilize fatty acids as an energy source. The sparing effect on glucose is illustrated in Figure 22-21 and reflects the tight coupling of oxidation of fatty acids to ATP production. When sufficient fatty acids are available ATP concentrations will be high and ADP concentrations low. ADP

CHAPTER22 Metabolic Homeostasis

F I G U R E 22-20 Regulation of fatty acid oxidation in cardiac muscle.

is required in two reactions in glycolysis; ATP is an allosteric inhibitor, and AMP is an allosteric activator of phosphofructokinase.

Ketone Body Production and Utilization Fatty acids released from adipose tissue are the source of ketone bodies (Figure 22-22). Fasting, high levels of glucagon and catecholamine, and a low level of insulin result in rapid lipolysis and ready availability of fatty acids. Fatty acids, after being converted to CoA thioesters, are oxidized in the mitochondria. The rate-limiting step in the oxidation process is the transport of fatty acyl-CoA

FIGURE 22-21 Glucose-sparingeffect of fatty acid oxidation. Note the inhibitionof glucose utilizationby high levels of ATP and citrate. OA, Oxaloacetate; F-1, 6-bisP,fructose-1,6-bisphosphate; 1,3-bisPG, 1,3-bisphosphoglycerate; 3-PG, 3-phosphoglycerate;PEP,phosphoenolpyruvate;Pyr, pyruvate.

SECTION22.6 LipidHomeostasis

507

Epinephrine

ucagon ~ Gl Glucocorticoids -~ acylglycerol ~ ac,os )Fatty~ FA-albumin

Insulin,~

ADIPOSETISSUE

//

/

/

/

MUSCLE

\

Oxidation

/ /

NADH FA

oso ~

t Oxa,oaco, ,

AcetoacetyI-CoA .."

,J \ Oxida;on .........

/ /

~"

D

.

HMG-CoA J

NADH,~,-y, J NAD~ " /

~ 13-Hydroxybutyrate~

I~

A~toacetyI-CoA

-F--''c"

TCA cycle

1"_- ~ Succinate~ ~ GTP\ c --

~

1~ X~

~ HB . _ . . I , ~

,/-'/

,&c

~ NADH+H" ot-Ketoglutarate/ r " NAD" /

HB

/

F I G U R E 22-22 Ketone body production and utilization. Ketone bodies are produced in the liver from fatty acids derived from adipocyte lipolysis. They are released and used as fuel in peripheral tissues. The initial step in acetoacetate metabolism is activation to acetoacetyl-CoA by succinyl-CoA. HMG-CoA,/3-hydroxy-/3-methylglutaryl-CoA; HB,/3-hydroxybutyrate.

across the inner mitochondrial membrane. The transport is a transesterification process and involves carnitine and two enzymes, carnitine palmitoyltransferase (CPT) types I and II. CPTI is inhibited by malonyl-CoA, primary substrate for fatty acid synthesis that occurs in the cytosol. In the presence of a low insulin:glucagon ratio, malonyl-CoA formation is inhibited, leading to active CPTI and promoting fatty acid oxidation and consequent ketone body production. The opposite situation occurs at a high insulin: glucagon ratio. Hepatic fl-oxidation, without oxidation of acetyl-CoA through the TCA cycle, produces a substantial amount of energy. At such a time, liver is actively engaged in gluconeogenesis so that mitochondrial oxaloacetate is depleted, TCA cycle activity is depressed, and acetyl-CoA levels rise. The last reaction in/3-oxidation is conversion of acetoacetyl-CoA to acetyl-CoA, with an equilibrium in favor of high levels of acetoacetyl-CoA. Thus, acetyl-CoA and acetoacetyl-CoA accumulate and form HMG-CoA; cleavage of this last compound yields acetoacetate, which is reduced to /3-hydroxybutyrate. Acetone results from nonenzymatic decarboxylation of acetoacetate. Ketone body formation occurs exclusively in liver (see Chapter 18) and is prominent in starvation and diabetes owing to the

low insulin:glucagon ratio in these conditions. The ketone bodies readily leave the liver and enter the bloodstream. Acetone is eliminated in the urine and exhaled by the lungs. Because of its odor, it has long been diagnostic in urine and in breath in clinical situations. Because production of ketone bodies was associated with starvation and disease, these compounds were not considered to be metabolically useful. During periods of starvation, acetoacetate and /3-hydroxybutyrate make a considerable contribution to fuel homeostasis. The ratio of their plasma concentrations in starvation and diabetes is about 1:5, reflecting high hepatic levels of NADH, which favors the reduction of acetoacetate to/3-hydroxybutyrate. In utilization of ketone bodies in skeletal muscle, /3-hydroxybutyrate is converted to acetoacetate and then to acetoacetyl-CoA, which is cleaved, and the acetyl-CoA formed is oxidized through the TCA cycle. Activation of acetoacetate to acetoacetyl-CoA requires conversion of succinyl-CoA to succinate. Under normal conditions, the brain cannot use ketone bodies because it lacks the enzyme needed to activate acetoacetate. However, this enzyme is induced in brain after about 4 days of starvation, permitting the brain to obtain 40-70% of its energy from ketone body oxidation while

508

needing less glucose from the liver. Two ketone bodies are acids and give rise to acidosis which is decreased by their elimination in urine. In a 24-hour fast, a 70-kg person hydrolyzes approximately 160 g of triacylglycerol, with release of 160 g of fatty acid and 16 g of glycerol. The glycerol is used for gluconeogenesis. Of the fatty acids, approximately 40 g is metabolized to ketone bodies in liver; the remainder and these ketone bodies provide for the energy needs of all other tissues.

22.7 Protein Synthesis and Nitrogen Homeostasis Protein Synthesis and Proteins as Energy Source While carbohydrates and lipids are essential for structural and functional needs of cells and organs, the principal changes in their fluxes reflect fuel homeostasis and energy requirements. Protein homeostasis is different because no protein serves purely as an energy storage form. All proteins--in addition to being potential sources of carbon for energy--serve some structural, catalytic, transport, or regulatory function. Many fluctuations observed in protein metabolism reflect changes in the need for the function of that protein. Thus, in starvation the total rate of skeletal muscle protein synthesis decreases but that of protein degradation increases because the amounts of proteases and enzymes involved in the catabolic routes for amino acids increase. Thus, starvation stimulates the specific synthesis of some proteins. Protein anabolism and catabolism are governed by many forces. Some rules apply: I. Which specific proteins are synthesized in a cell is governed entirely by the needs or functions of the cell at that time. 2. Synthesis of specific proteins is triggered by a specific signal. 3. Synthesis of a specific protein may not occur if the general "anabolic state" of the cell is low. 4. The anabolic state is reflected by metabolite availability and the presence of anabolic or catabolic hormones. 5. Synthesis occurs if the strength of the specific trigger overrides that of catabolic messages or other hormones or metabolites. 6. Degradation of a protein may occur if a) the protein has no function in the cell, b) a specific trigger to initiate degradation is present, or c) the state of the cell is sufficiently catabolic as a result of the presence or absence of specific metabolites or hormones.

CHAPTER22 Metabolic Homeostasis

Many hormones modulate the amounts and types of proteins synthesized and their rates of degradation. One group functions on very few target cells, e.g., TSH (thyroidstimulating hormone) on the thyroid gland, FSH (folliclestimulating hormone) and LH (luteinizing hormone) on the gonads, or ACTH (adrenocorticotropic hormone) on the adrenals. Another group has widespread action on protein metabolism, e.g., insulin, glucocorticoids, growth hormone, and thyroid hormone. Many sites of action of insulin and glucocorticoids remain to be elucidated. Glucocorticoids act at the gene level and promote peripheral tissue protein degradation, delivery of amino acids for gluconeogenesis, and adipocyte lipolysis. Insulin stimulates peripheral tissue protein synthesis by stimulating amino acid uptake and protein synthesis at the level of translation and by inhibiting protein degradation. At low insulin levels, muscle proteolysis occurs. As the levels increase, proteolysis decreases and protein synthesis is favored. Exercise decreases proteolysis and increases protein synthesis, whereas disuse results in muscle wasting and depressed protein synthesis. Exercise increases sensitivity to insulin, whereas disuse makes the tissue insulinresistant. Obesity, pregnancy, and glucocorticoids also increase insulin resistance.

Nitrogen Balance Unlike most carbohydrate and lipid, protein contains nitrogen required for incorporation into a wide range of compounds (Table 22-4). Although the synthesis of proteins fixes nitrogen into those proteins, they are synthesized in direct response to specific needs. Dietary protein nitrogen is used for the synthesis of nitrogen-containing compounds in response to specific hormones suchas insulin.

TABI,E 22-4

Compounds for Which Amino Acids Serve as Precursors

Amino Acid Precursor Glycine, aspartate, glutamine Aspartate, glutamine Glycine Arginine, methionine Arginine, glycine Tryptophan Tyrosine Histidine Arginine

Product Purines Pyrimidines Porphyrins Polyamines Creatine Serotonin, melatonin Catecholamines, dopamine, melanin Histamine Nitric oxide

SECTION 22.7

509

Protein Synthesis and Nitrogen Homeostasis

Excess nitrogen is excreted primarily as urea (Chapter 17). Since nitrogen is essential but cannot be stored, the body attempts to maintain "nitrogen balance." In positive nitrogen balance, more nitrogen is ingested than is eliminated. This occurs during growth, pregnancy, lactation, and recovery from trauma, such as major surgery. If less nitrogen is ingested than is excreted, the body is in negative nitrogen balance. In times when dietary nitrogen is not available, negative nitrogen balance occurs because some nitrogen is always excreted as urea and ammonia. Of the 20 amino acids in proteins, the body can readily synthesize eight if an appropriate nitrogen source is available. Two others can be synthesized from other amino acids of the diet: tyrosine from phenylalanine and cysteine from methionine. The rest must be provided in the diet (Chapter 17), since the body can synthesize none or an insufficient amount. The dietary requirement depends on several factors. Beside essential amino acids, the diet should provide the nitrogen required for synthesis of the nonessential amino acids.

Ammonia Toxicity Ammonia, a metabolite formed from many nitrogencontaining compounds, is used for biosynthesis. High ammonia levels are toxic. The energy requirements of the brain, which is sensitive to high levels of ammonia, are met almost exclusively by glucose oxidation, for which a source of oxaloacetate is essential. This condition is satisfied by the carboxylation of pyruvate, even though the pyruvate carboxylase is somewhat limiting. For efficient TCA cycle activity, considerable recycling of oxaloacetate is necessary, since de novo synthesis is limited. With high levels of ammonia, the equilibrium for glutamate dehydrogenase favors formation of glutamate and glutamine, which diminishes the ammonia level but decreases TCA cycle activity. The brain is thus deprived of its source of ATP generation. In addition, glutamate and aspartate have neurotransmitter functions. Within the liver, elimination of ammonia occurs via urea synthesis (Chapter 17). Since urea is uncharged, it does not disturb the acid-base balance. Many interorgan relationships in protein and nitrogen homeostasis arose because of the role that the liver plays in excess nitrogen excretion.

Nitrogen Transfer between Compounds and Tissues Nitrogen is eliminated from the body as urea and ammonia. Urea synthesized in the liver is excreted by the kidneys. Urinary ammonia is produced in the kidney. The nitrogen in other tissues is transported to the liver and kidney in

the form of amino acids. The general reactions involved in these processes are described below. Methods for Directly Transferring Nitrogen

Nitrogen for biosynthetic processes is derived from the o~-amino group of amino acids and from the amido group of glutamine and asparagine by transamination and transamidation, respectively (Chapter 17). Because of the wide variety of transaminases, transamination can provide the right balance of all nonessential amino acids and nitrogencontaining compounds derived from them. Transamidation is less ubiquitous. Other reactions utilizing the nitrogen of amino acids include the incorporation of glycine into purines and its partial incorporation into porphyrins. Reactions in Which Ammonia Is Released

Ammonia is produced by oxidative and nonoxidative deaminations catalyzed by glutaminase and glutamate dehydrogenase (Chapter 17). Ammonia is also released in the purine nucleotide cycle. This cycle is prominent in skeletal muscle and kidney. Aspartate formed via transamination donates its or-amino group in the formation of AMP; the amino group is released as ammonia by the formation of IMR Reactions That "'Fix" Ammonia

In muscle, ammonia is used to synthesize other nitrogen-containing compounds or is eliminated. In either case, glutamine and glutamate are formed by glutamine synthetase and glutamate dehydrogenase, respectively. The glutamate dehydrogenase reaction is readily reversible, but that of glutamine synthetase is not because it is driven by the hydrolysis of ATE Glutamine synthesis is the major mechanism for nonhepatic tissues to eliminate ammonia. In liver, ammonia is fixed by the formation of urea (Chapter 17).

Disposition of Dietary Intake of Protein Ingested protein is digested in a stepwise fashion in the stomach, small intestinal lumen, and small intestinal mucosal cells (Chapter 12). Peptides formed in the intestinal lumen are absorbed into the mucosal cells and degraded to free amino acids. The outflow of amino acids to the portal vein does not reflect the amino acid composition of the ingested protein. Thus, alanine levels increase twoto fourfold, and glutamine, glutamate, and aspartate are absent. These changes arise from amino acid interconversions within the intestinal cell. With the exception of the branched-chain amino acids (valine, leucine, and isoleucine), most amino acids

510

CHAPTER22 Metabolic Homeostasis

released from the intestine are metabolized by the liver. The liver carries out 1. Formation of other molecules from amino acid precursors, e.g., nonessential amino acids, purines, pyrimidines, porphyrins, and plasma proteins (such as albumin, VLDL, and transferrin), for its needs and those of other tissues; 2. Utilization of the amino acid carbon skeletons in gluconeogenesis or glycogen or lipid synthesis; and 3. Formation of urea. After a protein meal, the plasma amino acids are those delivered from the intestine and those synthesized by the liver. The levels of branched-chain amino acids are greatly increased because they bypass the liver, reaching a peak at about 4 hours and returning to normal by 8-10 hours. They account for up to 60% of the total amino acids entering the systemic circulation. After uptake by skeletal muscle and transamination, followed by oxidation of the oe-keto acid derivatives by the pathways described in Chapter 17, they can meet most energy requirements of muscle. The branched-chain amino acids have been called amino fats. Their or-amino nitrogen is converted to alanine, which is transported to liver for elimination as urea. Figure 22-23 summarizes the various pathways for amino acids following protein ingestion. Alanine is formed in intestine and muscle from many of the amino acids. Following a protein meal, insulin secretion increases, which stimulates peripheral tissue uptake of amino acids and net protein synthesis. On a diet high in carbohydrate and low in protein, insulin levels are elevated and glucagon levels are depressed. On a diet high in protein and low in carbohydrate, insulin and glucagon levels are elevated. While amino acids and glucose stimulate insulin release, amino acids stimulate and glucose inhibits glucagon release. Thus, on a high-carbohydrate diet, insulin promotes hepatic and peripheral tissue utilization of glucose without the need for hepatic gluconeogenesis. Because of the low level of glucagon, hepatic glucose formation is depressed. On a high-protein, low-carbohydrate diet, insulin secretion is stimulated by amino acids and results in their uptake by peripheral tissue and utilization for glucose uptake. However, glucose homeostasis is maintained because glucagon stimulates hepatic glucose release.

Protein Catabolism during Starvation During starvation, following depletion of hepatic glycogen, amino acids become the major source for glucose homeostasis because of the decrease in plasma insulin

Alanine + non-BCAA

BCAA N ot-Ketoacids N

Mus

I

~176

X

\

F I ( ; U R E 22-23 Amino acid metabolism following dietary protein intake. Digestion of protein produces amino acids. Within the intestinal cell, amino acid interconversions form alanine, which is delivered by the portal blood to liver, where it serves as the source of c~-amino nitrogen and pyruvate, which is converted to lipid and glucose. Excess nitrogen is converted to urea. Branched-chain amino acids (BCAAs) are not taken up by the liver but enter peripheral tissues such as muscle where they serve as an important fuel source. Their c~-amino nitrogen is transported to liver in the form of alanine.

level and the rise in glucocorticoid level. The pattern of amino acids released by skeletal muscle during starvation does not reflect the composition of muscle protein. Alanine and glutamine account for over half of the amino acids released. Alanine is taken up by liver, its carbon chain converted to glucose, and the nitrogen to urea. In early starvation, the principal site of glutamine metabolism is the gut. One product is alanine. The special role of glutamine in gut may be due to the high demand for glutamine in purine synthesis because of the active shedding of intestinal cells. In long-term starvation, a major site of glutamine metabolism is the kidney, since the excretion of ketone bodies requires NH + as a counterion formed from ammonia produced by glutaminase. The resulting glutamate is

511

SECTION22.8 Abnormalities of Metabolic Homeostasis Glucose production

200-

175-

150-

Glucose

125-

~L~nie

I00-

LIVER

Glutamine

Ala

~

75-

Urea Liver

25

-

~

-

..

-

..

-

-

-

.. . . . . . . . . .

_

_

..

_

..

..

. . . . . . . . . ..

..

~~,~ac;,200 mg/dl (11.1 mmol/1). Casual is defined as any time of day without regard to time since last meal. The classic symptoms of diabetes include polyuria, polydipsia, and unexplained weight loss. or 2. FPG >126 mg/dl (7.0 mmol/1). Fasting is defined as no caloric intake for at least 8 h. or 3. 2hPG >200 mg/dl during an OGTT. The test should be performed as described by WHO using a glucose load containing the equivalent of 75-g anhydrous glucose dissolved in water. In the absence of unequivocal hyperglycemiawith acute metabolic decompensation, these criteria should be confirmed by repeat testing on a different day. The third measure (OGTT) is not recommended for routine clinical use. FPG = Fasting plasma glucose; OGTT = Oral glucose tolerance test; 2hPG = 2 hour post load glucose. The diagnostic criteria are based upon the report by the Expert committee on the diagnosis and classification of Diabetes mellitus. Diabetes Care24(suppl. 1), 55 (2001).

in Table 22-5. Subjects who have fasting plasma glucose levels between _>110 mg/dL and 140 mg/dL (7.8 mmol/L), the test is considered positive and requires a diagnostic test consisting of oral ingestion of 100 g of glucose after a fasting for 8 hours, followed by 3-hour oral glucose tolerance test (Table 22-6). In the 50-g challenge test, if the threshold for the abnormality is decreased to _>130 mg/dL (7.2 mmol/L), the sensitivity for identifying patients with gestational diabetes mellitus increases from 80% to 90%. Since diabetes mellitus is an insidious disorder, testing of asymptomatic patients may be desirable under certain conditions, including age 45 years or older; obesity; firstdegree relatives of diabetics; members of high-risk ethnic population (e.g., Native American, Hispanic, AfricanAmerican); women who have delivered an infant weighing more than 9 lb (4.08 kg) or have had gestational diabetes mellitus; hypertension; abnormal lipid studies; recurring

513

TABLE 22-6

Screening and Diagnosis Scheme for Gestational Diabetes Mellitus (GDM)

Plasma Glucose

50-g Screening Test

100-g Diagnostic Test

140 mg/dl

180 mg/dl

Fasting

1-h 2-h 3-h

95 mg/dl 155 mg/dl 140 mg/dl

Screening for GDM may not be necessary in pregnant women who meet all of the following criteria: < 25 years of age, normal body weight, no first degree relative with diabetes, and not Hispanic, Native-American, Asian-, or African-American. The 100-g diagnostic test is performed on patients who have a positive screening test. The diagnosis of GDM requires any two of the fourplasma glucose values obtained duringthe test to meet or exceed the values shown above. To convert values for glucose to mmol/L, multiply by 0.05551. The diagnostic criteriais based upon the report by the American Diabetes Association on Gestational Diabetes Mellitus. Diabetes Care 23(suppl. 1), $77 (2000). skin, genital, or urinary tract infections; and previous impaired glucose intolerance. After the diagnosis of diabetes mellitus has been made and with the initiation of appropriate therapy (discussed later), assessment of other biochemical parameters is necessary in the management phase of the disorder to maintain the fasting blood glucose level as close to normal as possible and to prevent long-term complications. These biochemical tests include measurement of a stable form of glycosylated hemoglobin (hemoglobin Alc), determination of urine albumin excretion rate, measurement of serum fructosamine levels, and self-monitoring of blood glucose levels. Hemoglobin Aac levels are used to assess average glucose control over a 2 to 3-month period, since the, re,d hlc~c~d ce,ll'.~ life_span is about 120 days, EntLy~f glucose into red blood cells depends only on the prevailing plasma glucose concentration. (Formation of hemoglobin Alc, which is nonenzymatic, is discussed in Chapters 2 and 10). The normal hemoglobin Ale concentration is about 4-6%; spurious values for hemoglobin Alc levels can occur in uremic states, hemoglobinopathies (Chapter 28), hemolytic anemia and blood transfusion. Fructosamine is a generic term applied to the stable condensation product of glucose with serum proteins (albumin, with a circulating half-life of about 20 days, is the major contributor). Thus, serum fructosamine levels reflect glucose control over a period of 2-3 weeks. One of the chronic complications of diabetes mellitus is diabetic nephropathy, which leads to end-stage renal disease. An initial biochemical parameter of diabetic nephropathy in the asymptomatic state is a persistent

514

increase in urine albumin excretion rate between 20 and 200 #g/min (or 30-300 mg/d). This degree of albumin loss in the urine is called microalbuminuria and is a harbinger of renal failure and other complications of diabetes. It is important to identify individuals with microalbuminuria because with appropriate therapeutic intervention attenuation of loss of renal function can be accomplished. Therapeutic interventions include blood glucose control, treatment for high blood pressure if present, and the inhibition of formation of angiotensin II by use of angiotensinconverting enzyme (ACE) inhibitors. The formation of angiotensin II involves the following steps: release of the protease renin by justaglomerular cells in response to decreased renal perfusion pressure, release of angiotensin I from angiotensinogen by the action of renin, and conversion of angiotensin I to angiotensin II by ACE. Physiologic functions of angiotensin II in the kidney include restoration of normal renal blood flow and glomerular filtration and stimulation of aldosterone secretion, which affects Na + reabsorption (Chapter 32). In diabetes mellitus, renal hemodynamics are altered due to glomerular hyperfiltration. Angiotensin II has been postulated to contribute to the progression of renal disease. Some of the deleterious effects of angiotensin II may involve its action on vasomotor functions and induction of cytokines (e.g., TGF-/3, see Chapter 35) that lead to hypertrophy of mesangial cells. Thus, ACE inhibitors as well as angiotensin II receptor antagonists can prevent or slow the rate of progression of renal disease in diabetes mellitus. Clinical studies have shown that even in normotensive diabetic patients, ACE inhibitors are beneficial in the management of renal disease.

Chronic Complications of Diabetes Mellitus These stem from elevated plasma glucose levels per se and involve tissues that do not require insulin (e.g., lens, retina, peripheral nerve) for the uptake and metabolism of glucose. In these tissues, the intracellular level of glucose parallels that in plasma. The chronic complications, which cause considerable morbidity and mortality, are atherosclerosis, microangiopathy, retinopathy, nephropathy, neuropathy, and cataract. The biochemical basis of these abnormalities may be attributed to increased tissue ambient glucose concentration and may involve the following mechanisms: nonenzymatic protein glycation (Chapter 2), increased production of sorbitol, and decreased levels of myo-inositol. The ramifications of glycation of proteins are not clear. Sorbitol is produced from glucose by NADPH-dependent reduction catalyzed by aldose reductase. Sorbitol may also be converted to fructose by NAD+-dependent oxidation. Sorbitol (and fructose) diffuses poorly across cell membranes and accumulates

CHAPTER22 Metabolic Homeostasis

inside the cell causing osmosis-induced disturbances (e.g., cataract formation). The myo-inositol depletion may be due in part to the competition of glucose with myo-inositol for its intracellular transport; glucose and myo-inositol are strikingly similar in structure (Chapter 9). Myo-inositol can also be synthesized from glucose-6-phosphate. Decreased myo-inositol may lead to decreased phosphoinositide turnover. The latter yields at least two active catabolites (inositol polyphosphates and diacylglycerol) that function as second messengers (Chapter 30). The diminished activity of Na+,K+-ATPase found in nerve fibers is thought to be related to the altered phosphoinositide turnover. Potential therapeutic use of myo-inositol supplementation and aldose reductase inhibitor administration is being explored.

Management of Diabetes Mellitus The primary goal in the management of all types of diabetes mellitus (type 1, type 2, and GDM) is to maintain nearnormal plasma glucose levels in order to relieve symptoms (polydipsia, polyuria, polyphagia) and to prevent both acute and chronic complications. The glycemic control is assessed by monitoring of glucose (by self and in clinical settings), hemoglobin Alc, fructosamine, and microalbuminuria. In type 1 diabetes mellitus due to /~-cell destruction, administration of insulin is required throughout the person's lifetime. There are many insulin preparations that differ in duration of action (ultrashort, short, intermediate, and long acting) and in their origin (human, porcine, and bovine). Human insulin analogues lispro and aspart (B28) do not undergo polymerization and are rapid acting insulins. The side effects of insulin therapy include hypoglycemia and weight gain. The management of type 2 diabetes mellitus is based on both behavioral changes with lifestyle modifications and pharmacological measures. Diet, exercise, and weight loss (in the obese) are the cornerstone of treatment in maintaining euglycemia. Physical activity stimulates insulin sensitivity, whereas physical inactivity leads to insulin resistance. In the absence or failure of lifestyle and behavioral measures in the management of type 2 diabetes mellitus, pharmacological therapy is employed. Pharmacological agents used in the treatment of type 2 diabetes mellitus include insulins, sulfonylureas, ot-glucosidase inhibitors, biguanidines, and thiazolidinediones (Figure 22-26). Sulfonylureas bind to specific fl-cell plasma membrane receptors and cause closure of the ATP-dependent K + channel with accompanying depolarization. The depolarization leads to influx of Ca 2+ followed by insulin secretion. A benzoic acid derivative (repaglinide) is structurally similar to sulfonylurea without the sulfur

SECTION22.8 Abnormalities of Metabolic Homeostasis

515

Acarbose (an tx-Glucosidaseinhibitor) C,i_i2OH ,,,5.,,~

CH3 0

CH2OH ~" O

CH2OH ~ / O ~ , d "-tOH

HO HO

Glipizide (a sulfonylurea)

H3c-(-

0 [CI--NH--CH2--CH 2 ~

0 802 --NHJCI --NH--~

Metformin (a biguanidine)

Troglitazone (a thiazolidinedione) CH3

NH NH H3C II II H3C> N - - C - - N H - - C - - NH2

)

CH.

/7"'7N

.o-

o

N.

CH3 FIGURE 22-26

Structures of four classes of hypoglycemicagents. group and belongs to the class meglitinides. This drug inhibits a different K + channel than SUR in promoting secretion and has rapid-onset and short-acting actions. An example of an ot-glucosidase inhibitor is acarbose, a nitrogen-containing pseudotetrasaccharide. Acarbose inhibits the breakdown of carbohydrates into glucose by inhibiting intestinal brush-border ot-glucosidase and pancreatic amylase (Chapter 12) and brings about a reduction in postprandial hyperglycemia. Metformin belongs to the class of biguanidines and has many effects on carbohydrates and lipid metabolism. Its mechanism of action may include inhibition of hepatic gluconeogenesis and glycogenolysis and, in muscle, increased insulin receptor tyrosine kinase activity and enhanced GLUT4 transporter system. An adverse effect of metformin is lactic acidosis. Troglitazone is an example of the thiozolidinedione class of antidiabetic agents. It enhances insulin sensitivity in liver, muscle, and adipose tissue. Troglitazone promotes the conversion of non-lipid-storing preadipocytes to mature adipocytes (discussed later) with increased insulin sensitivity. The cellular target for the action of troglitazone, which functions as a transcription factor, is the nuclear receptor known as peroxisome proliferationactivated receptor-F (PPAR-F). PPAR-y upon binding

with troglitazone is activated and forms a heterodimer with retinoid X receptor (RXR). The PPAR-y/RXR heterodimer regulates the transcription of genes encoding proteins that control metabolic pathways (e.g., acetyl CoA-synthetase, lipoprotein lipase, GLUT4, mitochondrial uncoupling protein). An adverse effect of troglitazone is hepatic toxicity; periodic monitoring of liver function by measurement of serum alanine aminotransferase is recommended. When a diabetic patient becomes refractory to monotherapy, combination therapy (e.g., sulfonylurea and insulin) is used to achieve glycemic control.

Obesity Overweight and obesity are defined based on the body mass index (BMI), which is calculated as weight in kilograms divided by the square of height in meters (BMI -- kg/m2). A BMI between 25 and 29.9 is considered as overweight and >_30 is considered obese (Chapter 6). Body weight is determined by a balance between energy intake and energy expenditure. The energy expenditure is required to maintain basal metabolic functions, absorption and digestion of foods (thermic effect of food), physical activity and, in children, linear growth

~16

CHAPTER22 Metabolic Homeostasis

and development. If there is a net excess energy intake, BMI increases, eventually leading to overweight and ultimately to obesity and morbid obesity. In a population with stable genetic factors, increase in obesity is primarily attributable to consumption of excess foods with high fat content and decreased physical activity. Obesity is a worldwide health problem. It is a risk factor for development of diabetes mellitus, hypertension, and heart disease, all of which cause decreased quality of life and life expectancy. Feeding behavior and energy balance are regulated by a complex set of short-term and long-term physiological signals. Mechanisms that lead to obesity involve interactions between genetic, environmental, and neuroendocrine factors. The short-term regulators of hunger and satiety may include plasma levels of glucose and amino acids, cholecystokinin and other hormones, and body temperature. One of the long-term regulators of food intake and energy expenditure is an adipocyte-derived hormone leptin (Chapter 5). Leptin is a polypeptide of 167-amino acid

PPARy2\ (Active)~

Phosphorylation*

Preadipocytes

residues that functions in the afferent signal pathway of a negative feedback loop in regulating the size of adipose tissue and energy balance. The physiological role of leptin was established by using ob/ob and db/db mice that have identical phenotypes of obesity and diabetes mellitus. In the ob/ob mice, leptin levels are low due to mutations in the leptin gene, and in the db/db mice the leptin receptors are defective due to inactivating mutations. The role of leptin and its receptor was investigated by joining the circulatory systems (parabiosis) of ob/ob and db/db mice. In ob/ob mice, obesity, diabetes, and sterility are corrected by leptin administration. These animal studies have led to understanding of several aspects of obesity in humans. In humans, leptin is secreted in a pulsatile manner, in a nyctohemeral rhythm, and in proportion to size of the adipose tissue. The pathway of leptin's action involves the neuroendocrine system of the hypothalamus (Figure 22-27). When leptin levels are low, appetite increases via the activation of the neuropeptide Y

~ PhosphorylatedPPAR72 (Inactive)

J, Adipocytes

1 l

Leptin*

~

Hypothalamic LeptinReceptor* ~

~

Low Leptin Levels

~

Increased FoodConsumption

.

High Leptin Levels

NeuropeptideY/ AGRP ~ Neuropeptide Y Receptors*

~

~

Inhibition ~

POMC ]

Proteolysis*

MSH + other peptides Melanocortin-4 Receptors* Decreased Food Consumption

FIGURE 22-27 A modelfor the regulationof humanfood consumption.Asterisks(*) indicateobesity-causingmutations.Abbreviations are givenin the text.

SECTION 22.9

Metabolic Homeostasis during Exercise

pathway, eventually leading to stimulation of food intake. Neuropeptide Y contains 36 amino acid residues and has tyrosine at both the N and C termini (hence the designation Y). The actions of neuropeptide Y result in the stimulation of food intake followed by the restoration of depleted adipose stores and inhibition of sympathetic nervous system outflow with a consequent decrease in energy expenditure. When leptin levels are high, food intake decreases; this is mediated by the pro-opiomelanocortin (POMC)-ot-melanocyte stimulating hormone and melanocortin-4 receptor pathway. Inhibition of the melanocortin-4 receptor signal transduction system by the agouti protein leads to obesity in mice. Agouti protein (AGRP) is normally synthesized in hair follicles and antagonizes the action of ot-melanocytestimulating hormone in eumelanin synthesis (Chapter 17) by binding to melanocortin-4 receptor. In autosomaldominant mice that overexpress agouti protein in skin and hypothalamus, the phenotype is obese, yellow mice. In humans, the pathways of regulation of appetite and food consumption have been verified by identifying mutations in genes that participate in regulation of body weight (Figure 22-27). The regulators of conversion of preadipocytes to adipocytes also determine the adipose tissue mass. One of the regulators is a transcription factor known as peroxisome proliferator-activated receptor-F2 (PPAR-F2). Troglitazone, a hypoglycemic agent (previously discussed) that increases insulin sensitivity, is a synthetic ligand for PPAR-F2. However, contrary to studies in rodents, administration of troglitazone in humans does not cause significant weight gain as compared to other oral antidiabetic drugs. Phosphorylation of PPAR-F2 at ser114 makes the receptor less active in converting preadipocytes to adipocytes. A mutation converting proline to glutamine at position 115 blocks phosphorylation at the serine site, maintains the protein in the active state, and leads to obesity. Leptin therapy has corrected obesity in a child with congenital leptin deficiency. In obese individuals, the presence of circulating high levels of leptin has been attributed to resistance or some other defect in the leptin receptors. This apparent paradox of high leptin levels associated with obesity is analogous to insulin resistance seen in type 2 diabetes mellitus. In general, in the vast majority of obese patients, the molecular defects remain unknown. Diet and exercise are the mainstays in the management of obesity.

22.9 Metabolic Homeostasis during Exercise At rest, skeletal muscle utilizes the catabolism of fatty acids and branched-chain amino acids to maintain cellular

517

integrity. In exercise, oxygen utilization by muscle may increase 30-fold, and additional energy can arise from anaerobic glycolysis. The substrates used depend on the intensity of exercise and on training. High-intensity exercise can start almost instantaneously (e.g., the 100-m dash or the clean-and-jerk in weight lifting), but increase of oxygen utilization is limited by the rates of blood flow, respiration, and oxygen delivery to the tissue. After the onset of high-intensity exercise, the body and muscle are initially in oxygen deficit until a new steady-state, increased rate of oxygen uptake can be achieved. When the exercise ends, this oxygen debt is repaid during the recovery phase. The availability of oxygen determines the metabolic fuels utilized. Skeletal muscle has fewer mitochondria per unit weight than heart or liver. A major adaptation induced by endurance exercise training is an increase in the potential for delivery and utilization of oxygen: the heart hypertrophies; total blood volume increases; muscle capillarity increases; and skeletal muscle mitochondrial size, number, and capacity increase (see Chapter 21).

Exercise Generating Maximum Power The substrates for muscular activity are intracellular ATE phosphocreatine, glycogen, plasma glucose, fatty acids, ketone bodies, triacylglycerols, and dietary branchedchain amino acids. In exercise that generates maximum power, the primary sources are ATP and phosphocreatine. The total amount of ATE and of ATP generated by the adenylate kinase reaction (2ADP --+ ATP + AMP), is sufficient for only a fraction of a second. The immediate reserve is ATP produced by the creatine kinase reaction (phosphocreatine + ADP --+ ATP § creatine). Maximumpower-output exercise rapidly depletes phosphocreatine, can be sustained for short periods (15-20 seconds) and ends as phosphocreatine is depleted. Some anaerobic glycolysis from glycogen may occur during such a period but would reach a maximum rate only after the work had stopped. Maximum-power-output work exceeds the capacity of muscle to generate energy oxidatively even if this could be initiated rapidly.

High-Intensity Endurance Exercise This exercise is typified by an individual working in the range of 60-80% of maximum capacity of oxygen uptake (e.g., running distances of 5 km up to a marathon). The exercise is terminated by exhaustion. Phosphocreatine is the initial energy source; glycogenolysis ensues, anaerobically at first, with lactate production, but with increasing aerobic oxidation as oxygen availability increases. Glycogen utilization is the major fuel for the first 15 minutes, but

518 it is superseded by fatty acid and plasma glucose utilization. Most of the plasma glucose used is oxidized entirely but 10-20% is converted to lactate. During the exercise and depending on the past dietary history, branched-chain amino acid oxidation contributes some energy, and alanine is formed to eliminate the or-amino nitrogen. Lactate and alanine are used in the liver for gluconeogenesis and can provide 5-15% of the glucose required by skeletal muscle. The rest of the glucose is derived from glycogenolysis. Thus, in endurance exercise, hepatic glycogen serves as a major fuel for muscle, and muscle glycogen is the initial fuel. As plasma glucose and fatty acids are used, glycogenolysis in muscle diminishes; despite its smaller contribution, muscle glycogen plays a critical role since exhaustion occurs with its total depletion in spite of the availability of fatty acids and plasma glucose. Thus, "glycogen loading" is used to extend endurance. Exercisedepleted glycogen stores can be increased to above-normal levels for about 24 hours by high-carbohydrate refeeding. With elevated levels of tissue glycogen, an individual's endurance ability is increased. Several other phenomena contribute to the pattern of metabolite utilization in highintensity endurance exercise. 1. Although fatty acids are avidly utilized by skeletal muscle during exercise, their plasma concentrations do not differ significantly from those at rest. Their utilization is being balanced by production from adipocyte lipolysis, probably as a result of an elevated catecholamine level. 2. Glucose uptake by skeletal muscle increases with intensity of exercise even though levels of circulating insulin decrease. Either this glucose transport is insulin-independent or exercise enhances insulin sensitivity. 3. The increase in glucose uptake with increased intensity of exercise is matched by hepatic glucose production to the extent that arterial glucose concentrations are elevated by very intense exercise. Hepatic glycogenolysis probably is enhanced by a decrease in the level of insulin and by an increase in levels of glucagon and epinephrine. 4. Exercise may lower insulin levels even though plasma glucose concentrations are increased. This effect may result from ot-adrenergic inhibition of/3-cell secretion.

Low-Level Nonfatiguing Exercise This exercise is characteristic of an individual in normal occupational tasks that could reasonably be continued for 8-12 hours. The principal substrates used are similar to those in long endurance exercise but without depletion

CHAPTER22 Metabolic Homeostasis

of phosphocreatine and with minimal muscle glycogen utilization. The main source of energy is the aerobic oxidation of fatty acids, glucose, and branched-chain amino acids.

Supplemental Readings and References E H. Bennett, S. Haffner, B. Kasiske, et al.: Diabetic renal disease recommendations. American Journal of Kidney Disease 25, 107 (1995). E. E. Calle, M. J. Thun, J. M. Petrelli, et al.: Body-mass index and mortality in a prospective cohort of U.S. adults. New England Journal of Medicine 341, 1097 (1999). G. W. Cline, K. E Petersen, M. Krssak, et al.: Impaired glucose transport as a cause of decreased insulin-stimulated muscle glycogen synthesis in type 2 diabetes. New England Journal of Medicine 341, 240 (1999). S. Dagogo-Jack and J. V. Santiago: Pathophysiology of type 2 diabetes and modes of action of therapeutic interventions. Archives of Internal Medicine 157, 1802 (1997). R. DeFronzo: Pharmacologic therapy for type 2 diabetes mellitus. Annals of Internal Medicine 131,281 (1999). Expert Committee on the Diagnosis and Classification of Diabetes Mellitus: Report of the expert committee on the diagnosis and classification of diabetes mellitus. Diabetes Care 20, 1183 (1997). I. S. Farooqi, S. A. Jebb, G. Langmack, et al.: Effects of recombinant leptin therapy in a child with congenital leptin deficiency. New England Journal of Medicine 341,879 (1999). E L. Ferris III, M. D. Davis, and L. W. Aiello: Treatment of diabetic retinopathy. New England Journal of Medicine 341,667 (1999). J. M. Friedman and J. L. Halaas: Leptin and the regulation of body weight in mammals. Nature 395, 763 (1998). B. Glaser, P. Kesavan, M. Heyman, et al.: Familial hyperinsulinism caused by an activating glucokinase mutation. New England Journal of Medicine 338, 226 (1998). S. B. Heymsfield, A. S. Greenberg, K. Fujioka, et al.: Recombinant leptin for weight loss in obese and lean adults. Journal of American Medical Association 282, 1568 (1997). E Holleman and J. B. L. Hoekstra: Insulin lispro. New England Journal of Medicine 337, 176 (1997). S. J. Hunter and W. T. Garvey: Insulin action and insulin resistance: diseases involving defects in insulin receptors signal transduction, and the glucose transport effector system. American Journal of Medicine 105, 331 (1998). S. L. Kjos and T. A. Buchanan: Gestational diabetes mellitus. New England Journal of Medicine 341, 1749 (1999). A. Must, J. Sadano, E. H. Coakley, et al.: The disease burden associated with overweight and obesity. Journal of American Medical Association 282, 1523 (1999). O. E. Owen, K. J. Smalley, D. A. D'Alessio, et al.: Protein, fat, and carbohydrate requirements during starvation: anaplerosis and cataplerosis American Journal of Clinical Nutrition 68, 12 (1998). A. Rebollo and C. Martinez-A: Ras protein: recent advances and new functions. Blood 94, 2971 (1999). M. Ristow, D. Miller-Wieland, A. Pfeiffer, et al.: Obesity associated with a mutation in a genetic regulator of adipocyte differentiation. New England Journal of Medicine 339, 953 (1998). E. Ritz and S. R. Orth: Nephropathy in patients with type 2 diabetes mellitus. New England Journal of Medicine 341, 1127 (1999). J. N. Roemmich and A. D. Rogol: Role of leptin during childhood growth and development. Endocrinology and Metabolism Clinics of North America 28, 749 (1999).

Supplemental Readings and References M. W. Schwartz and R. J. Seeley: Neuroendocrine responses to starvation and weight loss. New England Journal of Medicine 336, 1802 (1997). P. R. Shepherd and B. B. Kahn: Glucose transporters and insulin action. New England Journal of Medicine 341,248 (1999). C. A. Stanley, Y. K. Lieu, B. Y. L. Hsu, et al.: Hyperinsulinism and hyperammonemia in infants with regulatory mutations of the glutamate dehydrogenase gene. New England Journal of Medicine 338, 1352 (1998).

519 J. A. Tamada, S. Garg, I. Jovanich, et al.: Noninvasive glucose monitoring. Journal of the American Medical Association 282, 1839 (1999). A. H. Xiang, R. K. Peters, E. Trgo, et al.: Multiple metabolic defects during late pregnancy in women at high risk for type 2 diabetes. Diabetes 48, 848 (1999). J. A. Yanovski and S. Z. Yanovski: Recent advances in basic obesity research. Journal of American Medical Association 282, 1504 (1999).

CHAPTER

23

Nucleic Acid Structure and Properties of DNA

The genetic information in all living cells is carried in molecules of deoxyribonucleic acid (DNA), which is primarily found in chromosomes. However, DNA is also present in cellular organelles such as mitochondria and chloroplasts. Viruses carry genetic information in either DNA or RNA (ribonucleic acid) molecules. When RNA viruses infect cells, the genetic information in RNA is converted to DNA prior to the replication and synthesis of new viral particles. In the case of certain RNA viruses (retroviruses) such as human immunodeficiency virus (HIV), the DNA that is copied from the infecting RNA is permanently integrated into the host chromosomes and the viral genome becomes an integral part of the cells' genetic information. The structure of DNA was elucidated by James Watson and Francis Crick in 1953. The structure they proposed made it apparent for the first time how genetic information in chemically stored in cells and how it is replicated and transmitted from one generation to the next. The proposed DNA structure also provided insight into the chemical nature of mutations and how they might occur during replication. Scarcely fifty years has passed since that monumental discovery and now we are deciphering the complete sequence of a human genome. This will lead to a chemical understanding of thousands of genetic disorders and the ability to diagnose, prevent, and treat many inherited diseases and cancers. In this chapter we discuss the structure and properties of DNA. Subsequent chapters examine the functions of DNA

such as replication, mutation, recombination, repair, and the expression of genes.

23.1 Components of Nucleic Acids A nucleic acid is a polynucleotide~a linear polymer of nucleotides that is defined by its three components (Figure 23-1):

1. A nitrogenous heterocyclic base (either a purine or a pyrimidine) attached to the l'-carbon atom of the sugar by an N-glycosidic bond. In DNA the purine bases are adenine (A) and guanine (G) and the pyrimidine bases are cytosine (C) and thymine (T). The bases in RNA are the same except that uracil (U), a pyrimidine, replaces thymine. 2. A cyclic five-carbon sugar which is ribose for RNA and deoxyribose for DNA. The difference in structure between ribose and 2'-deoxyribose is shown in Figure 23-1. The carbon atoms of the sugar are numbered with a prime to distinguish them from the carbon atoms in the base in the same nucleotide structure. 3. A phosphate group attached to the 5'-carbon atom of the sugar by a phosphodiester linkage. This phosphate group is responsible for the strong negative charge of both nucleotides and nucleic acids. 521

CHAPTER23 Structureand Propertiesof DNA

522

Amononuc~[

Adeninej 1 The nucleoside

Ribose

unit

deoxyribose Adenine A

NH~ I

N'~C%.~N:\\e I

I

HC~N~.C-~N/

CH

I OI

OI

OI

o

o

o

o--P-O-P,,

,,

5'

,

"C,xH, H

[ I

O

H/'C,

!

1

I

"1 X

OH

Ribose

Adenosine or deoxyadenosine

(X

or Deoxyribose

H

--

OH)

(X - H)

1

Adenosine monophosphate or deoxyadenosine monophosphate (AMP or dAMP) Adenosine diphosphate or deoxyadenosine diphosphate (ADP or dADP) Adenosine triphosphate or deoxyadenosine triphosphate (ATP or dATP) F I G U R E 23-1 Structure of the nucleotide in DNA and RNA. The base adenine is used as an example. See Table 27-1 for a complete nomenclature of nucleosides and nucleotides.

A base linked to a sugar is called a nucleoside; a base linked to a sugar linked to a phosphate is called a nucleotide or a nucleoside phosphate. The nucleotides in nucleic acids are joined to one another by a second phosphodiester bond that joins the 5'-phosphate of one nucleotide to the 3'-OH group of the adjacent nucleotide. Such a doubly esterified phosphate is called aphosphodiester group (Figure 23-2).

Base Pairing and Base Composition The molar content of the four bases in DNA (called the base composition) always satisfies the equalities [A] = [T] and [G] = [C] where [ ] denotes molar concentration. These equalities arise because of the base pairing rules in

F I G U R E 23-2 Structure of a phosphodiester group. The vertical arrows show the bonds in the phosphodiester group that are free to rotate. The horizontal arrows show the N-glycosidic bond about which the base can rotate freely. A polynucleotide consists of many nucleotides linked together by phosphodiester groups.

SECTION23.1 Componentsof Nucleic Acids H H~c~N\

\ / N--H---O\ \

523

The overall base composition of DNA varies considerably among organisms. Base composition is expressed as the fraction of all bases in DNA that are GC pairs divided by the total number of base pairs, such as ([G] + [C])/[all bases]. This fraction is termed the GC content or percent GC. For human beings and other primates, the value of the GC content is approximately 0.5. For lower organisms the value can vary widely; the most extreme variation is found in bacteria where the GC content varies from 0.27 to 0.76 from one genus to another; E. coli DNA has a GC value of 0.5, which may reflect its close association and evolutionary history with human beings. The higher the GC content of DNA, the more stable is the doublestranded helical molecule. This is because GC base pairs share three hydrogen bonds whereas AT base pairs share only two (Figure 23-3).

/CH 3

I c--c c--c N~C// \\ / \ N---H--N C--H / Deoxyribose \N-- C / \C--N / \H

O/ /

Adenine

\ Deoxyribose

Thymine

H / /O---H-- N\ /H H~c~N\ I c--c c--c /

~C

Deoxyribose

N--H---N C--H \N-- C / \C--N / k

Guanine

// N-- H- --O

H/

k

Deoxyribose

Cytosine

F I G U R E 23-3 The normal base pairs in DNA. Adenine in one polynucleotide chain pairs with thymine in the complementary chain; guanine pairs with cytosine. A-T base pairs are joined by two hydrogen bonds; G-C base pairs are joined by three hydrogen bonds.

DNA which require that A pair with T and G pair with C in the double-stranded DNA molecule (Figure 23-3). Because of the base pairing rules it follows that [purines] = [pyrimidines] in all DNA molecules.

Adenine H

\

/

H

\

/

N

N

I

O

/ N

H

I

N\

H

N"

I

/H

\H

Thymine H ~

N

O

Imino

Keto

I

Amino

H

O

Cytosine H \

Although the bases are chemically quite stable, certain hydrogen atoms bound to the bases are able to undergo tautomerization in which they change their locations on the bases (Figure 23-4). The preferred tautomeric forms of adenine and cytosine are the amino configurations, however, with low probability each can assume the imino configuration. The preferred tautomeric forms of guanine and cytosine are the keto configuration. However, again with low probability, each can assume the enol configuration.

Guanine

H

I

T a u t o m e r i z a t i o n of Bases

/H O'

I

Enol

F I G U R E 23-4 Tautomerization of bases in DNA. The most stable forms of adenine and cytosine are the amino conformations. With low probability these bases can tautomerize into the imino form; if this occurs during replication, an incorrect base pair (a point mutation) may result. The stable forms of guanine and thymine are the keto conformations; the enol conformations also can result in mistakes in base pairing during replication.

524

CHAPTER23 Structure and Properties of DNA

Tautomerization of the bases can occur as the free base and as polynucleotides. If tautomerization of a base should occur at the moment of replication of that region of DNA, an incorrect base may be inserted. For example, the tautomeric form of adenine can pair with cytosine or the tautomeric form of thymine can pair with guanine. During subsequent rounds of DNA replication these mismatched base pairs can result in point mutations in half of the DNA molecules in subsequent cycles of DNA replication and cell divisions. During replication, tautomerization of bases in DNA and ensuing mistakes in base pairing are extremely rare, but experiments show that point mutations do arise in this manner and contribute to genetic variation. However, most mistakes in base pairing that occur during DNA replication are corrected by enzymatic editing or repair functions.

Methylation of Bases After the four bases have been incorporated into DNA, they can be modified by methylation, the addition of methyl groups at various positions (Figure 23-5). The bases that are most frequently methylated are guanine and cytosine. Methylation of cytosine residues influences gene regulation in higher organisms, and about 70% of GC base pairs in mammalian cells are methylated. The pattern of methylation of cytosine residues is inherited and is specific

H

\ / N - - H ""O \\ / CH3 I cc--c N--C//' C~ / \ / N'"H--N \ Deoxyribose \ / /C - - H N--C \ //C N\ H~c~N\

H

O

Deoxyribose

Adenine

H\c~N

I /N~

Deoxyribose

Thymine

\

/ O...H--N // \

C\

\ / N - H... N\

/~-c

//c - c

N ---C

\

/ H Guanine

H H

~C /

C--N

//

N -- H... O

/

-- H

for each species. However, methylation of DNA is not universal; for example, the DNA in the fruitfly Drosophilia is completely unmethylated. Methylation of DNA is primarily the result of two enzymes, Dam methylase and Dem methylase. Dam methylase transfers a methyl group from S-adenosylmethionine to an adenine contained in any GATC sequence in DNA. Dem methylase acts in a similar fashion on cytosine residues in the sequence CCAGG. One or both cytosines in opposite strands of DNA are methylated. Methylated segments of DNA are recognized by proteins that interact with DNA in such processes as replication, recombination, and gene expression. Methylation of DNA also serves an important function in bacteria. Methylation of specific sequences in bacterial DNA protects the bacterial DNA from cleavage by endogenously synthesized restriction endonucleases. Many human genes are methylated differently in maternal and paternal chromosomes at CpG nucleotides, a mechanism that is referred to as imprinting. As a result of genomic imprinting, the expression of genes on maternal and paternal chromosomes differs; the loss of imprinting through mutation or some other mechanism can result in overexpression of critical genes and severe disease. Two inherited diseases that are the result of faulty imprinting are Prader-Willi/Angelmann's syndrome (PWS) and Beckwith-Wiedemann syndrome (BWS). The symptoms of PWS include neonatal hypotonia, hypogonadism, obesity, and short stature; the symptoms of BWS are omphalocele (abdominal wall defect), thickening of long bones, and renal abnormalities. The altered gene in BWS codes for insulin-like growth factor-2 (IGF2) and is located on the short arm of chromosome 15. Usually IGF2 is silent in the maternal chromosome and active in the paternal chromosome. In some cases of BWS, the child receives two copies of chromosome 15 from the father, a condition knows as disomy. These individuals have twice the normal level of IGF2 and suffer from "overgrowth." Low levels of IGF2 may also result from disomy and lead to abnormal "undergrowth." Other inherited diseases and cancers may result from mutations that affect imprinting and normal transmission of chromosomes (Chapter 26).

\

Deoxyribose

Cytosine

F I G U R E 23-5 Methyl groups can be added to bases in DNA. Except for some yeasts and insects, all DNA contains methylated bases. The attachment of methyl groups to bases at specific sequences in DNA acts to regulate the functions of DNA. One function of site-specific methylation in prokaryotes is to protect endogenous bacterial DNA from digestion by restriction endonucleases produced by the bacteria.

23.2 Physical and Chemical Structure of DNA The W a t s o n - C r i c k DNA Structure In DNA, two polydeoxynucleotide strands are coiled about one another in a double-helical structure as originally proposed by the Watson-Crick (W-C) model (Figure 23-6). The important features of the W-C model are as follows:

SECTION 23.2

Physical and Chemical Structure of DNA

525

F I G U R E 23-6 (a) Schematic diagram indicating the dimensions and helical structure of double-stranded DNA. (b) The structure of B-DNA showing the orientation of the atoms and the major and minor grooves in DNA. [From R. E. Dickerson, Sci. Am. December 1983, pp. 100-104; Copyright @1983 by Scientific American, Inc. All rights reserved. Illustration C. Irving Geis.]

1. The sugar-phosphate backbones of the double helix follow helical paths at the outer edge of the molecule. The sugar-phosphate strands are oriented in antiparallel directions such that the 5'-phosphate end of one strand is opposite the 3'-OH end of its partner. As a result of the antiparallel orientation, if one reads a sequence of bases in a 5' to 3' direction on one strand, one is reading the complementary bases on the other strand in a 3' to 5' direction (Figure 23-6a). The overall width of the double helix is 2.0 nm. 2. The two strands in the double helix are complementary because of the base pairing rules that dictate A = T and G = C. Because of the base

pairing rules, the information in DNA is redundant; knowing the sequence of bases in one strand dictates the sequence of bases in the opposite strand. 3. Each base pair in the double helix lies in a plane that is perpendicular to the axis of the helix (Figure 23-6b). Adjacent pairs of bases in DNA are separated by 0.34 nm and rotated with respect to one another so that 10 base pairs occupy each turn of the helix, which repeats every 3.4 nm. 4. Space filling models of DNA reveal two grooves that run the length of the molecule (Figure 23-6b). The major groove is wide and deep and the minor groove is narrow and shallow. These grooves in DNA provide

CHAPTER23 Structure and Properties of DNA

5~fi

space for other strands of nucleic acids and also for binding of regulatory proteins.

Alternative DNA Structures The W-C DNA structure is a right-handed helix as one looks down either axis of the molecule and the helix rotates in a clockwise direction. This conformation of DNA is called B-DNA and is the form found in solution and inside cells. The double helix is stabilized by a number of forces, including hydrophobic interactions and van der Waals forces, which also help stabilize single polynucleotide chains. Collectively, these two forces are known as stacking interactions because of their contribution to the stacked arrangement of the bases in DNA. Other stabilizing forces in DNA are the hydrogen bonds between the AT and GC base pairs. AT base pairs share two hydrogen bonds and GC base pairs share three. These hydrogen bonds are of sufficient strength to discriminate between insertion of a correct and an incorrect base during DNA replication. Although B-DNA is the physiologically significant form of DNA, two other conformations, A-DNA and Z-DNA, have been observed. When water is removed from solutions of DNA (such as in DNA prepared for x-ray analysis), a structure referred to as A-DNA is observed. While A-DNA is still a right-handed helix, the helix is wider and more condensed than in B-DNA. Also, the bases are tilted with respect to the helix axis and the minor groove in the molecule almost disappears. A-DNA is not believed to have any biological significance. A polynucleotide that consists of repeating CGCGCGCGCG base pairs forms a structure in solution that is a left-handed double helix with only one major groove. Also, because the repeating units are dinucleotide pairs, the phosphate groups in the backbone are rearranged to form a zig-zag configuration; this feature accounts for the term Z-DNA. While it is not known what biological role, if any, Z-DNA plays in cells, the fact that long CG tracts exist in DNA in vivo suggests that segments of DNA may assume different structural configurations and that DNA exists in a dynamic rather than a static state in chromosomes. P l a s m i d DNA Circular DNA molecules that are of great significance in nature as well as in numerous biotechnology applications areplasmids, which are widespread among bacteria. Plasmids are small circular DNA molecules consisting of just a few genes to more than a hundred. Among the important genes carried in plasmids are those coding for resistance to

a wide range of antibiotics and genes that allow plasmids to be transferred to other bacteria, even to other species of bacteria. The process of transfer of plasmid and chromosomal DNA from one bacterium to another is called

conjugation. Transfer of antibiotic resistance genes among bacteria in nature has created serious problems in the treatment of many infectious diseases such as tuberculosis, gonorrhea, pneumonia, staphylococcus, and others. The widespread use of antibiotics in agriculture and in hospitals has resulted in the selection and evolution of pathogenic microorganisms that are resistant to many, sometimes all, of the antibiotics normally used to treat infections by these microorganisms. Plasmids also have been genetically engineered in a multitude of ways so that they can carry and express foreign genes in bacteria. For example, the genes coding for human insulin and human growth hormone have been inserted into plasmids which are then reintroduced into bacteria such as E. coli. The genetically engineered bacteria are then used as biological factories to produce the desired drugs. C i r c u l a r a n d Supercoiled DNA Most DNA molecules isolated from prokaryotes and from some virus particles are circular. A circular molecule may be covalently closed, consisting of two unbroken complementary single strands, or nicked, i.e., having one or more interruptions (nicks) in one or both strands. With few exceptions, covalently closed circles assume the form of supercoils (Figure 23-7). The two ends of a linear DNA helix can be brought together and joined in such a way that each strand is continuous. Consider a molecule in which one of the ends is rotated 360 degrees with respect to the other in the unwinding direction, and then the ends are joined. If the hydrogen bonds re-form, the resulting covalent circle will twist in the opposite sense to form a twisted circle, in order to relieve the strain of underwinding. Such a molecule will resemble a figure 8 and will have one crossover point. If it is underwound by 720 degrees before the ends are joined, the resulting superhelical molecule will have two crossover points. In the case of a 720-degree unwinding of the helix, 20 base pairs must be broken (because the linear molecule has 10 base pairs per turn of the helix). However, such a DNA molecule tries to maintain a right-handed (positive) helical structure with 10 base pairs per turn; it will deform itself in such a way that the underwinding is compensated for by negative (left-handed) twisting of the circle. All naturally occurring, superhelical DNA molecules are initially underwound and, hence, form negative

SECTION 23.3

Denaturation of DNA

527 !

1.40

o

T

1.30

q:

Strand separation

,,i.0 m: 3

m 1.20 > I I I I I I I

> .o m (1.)

rr

1.10

-

/

, I t |

iLJ 1.00

...........

1

30

F I G U R E 23-7 Different states of a covalent DNA circle. (a) A nonsupercoiled covalent DNA having 36 turns of the helix. (b) An underwound covalent circle having only 32 turns of the helix. (c) The molecule in (b), but with four superhelical turns to eliminate the underwinding. In solution, (b) and (c) would be in equilibrium; the equilibrium would shift toward (b) with increasing temperature.

superhelices. In bacteria, the underwinding of superhelical DNA is not a result of unwinding before end joining but is introduced into preexisting circles by an enzyme called DNA gyrase, which is one of a class of enzymes called topoisomerases (Chapter 24). In eukaryotes, the underwinding is a result of the structure of chromatin, a DNA-protein complex of which chromosomes are composed. In chromatin, DNA is wound about specific protein molecules, in a direction that introduces underwinding.

23.3 Denaturation of DNA The three-dimensional structures of DNA, RNA, and proteins are determined by weak noncovalent interactions, principally hydrogen bonds and hydrophobic interactions. The free energies of these interactions are not much greater than the energy of thermal motion at room temperature, so that at elevated temperatures the structures of these molecules are disrupted. A macromolecule in a disrupted state is said to be denatured; the ordered state, which is presumably that originally present in nature, is called the native state. A transition from the native to the denatured state is called denaturation. When double-stranded (native) DNA is heated, the bonding forces between the strands are disrupted and the two DNA strands separate; thus, completely denatured DNA is single stranded. Much information about the structure and stabilizing interactions has been obtained by studying denaturation. Some property of DNA that changes as denaturation

1

:

l,

50

!

l

70

11

1

Tm 90

1

l

100

110

Temperature, ~ F I G U R E 23-8 Melting curve of DNA showing the melting temperature (Tm) and possible molecular conformations for various degrees of melting.

proceeds is measured, e.g., the absorption of ultraviolet light. A change in the ultraviolet absorbance (or some other property) as a function of temperature is called a melting curve (Figure 23-8). Many reagents either break hydrogen bonds or weaken hydrophobic interactions, and are powerful denaturants. Thus, denaturation is also studied by varying the concentration of a denaturant at a constant temperature. For DNA, the simplest way to detect denaturation is to monitor the ability of DNA in a solution to absorb ultraviolet light at a wavelength of 260 nm. The absorbance of DNA at 260 nm, A260, is not only proportional to its concentration (as is the case for most light-absorbing molecules) but also depends on the structure of the molecule; the more ordered the structure, the less light is absorbed. Therefore, free nucleotides absorb more light than a single-stranded polymer of DNA (or RNA), which in turn absorbs more light than a double-stranded DNA molecule. For example, solutions of double-stranded DNA, single-stranded DNA, or free bases, each at 50 #g/mL, have the following A260 values: Double-stranded DNA: Single-stranded DNA: Freebases:

1.00 1.37 A260 -- 1.60

A260 - -

A260 - -

If a DNA solution is heated slowly and the A260 is measured at various temperatures, a melting curve such as that shown in Figure 23-8 is obtained. The following features of this curve should be noted: 1. The A260 remains constant up to temperatures well above those encountered by living cells in nature.

528 2. The rise in A260 occurs over a range of 6-8~ 3. The maximum A260 is about 37% higher than the starting value. The hypothetical state of a DNA molecule in different regions of the melting curve is also shown in Figure 23-8. Before the rise begins, the molecule is fully double stranded. In the region of rapid denaturation, base pairs in various segments of the molecule are broken; the number of broken base pairs increases with temperature. A convenient parameter to characterize a melting transition is the temperature at which the rise in A260 is half complete. This temperature is called the melting temperature, Tin. The value of Tm varies both with base composition and experimental conditions. In particular. Tm increases with increasing percent G + C, which is a result of the hydrogen bonds in a GC pair (three) versus an AT pair (two). A higher temperature is required to disrupt a GC pair than an AT pair. Reagents such as urea and formamide, which can hydrogen-bond with the DNA bases, reduce Tm. These denaturing agents maintain the unpaired state at a temperature at which a broken base pair would normally pair again, so that permanent melting of a section of paired bases requires less thermal energy. Other reagents either enhance the interaction of weakly soluble substances (such as the nucleic acid bases) with water or disrupt the water shell; such substances should weaken hydrophobic interactions. An example of the former type of substance is methanol, which increases the solubility of the bases. Sodium trifluoracetate is an example of the second type. The addition of both these reagents greatly reduces Tm because hydrophobic interactions are also important in stabilizing the DNA structure. In fact, the three-dimensional structure of DNA is one that minimizes contact between bases and water and maximizes the contact of the highly soluble phosphate group with water. Minimization of base-water contact is accomplished by stacking of the bases, which occurs even in singlestranded DNA. The bases of double-stranded DNA are more stacked than those in single-stranded DNA because of the hydrogen bonds between the two strands. Both hydrogen bonds and hydrophobic interactions are weak and easily disrupted by thermal motion. Maximum hydrogen bonding is achieved when all bases are oriented in the right direction. Similarly, stacking is enhanced if the bases are unable to tilt or swing out from a stacked array. Clearly, stacked bases are more easily hydrogen-bonded, and correspondingly, hydrogen-bonded bases, which are oriented by the bonding, stack more easily. Thus, the two interactions act cooperatively to form a very stable structure. If one interaction is eliminated, the other is weakened, which explains why Tm drops so markedly following

CHAPTER23 Structure and Properties of DNA

addition of a reagent that destroys either type of interaction. When hydrogen bonds and hydrophobic interactions are eliminated, the helical structure of DNA is disrupted and the molecule loses its rigidity. This collapse of the ordered structure is accompanied by complete disentanglement of the two strands. At high pH, the charge of several groups engaged in hydrogen bonding is changed and base pairing is reduced. At a pH greater than 11.3, all hydrogen bonds are eliminated and DNA is completely denatured. When a DNA solution is heated above 90~ the value of A260 increases by 37% and the solution consists entirely of single strands whose bases are unstacked. If the solution is then rapidly cooled to room temperature and the salt concentration is greater than 0.05 mol/L, the value of A260 drops significantly because random intrastrand hydrogen bonds re-form between distant short tracts of bases whose sequences are complementary (or nearly so). After cooling, about two-thirds of the bases are either hydrogenbonded or in such close proximity that stacking is restored and the molecule is very compact. In contrast, if the salt concentration is 0.01 mol/L or less, the electrostatic repulsion due to unneutralized phosphate groups keeps the single strands sufficiently extended that the bases cannot approach one another. Thus, after cooling, no hydrogen bonds are re-formed. At a sufficiently high DNA concentration and in a high salt solution, interstrand hydrogen bonding competes with the intrastrand bonding just mentioned. This effect can be used to re-form native DNA from denatured DNA.

23.4 Renaturation of DNA If a DNA solution is heated to a temperature at which most (but not all) hydrogen bonds are broken and then cooled slowly to room temperature, A260 drops immediately to the initial, undenatured value and the native structure is restored. Thus, if strand separation is not complete and denaturing conditions are eliminated, the helix rewinds. A related observation is that if two separated strands come in contact and form even a single base pair at the correct position in the molecule, the native DNA molecule will re-form. This phenomenon is called renaturation, or

reannealing. Two requirements are necessary for renaturation to occur. 1. The salt concentration must be high enough that electrostatic repulsion between the phosphates in the two strands is eliminated--usually 0.15-0.50 M NaC1 is used.

SECTION23.4 Renaturation of DNA

529

2. The temperature must be high enough to disrupt the random, intrastrand hydrogen bonds described above. However, if the temperature is too high, stable interstrand base pairing will not occur or be maintained. The optimal temperature for renaturation is 20-25~ below the value of Tm. Renaturation is slow compared with denaturation. The rate-limiting step is not the rewinding of the helix but the precise collision between complementary strands such that base pairs are formed at the correct positions. Since two molecules participate in the rate-limiting step, renaturation is a concentration-dependent process requiring several hours under typical laboratory conditions. In particular, the kinetics of association follows a simple second-order rate law with the association rate increasing with DNA concentration. The kinetics are conveniently described by an equation that relates the fraction, f , of single-stranded (dissociated) DNA and the time elapsed after exposing a DNA sample to renaturing conditions (high salt concentration, elevated temperature): f = 1/(1 + kCot) in which Co is the initial DNA concentration, t is the elapsed time in seconds, and k is a rate constant. A plot of f versus the logarithm of the product Cot yields a sigmoid curve commonly called a Cot ("cot") curve. A set of such curves for several DNA samples is shown in Figure 23-9. A notable feature of these curves is that the renaturation rate is related to the molecular weight of the DNA. A useful index for characterizing these curves is Co/tl/2, with the value of Cot corresponding to renaturation of half of the DNA ( f = 1/2). Comparison of the Co/tl/2 values for E. coli DNA (M.W. = 2.7 • 109) and T4 DNA (M.W. = 1.1 • 108) shows that T4 DNA renatures roughly 50 times faster than E. coli DNA. The reason

FIGURE 23-9 Reassociation "Cot" curves for bacteriophage T4 DNA, E. coli DNA, and mouse DNA. [Reproduced with permission from D. M. Prescott: Cells: Principles of Molecular Structure and Function. Jones and Bartlett, London, 1988.]

F I G U R E 23-10 (a) Hypothetical DNA molecule having a base sequence that is 3% of the total length of the DNA and is repeated five times. The dashed lines represent the nonrepetitive sequences: they account for 85% of the total length. (b) Renaturation curve for the DNA in (a). Time is logarithmic to keep the curve on the page.

is that if the two DNA samples have equal molar concentrations of nucleotides, the T4 sample will contain many more DNA molecules than the E. coli sample. Cot analysis is not-usually done with intact DNA molecules but rather with fragments having lower molecular weights. This breakage does not affect the relative values of Co/tl/2 since the number of different kinds of fragments of T4 DNA is smaller and hence their concentration larger than the corresponding number of fragments in the E. coli sample. Studies of a variety of prokaryotic DNAs show that the value of Co/tl/2 is directly related to the total size of the DNA of the organism (the genome size). However, this relation does not apply to the DNA of eukaryotes because of the presence of highly repetitive sequences. The fragmentation of the DNA molecules allows a new feature of base sequences to be seen. Figure 23-10(a) shows a long hypothetical DNA molecule having a repeating base sequence. If the unbroken denatured DNA were allowed to renature, the Cot curve would be a smooth curve in which Co/tl/2 would be proportional to the size of the DNA. However, if the DNA were broken into small pieces, several fragments from each molecule would contain the repeated sequence, and these fragments would renature more rapidly than the bulk of the sequences. For example, if the repeated sequence contains 3% of the total number of bases of the DNA and there are five copies of the repeated sequence in the DNA, the Cot curve would be that shown in Figure 23-10b, in which the more rapidly renaturing component accounts for 15% of the transition; hence, the conclusion that 15% of the DNA (on a weight basis) contains a repeating sequence. Approximately 30% of human DNA contains sequences of bases that are repeated 20 times or more.

530

23.5 Repetitive DNA Sequences Cot curves from various eukaryotic organisms show the presence of repetitive sequences to varying degrees. Unique sequences represent genes, approximately 100,000 in the human genome, but constitute only 1-2% of the total genetic information in the human genome. Some genes may be slightly repetitive if they are related to pseudogenes, sequences of DNA that resemble genes but that are no longer expressed. About one-third of the human genome consists of sequences of bases that are repeated at least 20 times or more. Moderately repetitive refers to sequences that usually contain between 10 and several hundred copies; highly repetitive refers to sequences that are repeated thousands or millions of times throughout the genome. The distinction among various classes of repetitive sequences is frequently blurred and additional categories are sometimes defined. Repetitive sequences are also defined by the number of base pairs in each repeated segment. Sequences that have from 100 to 500 bp are referred to as SINES (short interspersed repeated sequences) and sequences that have several thousand base pairs are referred to as LINES (long interspersed repeated sequences.) Thus, repetitive DNA sequences can be described both by the length of the segment and the degree to which it is repeated. Some examples of moderately repetitive sequences are transposable elements including retroviruses, histone genes that are repeated 30-40 times in the human genome, and genes coding for ribosomal RNAs and transfer RNAs. Most of the moderately repetitive genes do not code for proteins but serve other functions in the cell. Histones are involved in condensation of DNA in chromosomes and both ribosomal RNAs and transfer RNA are involved in protein synthesis. One special class of a highly repetitive sequence is satellite DNA which consists of 6-8 bp (such as ATAAACT) and may account for 10-20% of the total DNA. The term satellite DNA derives from the observation that if fragmented DNA is centrifuged in a density gradient solution that separates DNA by weight, a small band of DNA is observed that is separate from the bulk of the DNA. The base composition of satellite DNA is AT-rich, which explains why it separates from the rest of the DNA. Satellite DNA is thought to play a structural role in chromosomes. Certain satellite DNA sequences are concentrated near the centromeres of chromosomes, the site where spindle fibers attach when sister chromatids are separated. Other satellite DNA sequences are located in

CHAPTER 23

Structure and Properties of DNA

telomeres, structures at the ends of chromosomes that play a critical role in DNA replication (Chapter 24). Another class of highly repetitive sequences is the Alu family, which consist of about 300 bp that are repeated millions of times throughout the genome. Any Alu sequence is at least 85% homologous in base sequence to any other Alu sequence; hence, the family of genes has been highly conserved. Each Alu sequence contains a restriction site that is recognized by the AluI restriction enzyme, from which the name of the family of sequences is derived.

23.6 Degradation of DNA Nucleases A variety of enzymes break phosphodiester bonds in nucleic acids; deoxyribonucleases (DNases) cleave DNA and ribonucleases (RNases) cleave RNA. DNases usually are specific for single- or double-stranded DNA although some DNases can cleave both. DNases can act as exonucleases in which they remove one nucleotide at a time from either the 3' or 5' end of the strand. Other DNases function as endonucleases and are specific for cleaving between particular pairs of bases.

Restriction E n z y m e s An important class of DNA endonucleases are restriction enzymes (restriction endonucleases) that recognize specific sequences of bases in DNA (a restriction site) and make two cuts, one in each strand that generates fragments of double-stranded DNA. Microorganisms use their restriction enzymes to degrade any foreign DNA that may enter the cell in the form of viruses, plasmids, or naked DNA. Hundreds of different restriction enzymes have been isolated from microorganisms; these enzymes generally recognize sequences of four, six, eight, or rarely more bases that have an axis of symmetry (Table 23-1). Restriction sites that have an axis of symmetry are called palindromes, which means that the restriction site can be rotated 180 degrees and the sequence of bases will remain the same. Restriction enzymes can cut DNA in either of two ways. Each strand can be cleaved along the axis of symmetry generating fragments of double-stranded DNA that have blunt (flush) ends. Symmetrical cuts that are staggered around the axis of symmetry generate fragments of doublestranded DNA that have single-stranded cohesive ends (Figure 23-11). Fragments of DNA with cohesive ends

SECTION23.6 Degradation of DNA

531

TABLE 23-1 Some Restriction Endonucleases and Their Cleavage Sites* Name of Enzyme

Microorganism

Target Sequence and Cleavage Sites Axis of symmetry

Generates cohesive ends

Escherichia coli RY 13

EcoRI

Bacillus amyloliquefaciens H

BamHI

Bacillus globigii

BgIII

Haemophilus aegyptius

HaeII

Haemophilus influenzae R d

HindIII

Providencia stuartii

PstI

Streptomyces albus G

SaII

Xanthomonas badrii

XbaI

Thermus aquaticus

TaqI

Nocardia otitdis

Not I

Streptomyces sp.

Sgf I

G C G C

GSA A C T T GZG A C C T AZG A T C T Pu G C PyTC G AsA G T T C C T G GTA C GZT C CzA G T C T A GzA T C A G CSG G G C C C G A G CTT

T C ATG C C GTG C T G?A C $ Py C G Pu C T T G ATA C A$G G T C G A C C TTG A G A T CTT G A CTT C C G G GTC T$C G A G C

T

G

G

C

C

A

A

C

C ~ G

G

T

C

C ~ G

G

C

C

C ~, G

G

G

G

G

G ~ C

C

C

T A T A T A G

C G C G

Generates flush ends

Brevibacterium albidum

Haemophilus aegyptius Serratia marcescens

BalI

HaeI

SmaI

| | | |

*The vertical dashed line indicates the axis of dyad symmetryin each sequence. Arrows indicate the sites of cutting. The enzyme TaqI yields cohesive ends consisting of two nucleotides, whereas the cohesive ends produced by the other enzymes contain four nucleotides. The enzyme HaeI recognizes the sequence GGCC whether the adjacent pair is A .T or T .A, as long as dyad symmetryis retained. Pu and Py refer to any purine and nvrimidin~ rP.~n~tivolv x.,

l

.,

are useful for constructing novel DNA molecules, which is the goal of recombinant DNA (rDNA) technology. Since a restriction enzyme recognizes a unique sequence, the number of cuts and the number of DNA fragments depends on the size of the molecule. In general, restriction sites consisting of four bases will occur more

frequently by chance than sites consisting of six or eight bases. Thus, four-cutters will generate more fragments than six-cutters which, in turn, will generate more fragments than eight-cutters. A particular restriction enzyme generates a unique family of fragments for a particular DNA molecule.

532

CHAPTER 23

(a) Cuts on axis of symmetry

........ ........

i

TCIGA AGjCT

........

I S e p a r a t i o n of fragments

........ ........

TC AG

3 I i i 5

5" GA ........ CT ........ 3"

Flush-end molecules

(b) Cuts symmetrically placed around axis of symmetry

........ ........

I

GAAITTC CTTIAAG

t I Separation of fragments

..... ........

G

3"

........

CTTAA

5" AATTC

........

Structure and Properties of DNA

has a strong interaction with (ideally) a specific DNA target and can be detected after the interaction. DNA probes consisting of 20 bases or fewer usually will have a unique target even in a large set of DNA molecules. The probability that any base will follow any other base in DNA is one in four or 0.25. Therefore, the probability of a specific sequence of 20 bases occurring in a DNA molecule by chance is 0.252~ a vanishingly small number. The use of DNA probes in various aspects of medical diagnostics is increasing rapidly. DNA probes can be used to identify infectious agents if sequences specific to different pathogens are known. Identification of a pathogen by DNA probes can be done in hours as compared to days or weeks by conventional culturing of microorganisms. DNA probes are now used routinely to detect the presence of mutant alleles in fetal cells obtained by amniocentesis, as well as in cells removed from affected adults or carriers. Many inherited disorders, such as sickle cell disease, cystic fibrosis, Huntington's disease, Duchenne's muscular dystrophy, and dozens of other Mendelian (single-gene) disorders, can now be diagnosed in fetuses and adults. In addition to inherited disorders, DNA probes are used to detect the presence of active oncogenes or inactive tumor suppressor genes in cancerous tissues removed from patients (Chapter 26).

G ........ 5"

Cohesive-end

3 molecules

F I G U R E 23-11 Two types of cuts made by restriction enzymes. The arrows indicate the cleavage sites. The dashed line is the axis of symmetry of the sequence.

Figure 23-12a shows the positions of restriction sites in bacteriophage lambda (k) DNA for the restriction enzymes EcoRI and BamHI. This arrangement of restriction sites is known as a restriction map. The family of fragments generated by one or more restriction enzymes is detected by agarose gel electrophoresis that separates DNA molecules according to their molecular weights (Figure 23-12b).

23.7 Diagnostic and Clinical Applications of DNA DNA Probes The ability to isolate specific fragments of DNA containing known sequences of genes gives rise to DNA probes that can be used for a variety of diagnostic, forensic, and therapeutic purposes. DNA probes can be labeled with either radioactive or nonradioactive markers. A DNA probe

Southern Blot Analysis The Southern blot (named for its inventor, E. M. Southern) is a method for hybridizing one or more labeled DNA probes to a large number of DNA fragments and discriminating among them. The procedure depends on the ability of denatured DNA single strands to bind tightly to nitrocellulose under certain conditions. The Southern blot procedure is outlined schematically in Figure 23-13. The DNA to be investigated is digested with several restriction enzymes to generate DNA fragments of varying sizes which are then separated by agarose gel electrophoresis. After the fragments are separated, the gel is immersed in a denaturing solution that converts all DNA to single strands. Then the gel, which is in the form of a broad, flat slab, is placed on top of filter paper supported by a glass plate. A nitrocellulose filter is placed over the gel and covered with another sheet of filter paper. Finally, paper towels and weights are placed on top of the gel. The glass plate supporting the filter papers and gel is adjusted so that the ends of the filter paper are suspended in buffer and act as wicks. The buffer moves upward through the gel and into the wad of paper towels that act to absorb the buffer. The DNA migrates from the gel to the nitrocellulose filter that binds the single-stranded DNA fragments.

Diagnostic and Clinical Applications of DNA

SECTION 23.7

533 EcoRI

EcoRI 0

43.9

53.5

64.9

79.9

91.8

t

t

I

t

I

t

43.9

9.6

11.4

15.0

11.9

BamHI

100 8.2

o

BamHI 0

10.6

45.3

56.5

69.3

83.8

F

i

t

I

I

I

10.6

34.7

11.2

12.8

14.5

100 16.2

4

(a)

5/6

(b)

F I G U R E 23-12 (a) Restriction maps of ~. DNA for EcoRI and BamHI nucleases. The vertical bars indicate the sites of cutting. Thc top numbers indicate the percentage of the total length of DNA measured from the gene A end of the molecule. The bottom numbers are the lengths of each fragment, again expressed as percentage of total length. (b) Gel electrophoretogram of EcoRI and BamHI restriction enzyme digests of ~. DNA. The bands labeled "cohered ends" contain molecules consisting of two terminal fragments, joined by the normal cohesive ends of ~. DNA. Numbers indicate fragments in order from largest 1 to smallest 6. Bands 5 and 6 of the BamHI digest are not resolved.

~ ~_1,.,. ~ - _ ~ . ~

,,.~_~#~-:

DNA restriction fragments

Agarose gel electrophoresis

,ii H I tt/ t . I /III

[/////I

/I I I/ I #I I II m~ I I I I I/1I ~ - -

[

Ill/ I l l

"

~

..........

/ Film / I

Fragments separated by size

I

.Weight Transfer of ~ towels DNA fragments litrocellulose filter from gels to nitrocellulose ~el filter

1

# , . , .,,... ] /////# ///// / # t /i/~/t / / t t t .

////,1''1/'1/

[

Nitrocellulose filter with DNA fragments

Radioactive D N A probe hybridized to fragments on filter

. Autoradiograph showing -positions of hybrid DNA

F I G U R E 23-13 Procedure for a Southern blot. This analysis allows detection of a specific fragment of DNA by hybridization of a radioactive probe with a complementary sequence.

The nitrocellulose filter is dried and exposed to high temperature to keep the DNA in a denatured state. A labeled DNA probe containing a known sequence or gene is then incubated with the nitrocellulose filter under a variety of hybridization conditions to identify a particular fragment of DNA from the sample. Strands of the DNA probe bind to fragments of the separated DNA that contain similar or identical sequences and hybridize together to form double-stranded DNA. After the hybridization reaction is complete, the solution is cooled and the itrocellulose filter is washed to remove unbound probe DNA and dried. The labeled DNA fragments can be visualized by autoradiography (if the probe was radioactive) or by fluorescence. The DNA fragments of interest also can be eluted from the nitrocellulose filter for further analysis.

Polymorphisms Approximately 25% of all human genes occur among individuals in a population in multiple allelic forms referred to as polymorphisms. For example, the ABO locus, an important factor in blood transfusions, consists of three different alleles (alternative states of a gene) called A, B, and O. Individuals can be homozygous or heterozygous for any pair of A, B, and O alleles. The HLA family

534 of genes that play a vital role in the rejection of tissue transplants consists of hundreds of different alleles (Chapter 35). No two individuals, except for identical twins, share exactly the same set of HLA alleles. For a gene to be polymorphic, alternative alleles must be present in at least 1% of individuals in a population. The relative frequencies of the different alleles at a polymorphic locus usually vary from one human population to another. The remaining 75% of human genes consist of a single allelic form and are said to be monomorphic. For example, all human beings have the same ot and/~ genes for hemoglobin, presumably because the protein structure has been optimized by millions of years of human evolution. All persons, irrespective of race or ethnicity, have hemoglobin genes that are identical in sequence. The only exceptions to the monomorphic state of the hemoglobin genes are the rare individuals who inherit or acquire a mutation in the ot or/3 genes that gives rise to the several hundred characterized human hemoglobinopathies (Chapter 28). Restriction enzyme sites in DNA also are highly polymorphic in human populations. The pattern of DNA fragments produced by digesting DNA from different individuals will usually show a difference if a sufficient number of fragment sizes are examined; again, only identical twins have identical restriction enzyme sites. The different fragments produced by digesting DNA with one or more restriction enzymes are called restrictionfragment length

polymorphisms (RFLPs). RFLPs have been extremely useful in constructing a restriction map of the human genome (which can be correlated with the genetic and physical maps), in screening human populations for the presence of mutant alleles, and for diagnosing hereditary disorders in fetuses and prospective parents. An early success in mapping the human genome was determining the location of more than 10,000 RFLPs on the 23 human chromosomes. Most mutant genes that cause hereditary disorders are linked to specific RFLP differences among affected and unaffected persons that can be detected by using DNA probes carrying the gene in question. For example, the single-base change found in all cases of sickle cell anemia also changes a restriction site located within the/3-globin gene that is recognized by the restriction enzyme MstII (Figure 23-14). When DNA from unaffected and affected individuals is digested with MstII and a Southern blot analysis is performed with the appropriate probe, affected individuals show a different pattern of DNA fragments as compared with unaffected individuals. Huntington's disease (HD) is an autosomal dominant disorder characterized by progressive chorea, dementia,

CHAPTER 23

Structure and Properties of DNA

FIGURE 23-14 Detection of sickle cell anemiaby RFLPanalysisand Southern blot.

and ultimately death. It is a late-onset hereditary disease that usually manifests after age 40. Any son or daughter who had a parent with Huntington's disease has a 50% probability of inheriting the mutant allele from the affected parent. A HindIII restriction site is closely linked to the HD gene in asymptomatic family members who carry the mutant allele but is absent in family members who have not inherited the mutant allele. Southern blot analysis can determine which family members are destined to die of the disease later in life. (Not all families that carry a HD gene mutation have this particular mutation that was characteristic of the original large family investigated.) Genetic testing for susceptibility to Huntington's disease (and many others) raises profound ethical questions for society, for families, and for individuals who may or may not have inherited disease-causing mutant alleles, and who may or may not want to know their disease status. Persons diagnosed with a gene for an inherited disorder may face discrimination in obtaining health insurance, life insurance, job opportunities, and so path. Hundreds of Mendelian (single-gene) disorders can now be detected with appropriate DNA probes (Table 23-2). However, genetic screening generally is not advised, particularly if no medical treatment is available for the particular disease.

SECTION 23.7

Diagnostic and Clinical Applications of DNA

535

TABLE 23-2

A Partial List of Diseases for Which Single Genes Have Been Isolated and Characterized* Familial amyloid polyneuropathies Phenylketonuria Familial hypercholesterolemia Christmas factor deficiency Retinoblastoma, Wilson's disease Type IV collagen deficiencies Hydatidiform moles and choriocarcinomas Tay-Sachs disease Alzheimer' s disease Gaucher' s disease Charcot-Marie-Tooth 1A disease Williams syndrome Von Hippel-Lindau disease Breast cancer (BRCA1) Prostate cancer

Niemann-Pick disease X-linked Charcot-Marie-Tooth disease Familial colon cancer Hereditary retinal degenerative diseases Ototoxic deafness XSCID Compulsive disorders Myotonic muscular dystrophy Familial dysalbuminemic hyperthuroxinemia Long QT syndrome Cerebral autosomal dominant arteriopathy Cancer-associated mar binding protein Breast or ovarian cancer Idiopathic dilated cardiomyopathy Hereditary neuropathy with liability

*Each of these mutant alleles contributes to a greater or lesser degree to the pathology of the disease or inherited disorder. As of December 1997, patents had been issued to universities or companies for the commercial use of these genes.

Forensic DNA Analysis RFLPs are also widely used in forensic pathology, criminology, and cases of contested paternity. A particular set of DNA probes is specific for hypervariable sequences in the human genome that are inherited in a Mendelian pattern identical to the inheritance of genes. Hypervariable sequences are highly polymorphic minisatellite loci that are unique to each individual just as each individual has a unique set of genes. These hypervariable sequences can be detected by a special set of DNA probes called Jeffreys probe after their discoverer. A Southern blot analysis of fragments of DNA from an individual using the Jeffreys probe constitutes a unique "genetic fingerprint." Only monozygous twins have the same pattern of hypervariable sequences and identical genetic fingerprints (Figure 23-15). DNA obtained from a blood stain, a human hair cell, or even a few sperm provides enough material to match with the DNA obtained from a suspect. DNA testing of suspects in rape, murder, and other crimes in which a sample of DNA was obtained at the scene of the crime is now a routine procedure. DNA sequences can be amplified by the polymerase chain reaction (PCR) technique. Only an infinitesimal amount of DNA is needed, e.g., a sample of DNA from a single hair follicle or sperm. The DNA to be amplified is mixed with three other components in an automated PCR procedure:

1. A heat-stable DNA polymerase that is isolated from a thermophilic bacterium, 2. An excess of two short primer DNAs that are complementary to opposite strands of the DNA fragment that is to be amplified, and 3. An excess of deoxyribonucleotide triphosphates. PCR consists of repeated cycling of three reactions: denaturation of the DNA by heating, reannealing of the primers with the target DNA by cooling, and synthesis of new DNA strands. The sequence of reactions is automatically repeated at defined intervals to yield an exponential increase in the amount of DNA. Twenty cycles of PCR amplify DNA by about a factor of 106 and 30 cycles by about 109. PCR amplification of DNA is one of the most widely used techniques in medical research and diagnostics. PCR is used in forensic pathology (to identify human remains), in rapid identification of infectious microorganisms, in diagnosis of inherited diseases, and in archaeology and anthropology where small DNA samples can be recovered.

Sequencing DNA Until the development of automated DNA sequencing machines in the 1990s, two techniques were used to sequence the bases in a segment of DNA. Each of the techniques involves the isolation of a restriction fragment containing a few hundred or a few thousand base pairs. The DNA is denatured and each strand is sequenced separately so

536

CHAPTER 23

Structure and Properties of DNA

radioactivity and used in four separate reactions in vitro in which a single-stranded DNA template is copied. A series of radioactive DNA fragments are separated according to length by agarose gel electrophoresis (fragments differing by only one nucleotide are separated), and the DNA sequence can be read directly from the pattern of radioactive bands in the gel. Another sequencing technique is the Maxam-Gilbert method (developed by Alan Maxam and Walter Gilbert). In this method, a single DNA strand is labeled at the 5' end with radioactive phosphorus (p32). The radioactive DNA is divided into four portions and each one is exposed to different chemical reactions. Each reaction causes a 5' cleavage adjacent to either 1. 2. 3. 4.

AorT, G alone, C or T, or C alone.

The reactions are carried out for a short time so that, on average, only one cleavage occurs in each DNA molecule. This produces a set of DNA fragments (one set for each reaction), whose length identifies that position of a particular base. For example, a fragment containing 19 nucleotides in the G-only reaction mixture identifies G at position 20 from the 5' end. Similarly, a fragment containing 27 nucleotides present in the C or T reaction but not in the C-only reaction indicates that T is at position 28. The lengths of DNA fragments are determined by polyacrylamide gel electrophoresis, a technique that can separate DNA fragments differing in length by only one nucleotide.

Gene Therapy

F I ( ; U R E 23-15 Genetic fingerprint of monozygotic twins (columns 2 and 3) and their parents (columns 1 and 4). The arrows indicate hypervariable sequences inherited from the father. Similar hypervariable sequences can be identified from the mother. [Reproduced with permission from A. J. Jeffreys, V. Wilson, and S. L. Thein: Individual-specific "fingerprints" of human DNA. Nature 316, 76 (1985). @ 1985 by Macmillan Magazines Ltd.]

that the sequences can be compared in order to eliminate errors. One sequencing technique is the Sanger method (developed by Fred Sanger) which uses dideoxynucleotides that stop chain elongation at the site of their incorporation. The four dideoxynucleotide substrates are labeled with

One expected advance in medical treatment is the use of gene therapy to ameliorate or cure a variety of inherited disorders as well as diseases caused by somatic mutations such as cancer. The goal of gene therapy is the replacement of a defective, disease-causing gene in an individual by normal, functioning copies. In essence, gene therapy is a novel form of drug therapy; it uses the biochemical capacity of a patient's cells to synthesize the therapeutic agent from the introduced gene. The first attempts at gene therapy were directed at trying to correct severe combined immunodeficiency (SCID) which is caused, in some cases, by a deficiency of adenosine deaminase (ADA), that is expressed in all tissues. ADA deaminates both adenosine and deoxyadenosine and, in the absence of ADA, deoxyadenosine accumulates in cells. Deoxyadenosine can be phosphorylated by the enzyme deoxcytidine kinase to produce deoxyadenosine

531

SECTION23.8 DNA Vaccines

triphosphate, a toxic substance that kills dividing cells. As a result, individuals with an ADA deficiency are defective in both humoral and cell-mediated immunity. In the mid-1980s, the first clinical gene therapy trials were attempted to correct ADA deficiency in two children with SCID. The ADA gene was cloned into a viral vector and inserted into peripheral T cells removed from the affected children. After a period of growth of the genetically modified cells in vitro, they were reintroduced into the patients. Synthesis of ADA could be detected for a time but the activity disappeared as the introduced cells died. Several other trials of gene therapy for ADA deficiency showed promise, but it was still necessary to maintain the children with SCID on enzyme replacement therapy. Finally, in 2000, successful treatment of an X-linked form of SCID (SCID-X1) by gene therapy was reported by a French medical team. Almost a year after a normal gene was introduced into their cells, two children were still synthesizing the enzyme that they lacked. This was the first success for gene therapy in curing a disease after years of effort. Since the 1980s, hundreds of clinical trials of gene therapy for cystic fibrosis, osteogenesis imperfecta, Gaucher's disease, Fanconi's anemia, and several forms of cancer have been attempted. The problems that need to be overcome in developing a successful strategy for gene therapy are: 1. Development of safe and effective vectors for cloning the gene and inserting it into palients cells, 2. Regulation of expression of the desired gene product so that it is produced at the right time and in the correct amounts in the appropriate tissues, and 3. Stability of the inserted genetic construct in cells so that the product will continue to be produced. Many different viral vectors have been developed to deliver genes to various organs in the body; these include retroviruses that can insert themselves and the genes they carry into the chromosomes of cells; adenovirus, a DNA virus that causes respiratory infections, which has been inactivated by removal of many viral genes; and lentiviruses, slow-growing retroviruses. All of the viral vectors carry with them some risks. In 1999, a gene therapy patient died of complications from the use of adenovirus that was being tested as a therapy for an inherited liver disease caused by a deficiency in the enzyme ornithine transcarbamylase (OTC). Despite the problems that have beset gene therapy trials, it is expected that development of safer vectors and new techniques for delivering genes to cells will eventually make gene therapy a vital part of medical treatment.

23.8 DNA Vaccines An extension of gene therapy, and one that may turn out to be of worldwide importance, is the use of naked DNA as a vaccine to prevent viral diseases. For example, plasmid DNA can be injected into tissues; upon entry, the DNA expresses any cloned gene, such as a viral antigen, that is carried by the plasmid. DNA vaccines have the advantage that the viral protein that is expressed in the cells stimulates both humoral and cell-mediated immunity. Fragments of the synthesized viral protein are carried to the cells' surface where they stimulate CD8 + cytotoxic T cells and, thereby, cell mediated-immunity (Figure 23-16). In experiments with mice, DNA vaccines have proven to be very effective. The gene coding for the core protein of the influenza virus was cloned into a plasmid vector and injected into mice. The mice developed immunity not only to the strain of influenza from which the gene was derived but from other strains of influenza virus as well. Inducing an immune response with a viral core antigen is thought to be more effective than capsid antigens because viral core proteins from related viral strains do not differ much in structure or antigenicity. Viral capsid proteins, on the other hand, evolve rapidly; such changes, for example, account for the different strains of influenza virus that arise each year. Before DNA vaccines can replace conventional vaccines that use inactivated or attenuated viruses, the safety of the plasmid vectors must be rigorously proved. The plasmids might occasionally integrate into the host genome or they might stimulate an immune response to tissues containing the plasmid DNA. Either event might dictate against the widespread use of DNA vaccines.

Antibodies to DNA Both single- and double-stranded DNA are antigenic; antibodies to DNA are normally found in the circulation, but in some individuals overproduction of DNA antibodies causes disease. In particular, patients with systemic lupus erythematosus (SLE) show abnormal levels of antibodies to double-stranded DNA. Antibodies to singlestranded DNA also bind to bases, nucleosides, nucleotides, oligonucleotides, and the ribose-phosphate backbone of RNA. Antibodies to double-stranded DNA also bind to base pairs, chromatin, nucleosomes, type IV collagen, and the deoxyribose-phosphate backbone of DNA. Antibodies to DNA consist of both IgM and IgG classes. Healthy individuals usually have low-affinity IgM antibodies to DNA; however, if these undergo an isotype switch to IgG, they may become pathogenic. Tests for DNA antibodies help establish a diagnosis of SLE,

538

CHAPTER 23

Structure and Properties of DNA

!"1(;1 Ri'~ 2 3 - 1 6 How a DNA vaccine might work. A viral gene coding for a core or capsid protein is inserted into a plasmid. The plasmids are injected into cells where the plasmid migrates to the nucleus. Viral proteins are expressed in the cells producing both a cell-mediated and humoral immune response. [Reproduced with permission from W. M. McDonnel and E K. Ashari, DNA vaccines, N. Engl. J. Med. 314, 44 ( 1996)].

although the level of such antibodies is not always predictive of the severity of the disease. Genetic susceptibility also seems to play a role in the pathogenicity of DNA antibodies in some individuals. Antibodies to double-stranded DNA may cause glomerulonephritis by forming antibody-DNA complexes that become trapped in the glomeruli. In some cases, DNA antibodies can be recovered from damaged tissues in patients with SLE or glomerulonephritis, indicating that the antibodies are involved in tissue damage. The production of DNA antibodies is reduced by immunosuppressive drugs, and immunosuppressive therapy is the accepted treatment for SLE and related diseases caused by antibodies directed against DNA.

23.9 The Human Genome Project A complete human genome, the total genetic information in a haploid set of chromosomes, contains approximately 3.1 billion bases and 50,000-1000,000 genes that encode proteins. The H u m a n Genorne Project was established

with federal funds to obtain the complete sequence of a human genome or, more accurately, the sequence of pooled DNA extracted from cells donated by several anonymous individuals. Initially a public research effort, the sequencing task later was undertaken by a private company, Celera Genomics (Rockville, MD). The ensuing competition led to a joint announcement by the U.S. Government and Celera Genomics on June 26, 2000 that the human genome had essentially been sequenced although there were still gaps and errors in the sequence. The two groups agreed to continue cooperating to complete the sequence of a generic human genome as soon as possible. Any two human genomes are approximately 99.9% identical in sequence. Of course, the 0.1% represents a difference of 3 million bases between any two individuals, and these base differences may have profound effects on disease susceptibility, behavior, intelligence, personality, and other traits. Approximately 75% of all human genes have the same DNA sequence in all individuals except for those with rare mutations. The type of genetic markers employed in the dissection of the human genome include

SECTION 23.9

The Human 6enome Project

1. Restriction sites: sequences of DNA digested by restriction enzymes;

2. Variable-number tandem repeats (VNTRs): sequences of bases that are repeated in tandem almost without change up to 40 copies; 3. Simple sequence repeats (SSRs): sequences of bases, most commonly CA, that are repeated dozens of times and occur at thousands of sites in the genome; and 4. Single-nucleotide polymorphisins (SNPs): sites in human DNA that differ by a single base pair. Because SSRs and SNPs occur so frequently in the genome, they can be used to distinguish one chromosome from another in an individual. An SSR may occur 19 times at one location in a particular chromosome and 21 times in a homologous chromosome. Since an SSR is inherited like a gene, the variability can be followed from person to person in a pedigree. The length of a particular SSR can be determined by gel electrophoresis without any knowledge of the exact sequence. SNP differences occur about once in every thousand bases in DNA. That is, at a position on a chromosome where an A - T base pair occurs, a G-C pair might occur on the homologous chromosome. Because only a small percentage of the total number of nucleotides in the human genome is actually used to encode information for the synthesis of different proteins, techniques have been developed to search for and map genes that are expressed. Knowing the location of these protein-encoding genes is extremely useful as mutated forms of these expressed genes are likely implicated in many diseases. Two techniques have contributed to the identification of many active genes in the human genome. Expressed sequence tags (ESTs) for as many as 50,000 human genes have been generated by isolating messenger RNAs and copying them into DNA. These DNA probes can then be used to identify the location of the expressed genes on the various chromosomes by hybridization. Specific chromosomal sites also can be identified by using PCR primers to amplify a small segment of a chromosomal locus. The sites identified by using PCR amplification are called sequence

tagged sites (STSs). Completing the sequencing of the human genome is only the first step in the revolution in medicine that many are predicting will result in the 21st century. However, knowing the base sequence, chromosomal position, and even the function of the protein that is encoded in the gene is only the first step in using the information to benefit patients with inherited defects (Figure 23-17). The sequence information does not specify how and when a gene is turned or or off, in what cells the gene is active or inactive,

539 or in what stage of development expression of the gene is needed. The expression of most genes is modulated in complex ways and coordinated by numerous intracellular and extracellular factors. How a particular protein affects the biochemistry, physiology, and phenotype of an individual must be understood before therapeutic messures can be developed. Often inherited defects are manifested during fetal development and the damage is irreversible by the time the child is born. Some of the anticipated medical, social, and ethical consequences deriving from the human genome project are described below. Genetic testing: Hundreds of genetic tests are available that can be used to determine if a person carries a defective gene and to estimate the risks of developing a disease as well as the risk of passing the gene on to progeny. In many instances, determining the risk of passing on genes for Hungtington's disease, cystic fibrosis, hemophilia, Duchenne's muscular dystrophy, or sickle cell anemia provides prospective parents with information useful in making reproductive choices. However, an asymptomatic person with a family history of Huntington's disease faces a difficult choice. Finding out while young that you carry a gene that eventually will produce a lethal, neurological disease creates enormous emotional and psychological stress since there is no treatment or cure that can be offered. Faced with the choice of genetic testing for Huntington's disease, many possible carries of the gene opt not to be tested. Genetic tests also can determine if a person carries genes that increase the chances of developing breast cancer, colon cancer, and other polygenic-multifactorial diseases. Again, in most cases, only marginally helpful medical help can be offered to individuals who test positive for cancer-causing susceptibility genes. Some young women who are homozygous for the BRCA1 gene have elected to undergo prophylactic bilateral mastectomy to reduce the risk of breast cancer later in life. If one tests positive for colon cancer susceptibility genes, the only recourse is frequent examination of the colon and immediate removal of any polps that appear. Clearly, the consequences of many of the genetic tests currently available create serious problems for those who discover that they carry disease-causing genes of one kind or another. As hundreds of additional genetic tests become available, patients and physicians will have to make difficult choices regarding the risks and benefits of genetic testing. Genetic discrimination: Profound social, ethical, and financial questions are raised by wide spread use of genetic testing and identification of persons at risk for diseases. Employers and insurance companies would like to have access to such information since it would be in their financial interest to know who is likely to become sick or disabled.

540

CHAPTER23 Structure and Properties of DNA

F I G U R E 23-17 A sampling of single-gene defects (Mendelian disorders) that have been mapped to specific chromosomal loci. More than 8000 inherited disorders and defects have been identified as stemming from mutations in single genes, and many of the altered base changes have been mapped to specific chromosomal loci. As the analysis of the DNA sequence of the human genome proceeds, it is expected that eventually thousands more mutated alleles that cause inherited disorders and defects will be identified and mapped.

SECTION 23.9

The Human 6enome Project

FIGURE 23-17

(Continued)

5/11

542 On the other hand, such use of personal medical information would generate a society that accepts the concept of genetic discrimination. Since our society has made great efforts to eliminate sex, racial, and age-related discrimination, the overwhelming sentiment is to not allow genetic discrimination. The question for society is how to guarantee that genetic discrimination will not be practiced either legally or illegally as more and more DNA information becomes part of peoples' medical records. Laws against genetic discrimination have been introduced in many states but there is no federal law (as of 2000) that guarantees a person's right to genetic privacy. Drug design: One of the anticipated benefits of the identification of human genes involved in diseases is the development of highly specific drugs that can be targeted to correct the biochemical and physiological consequences of a defective gene and its abnormal protein. It is expected that drugs eventually can be developed that will be targeted to correct the effects of specific mutations that individuals carry. Drug design and drug therapy may evolve to the point where each person's genotype determines the drug that will be used. Pharmacogenomics is the study of the relation between human genetic variability in the activity, toxicity, and metabolism of drugs. Examples include deficiencies of catabolic enzymes dihydropyrimidine dehydrogenase and thiopurine S-methyl transferase which can cause profound toxicity to fluoropyrimidine and mercaptopurine, respectively. These are used in cancer chemotherapy (Chapter 27).

23.10 Genomics and Proteomics Enormous progress has been made in recent years in deciphering the genetic basis of different human phenotypes and clinical diseases that result from inherited mutations. Technological breakthrough have made the identification of genes and their sequencing a relatively straightforward procedure. Genomics is the study of DNA and the genes that it contains. While genomics has resulted in the characterization of many human genes that are responsible for abnormal phenotypes and genetic disorders, identification of the protein corresponding to the gene is often difficult and, in many cases, is still unknown long after the gene has been identified and sequenced. Often the activity of the gene does not correlate with the amount of the protein or its activity in the cell. Posttranslational modifications cannot be determined by genomics, which only allows determination of the primary amino acid sequence as deduced from the nucleotide sequence of the gene. Proteomics is the direct study of proteins produced by both healthy and diseased cells. For example, twodimensional gel electrophoresis can be used to display

CHAPTER 23

Structure and Properties of DNA

and quantify thousands of proteins within a cell. A change in the level of a particular protein, either higher or lower, can be detected by differential protein abundance analysis. When a cell receives a chemical signal, such as the binding of a growth factor or cytokine, the immediate response is at the level of proteins that are unrelated to the target gene. Cell surface receptor proteins are modified in response to the external signal. Also, transmission of information from the cell surface to the nucleus is mediated by the physical movement of proteins. Proteomic technologies have been developed that can detect and analyze such protein changes. In addition, the level of a particular protein in the cell may change dramatically in response to a change in transcriptional regulation of one or more genes, and this can be monitored (Chapter 26). Proteomics is a valuable new technology used in the development of novel and more effective drugs following the identification and characterization of disease-causing proteins. Proteins also provide useful molecular markers for the diagnosis of specific diseases. In essence, proteomics provides the link between genes, proteins, and diseases and complements the information obtained by genomics.

Supplemental Readings and References Structure of DNA B. Alberts, A. B. Bray, J. Lewis, et al.: Molecular Biology of the Cell. New York: Garland (1994). S. Henikoff, E. A. Greene, S. Pietrokovski, et al.: Gene families: The taxonomy of protein paralogs and chimeras, Science 278, 609 ( ! 997). D. W. Ross: The human genome: information content and structure. Hospital Practice, June 15, 49 (1999).

Sequencing DNA L. Alphey: DNA Sequencing. New York: Springer-Verlag (1997). F. R. Blattner, G. Plunkott, C. A. Bloah, et al.: The complete genome sequence of Escherichia coli K-12. Science 277, 1453 (1997). F. S. Collins: Sequencing the human genome. Hospital Practice, January 15, 35 (1997). E S. Collins: Medical and societal consequences of the human genome project, New England Journal of Medicine 341, 28 (1999). N. Rosenthal: Fine structure of a gene--DNA sequencing. New England Journal of Medicine 332, 589 (1995). L. Rowen, G. Mahairas, and L. Hood: Sequencing the human genome. Science 278, 669 (1997).

Properties of DNA B. H. Hahn: Antibodies to DNA. New England Journal of Medicine 338, 1359(1998). D. E. Housman: DNA on trial--The molecular basis of DNA fingerprinting. New England Journal of Medicine 332, 534 (1995).

543

Supplemental Readings and References S. E Naber: Molecular pathology--diagnosis of infectious DNA. New England Journal of Medicine 331, 1212 (1994). W. Reik and M. A. Surami, eds.: Frontiers in Molecular Biology. New York: Oxford University Press (1997). C. W. Schmid, N. Deka, and G. Matera: Repetitive human DNA: The shape of things to come. In Chromosomes: Eukaryotic, Prokaryotic, and Viral. Volume 1. K. W. Adolph, ed., Boca Raton, FL: CRC Press (1990). R. A. Seder and S. Gurunathan: DNA vaccines--Designer vaccines for the 21st century. New England Journal of Medicine 341, 277 (1999).

Gene Therapy H. M. Blau and M. I. Springer: Muscle-mediated gene therapy. New England Journal of Medicine 333, 1554 (1995). M. Cavazzana-Calvo, S. Hacein-Ray, G. de Saint Basile, et al.: Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science 288, 669 (2000). S. J. Eck, ed.: Gene therapy. Hematology/Oncology Clinics of North America 12 (1998).

W. M. McDonnell and E K. Askari: DNA vaccines. New England Journal of Medicine 334, 42 (1996). L-C. Tsui and E Durie: Genotype and phenotype in cystic fibrosis. Hospital Practice, June 15, 15 (1997).

Human Genome Project G. J. Annas: Rules for research on human genetic variation--Lessons from Iceland. New England Journal of Medicine 342, 1830 (2000). E. Green: The Human Genome Project and its impact on the study of human disease. In Metabolic and Molecular Bases of Inherited Disease, C. R. Scriver, ed., 8th edition. New York, McGraw-Hill (2000). V. Hatzimanikatis, L. H. Choe, and K. H. Lee: Proteomics: Theoretical and experimental considerations. Biotech. Prod. 15, 312 (1999). N. A. Holtzman and T. M. Marteau: Will genetics revolutionize medicine? New England Journal of Medicine 343, 141 (2000). G. J. B. van Ommen, E. Bakker, and J. T. den Dunnen: The human genome project and the future of diagnostics, treatment, and prevention. The Lancet 354, si8, (Molecular Medicine) (1999). D. B. Searls: Using bioinformatics in gene and drug discovery. Drug Discovery Today 5, 135 (2000).

CHAPTER

24

DNA Replication, Repair, and Mutagenesis

DNA is the permanent repository for all genetic information in every cell and, as such, must maintain the fidelity of that information from one cell generation to the next as well as from one human generation to the next. This means, in the case of human beings, accurately copying the approximately 6 x 10 9 base pairs in each diploid cell prior to its undergoing mitosis (somatic cells) or meiosis (germ cells). Over billions of years of biological history, cells have developed intricate mechanisms to ensure faithful replication of DNA molecules and for the detection and repair of errors in the sequence of bases when they do occur. However, errors m u s t occur; the evolution of new organisms and new species depends on a supply of new genomes, the source of genetic diversity on which natural selection acts and which fuels biological evolution. This is the paradox of DNA replication. On the one hand, DNA must replicate itself with the utmost accuracy; on the other hand, it must allow a low frequency of genetic change to occur during each round of replication, particularly during meiosis, which produces the cells (sperm and ova) that pass DNA to succeeding generations. This apparent paradox is resolved by the mechanisms of mutation and recombination that provide for a small but essential amount of genetic change to occur in each

generation. Mutations occur spontaneously during DNA replication because not all of the replication errors or damage to templates are detected and repaired by the enzymes responsible for proofreading and repairing DNA. The rate of mutations can be markedly increased by ionizing radiation (x-rays), mutagenic chemicals (cigarette smoke), or retroviruses that integrate in DNA (HIV). Thus, mutations are essential to the species because they allow for adaptive changes but often are harmful to the individual by giving rise to cells that cause cancer, congenital defects, and other diseases. Recombination occurs primarily in germ cells of eukaryotes and during cell division in partially diploid prokaryotes. In general, recombination does not occur in mitosis of eukaryotic cells. However, recombination is an integral part of meiosis in which recombinant sperm and ova provide new genotypes and phenotypes in each generation which may be favored by natural selection. In this chapter, the basic mechanisms of DNA replication, recombination, repair, and mutation are discussed. Much of our understanding of these mechanisms derived from the study of bacteria. However, as further evidence for the unity of biology, the processes that replicate and modify DNA in bacteria also apply to human DNA with some modifications.

545

546

CHAPTER 24

DNA Replication, Repair, and Mutagenesis

24.1 DNA Replication

TABLE 24-1

P r o b l e m s of Replication

Comparison o f Parameters o f D N A Replication in E. coli and Human Cells

The replication process requires that each double-helical molecule of DNA produce two identical molecules of DNA. This means that wherever a G-C or A-T base pair occurs in the parental molecule, the identical base pair must occur in the progeny molecules. However, many factors interfere with accurate replication of DNA. If an A should pair with C or G with T as a result of tautomerization (Chapter 23), a point mutation (a change in one base pair) will result. Occasionally, a segment of DNA will be replicated more than once (duplication) or a segment may fail to be replicated (deletion). These and other aberrations in DNA replication do occur, but the mechanism of replication has evolved to minimize such mistakes.

DNA

E. coli

Amount of DNA -~3.9 x 106 bp Rate of replication at -850 bases/sec each replication fork Number of origins of one replication Time for cell division -30 min

Human -3 • 109 bp -6-90bases/sec 103 to 104 --24 hours

The information in each strand of the double helix serves as the template for the construction of a new double-helical DNA molecule; this is called semiconservative replication since one old strand of DNA is paired with one new strand to produce the daughter DNA molecule. All DNA molecules in all organisms replicate semiconservatively. This basic fact was first proved by Matthew Meselson and Frank Stahl who labeled DNA in E. coli by growing cells for several generations in medium containing a heavy isotope of nitrogen (15N). Cells were then transferred to medium containing 14N and continued to grow exponentially. After one generation of growth in 14N medium, the DNA was extracted from a sample of cells and analyzed by cesium chloride density centrifugation. All DNA was of intermediate (hybrid) density; one strand contained JSN and the newly synthesized strand contained 14N. After two generations of growth in 14N medium, 50% of the DNA contained only 14N (light DNA) and 50% is hybrid. This important experiment confirmed a prediction of the Watson-Crick model for DNA. Subsequent experiments also showed that human DNA undergoes semiconservative replication.

from one DNA molecule to another just as genes can be moved by genetic engineering techniques. In both bacterial and plasmid DNA, replication is initiated at a unique origin and proceeds in both directions along the DNA molecule to a termination site that is located approximately 180 degrees from the origin. Thus, replication of DNA is, in most cases, bidirectional replication. The ability of DNA to replicate from many origins and in both directions means that cells can replicate all of their DNA in a fairly short period of time. The shortest generation time for the bacterium E. coli is approximately 30 minutes in rich medium in the laboratory whereas the shortest division time for a human cell is approximately 24 hours (Table 24-1 ). Unwinding a double helix during replication presents a serious mechanical problem. Either the two daughter DNA molecules at the replication fork (the Y-shaped fork) must rotate around one another, or the unreplicated segment of DNA must rotate (Figure 24-1). This necessity for DNA rotation during replication creates topological problems for covalently closed circular DNA in bacteria and for the enormously long condensed and folded DNA in human chromosomes. The unwinding of strands of the DNA double helix is accomplished by a special group of enzymes called helicases; positive and negative coiling of DNA to maintain the topology necessary for replication is the function of another group of enzymes called topoisomerases (discussed in a later section).

Origin and Direction of DNA Replication

Replicons

Another unifying principle of DNA replication is that each molecule of DNA has one or more specific origins ofreplication where DNA synthesis begins. In bacteria that carry their genetic information in a circular DNA molecule, only one origin of replication exists. Plasmid DNA also contains a unique origin of replication. Origins of replication can be moved from one location in DNA to another, or

Much of the basic understanding of DNA replication was gleaned from studying the replication ofplasmids (Chapter 23): small, circular, extrachromosomal DNA elements in bacteria. Some plasmids, such as the F (fertility) factor in E. coli, have an origin of replication but replicate only unidirectionally from that single origin by a rolling-circle mechanism of replication (Figure 24-2). If an F factor

Semiconservative Replication

SECTION24.1 DNA Replication

FIGURE 24-1 Semiconservative replication of DNA. The two replicas consist of one parental strand plus one daughter strand. Each base in a daughter strand is selected by the requirement that it form a base pair with the parental base.

integrates into the bacterial chromosome, it can replicate its own D N A and bacterial D N A as well during conjugation, a process in which bacteria physically join and transfer D N A from the donor bacterium to the recipient bacterium. In this situation, bacteria replicate D N A simultaneously in a bidirectional manner from their own origin of replication (ori c) and unidirectionally from the origin of the integrated F factor (oriF). Such replication clearly raises topological problems during D N A replication (Figure 24-3). Studies of bacterial and plasmid D N A replication led to the concept of a replicon, which is: 1. A site on D N A consisting of a sequence of nucleotides that defines an origin of replication, and 2. Structural genes coding for proteins that recognize and bind to the origin to initiate D N A replication.

FIGURE 24-2 Rolling-circle or cr replication. Newly synthesized DNA is shown in color.

5/17

FIGURE 24-3 Replication of bacterial DNA with an integrated F factor. There are two origins of DNA replication, orir and oriF. Replication is bidirectional from orir and unidirectional from oriF. Somehow the cell solves this topological dilemma when cell division occurs.

These genes and the segment of D N A that is replicated from the origin are collectively defined as a replicon. Thus, a bacterial c h r o m o s o m e or a plasmid is a single replicon, whereas a single h u m a n c h r o m o s o m e may contain thousands of replicons.

Discontinuous D N A Replication In the diagram of replication shown in Figure 24-1, both daughter molecules are shown as having continuous strands. In truth, D N A cannot replicate by copying both strands continuously because D N A polymerases, the enzymes that synthesize new DNA, can only add nucleotides to a T - O H group. Because of the antiparallel nature of the

548

CHAPTER24 DNA Replication, Repair, and Mutagenesis 3'-, ~/5'

l llllllllllll

#

.

I'! l i, l llillll

Ligase seals nick

I i I I ! !11 l l I !I

Site of missing nucleotide

Ligase cannot seal a gap

NOactivity

(a)

(b)

F I G U R E 24-4 Action of DNA ligase. (a) A nick having a 3'-OH and a 5'-P terminus sealed (left panel). (b) If one or more nucleotides is absent, the gap cannot be sealed.

DNA strands, synthesis of one new strand of DNA can be continuous but synthesis along the other strand must occur by discontinuous replication. At each replication fork one DNA strand has a free 3'-OH group; the other has a free 5'-PO 2- group. Since DNA can only elongate in a 5' to 3' direction, synthesis of one strand (the leading strand) is continuous; synthesis of the opposite strand (the lagging strand) is discontinuous. Short fragments of DNA are synthesized in the replication fork on the lagging strand in a 5'to 3' direction; these are called Okazaki fragments after the scientist (Reiji Okazaki) who first demonstrated their existence during replication. These Okazaki fragments are joined in the replication fork by the enzyme DNA ligase that can form a phosphodiester bond at a single-strand break in DNA. DNA ligase joins a 3'-OH at the end of one DNA fragment to the 5'-monophosphate of the adjacent fragment (Figure 24-4). However, if even one nucleotide is missing in the DNA strand, ligase cannot seal the sugar-phosphate backbone.

24.2 Enzymology of DNA Replication The fundamental enzymology of DNA replication derives from both in vivo and in vitro studies with cells and extracts derived from E. coli. Many of the enzymes involved in DNA replication were identified by isolation of conditional lethal mutants of the bacterium, e.g., mutants that are unable to replicate DNA (and unable to grow) at high temperatures (42~ but that replicate and grow normally at low temperatures (30~ The synthesis of DNA is a complex process because of the need for faithful replication, enzyme specificity, and topological constraints. Approximately 20 different enzymes are utilized in bacteria to replicate DNA. In addition to polymerization reactions, DNA replication requires accurate initiation, termination, and proofreading to eliminate errors. DNA Polymerases Three DNA polymerases have been characterized in E. coli and are designated polymerase I, polymerase II, and polymerase III (Table 24-2). Although present in very low concentration in the cell, polymerase III, also called replicase, is the polymerase that elongates both strands of the bacterial DNA in the replication fork. Polymerase I is primarily a DNA repair enzyme and is responsible for excision of the short RNA primer that is required to initiate DNA synthesis on both the leading and lagging strands of DNA during replication. It also can remove mismatched base pairs during replication and fill in gaps in singlestranded DNA that is joined in a double helix. The function of polymerase II is not clear but it probably also has repair functions. All DNA polymerases select the nucleotide that is to be added to the 3'-OH end of the growing chain

T A B L E 24-2

Properties orE. coli DNA polymerases Property

Molecular weight Molecules/cell Nucleotides/sec 3' exonuclease activity 5' exonuclease activity Biological activity

Polymerase I

II

III

105,000

90,000

130,000

-400 -20 yes yes RNA primer excision, DNA repair

-100 --5 yes no SOS DNA repair?

-10 -1,000 no no Replicase

SECTION 24.2

Enzymologyof DNA Replication

549

accomplished by a potent pyrophosphatase, a widely distributed enzyme that breaks down pyrophosphate to inorganic phosphate. Hydrolysis of pyrophosphate has a large negative free energy, so essentially all of the pyrophosphate is rapidly removed. No DNA polymerase can catalyze the reaction between two free nucleotides, even if one has a 3'-OH group and the other a 5'-triphosphate. Polymerization can occur only if the nucleotide with the 3'-OH group is hydrogen-bonded to the template strand. Such a nucleotide is called aprimer (Figure 24-5). The primer can either be a single nucleotide or the terminus of a hydrogen-bonded oligonucleotide. When an incoming nucleotide is joined to a primer it supplies another free 3'-OH group, so that the growing strand itself is a primer. Since polymerization occurs only at the 3'-OH end, strand growth is said to proceed in the 5' --+ 3' direction. All known polymerases (both DNA and RNA) are capable of chain growth only in the 5' --+ 3' direction. This unidirectional feature ofpolymerases complicates the simultaneous replication of both strands of DNA. Polymerization is not confined to addition of a nucleotide to a growing strand in a replication fork. For example, pol I can also add nucleotides to the 3'-OH group at a single-strand (a nick) in a double helix. This activity results from the ability of pol I both to recognize a T-OH group anywhere in the helix and to displace the base-paired strand ahead of the available 3'-OH group.

and catalyze formation of the phosphodiester bond. The substrates for DNA polymerases are the four deoxynucleoside-5'-triphosphates (dATE dCTP, dGTP, and dTTP) and a single-stranded template DNA. The overall chemical reaction catalyzed by all DNA polymerases is Poly(nucleotiden)-3'-OH + dNTP Z poly(nucleotiden+ 1)-T-OH + PPi in which PPi represents pyrophosphate cleaved from the dNTE That is, a reaction occurs between a 3'-OH group at a terminus of a DNA strand and the phosphoryl group (the one linked to the sugar) of an incoming nucleoside triphosphate. Even though the hydrolysis of the nucleoside triphosphates has a large negative A G, the polymerization reaction as written still has a positive A G at concentrations present in a cell and in laboratory reactions (+0.5 kcal/mol = 2.1 kJ/mol). Thus, in the absence of any other reaction DNA polymerases would catalyze depolymerization rather than polymerization. Indeed, if excess pyrophosphate and a polymerase are added to a solution containing a partially replicating DNA molecule, the polymerase acts like a nuclease. In order to drive the reaction to the right, pyrophosphate must be removed, and this is

Product

Template

(a)

P-5'

3'-OH

HO-3'

(b)

[c)

,. . . . . . .

HO-3'

P-5'

3'-OH ,

(e)

NO-3'

3'-OH ...

.

.

P-5' .

.

P-5' HO-3', .

.

5'-P

HO-3'

Nick~

3'-OH 5'-9

P-5'

(f)

P-5'-5'-P

P-5' HO-3',,L

No synthesis

5'-P

HO-3'w.

(d)

No synthesis

5'-P

.

3'-OH

NO s y n t h e s i s

P-5'--

5'-9

NO-3'

5'-P

HO-3'

-

.

3'-OH 5'-P

3'-OH .

~

P-5' -

-

= 5'-P

FIGURE 24-5

Effect of various templates used in DNA polymerizationreactions. A free 3f-OHon a hydrogen-bondednucleotide at the strandterminus and a non-hydrogen-bondednucleotide at the adjacentposition on the template strand are needed for strand growth. NewlysynthesizedDNA is shownin color.

550

CHAPTER24 DNA Replication, Repair, and Mutagenesis

F I G U R E 24-6 Strand displacement and nick translation on linear and circular molecules. In nick translation, a nucleotide is removed by an exonuclease activity for each nucleotide added. The growing strand is shown in color.

This reaction is called strand displacement (Figure 24-6). Not all DNA polymerases can carry out strand displacement. Pol I has several enzymatic activities other than its ability to polymerize; one of these is important in maintaining continuity of the daughter strand, and the other improves the fidelity of replication. These two functions are a 5' --+ 3' exonuclease activity and a 3' --+ 5' exonuclease activity.

Exonuclease Activities of Polymerase I Occasionally DNA polymerase, in error, adds a nucleotide to the 3'-OH terminus that cannot hydrogen-bond to the corresponding base in the template strand. Such a nucleotide would clearly alter the information content of the daughter DNA molecule, and mechanisms exist for correcting such incorporation errors. Once having added an incorrect nucleotide and moved on to the next position, pol I cannot add another nucleotide because the enzyme requires a primer that is correctly hydrogen-bonded. Where such an impasse is encountered, a 3' --+ 5' exonuclease activity, which may be thought of simply as pol I running backward or in the 3' --+ 5' direction, is stimulated and the unpaired base is removed. After removal of this base, the exonuclease activity stops, polymerizing activity is restored, and chain growth resumes. This exonuclease activity is called the proofreading or editing function of polymerase I. Pol I also has a potent 5' ~ 3' exonucleolytic activity (Figure 24-7). This activity is directed against a basepaired strand and consists of stepwise removal of nucleotides one by one from the 5'-P terminus. Furthermore, the nucleotide removed can be either of the deoxyribo or the ribo type. The 5' ~ 3' exonucleolytic activity also can be coupled to the polymerizing activity. Recall that pol I can add nucleotides to a 3'-OH group at a nick and displace

the downstream strand. Under certain conditions, the displacement reaction does not occur and instead the 5' ---> 3' exonuclease acts on the strand that would otherwise be displaced, removing one downstream nucleotide for each nucleotide added to the 3' side of the nick. Thus, the position of the nick moves along the strand; this reaction is called nick translation. It is an important laboratory procedure for producing labeled DNA that can be used as probes; simply by carrying out the reaction in the presence of radioactive or chemically labeled nucleotides, an unlabeled DNA molecule with nicks in both strands can be converted to a labeled molecule. DNA probes are used for many diagnostic and forensic purposes (Chapter 23). The main function of the 5' --+ 3' exonuclease activity is to remove ribonucleotide primers that are used in DNA replication. Pol I also has a 5' --+ 3' endonuclease activity. An endonucleolytic cut is made between two base pairs that follow a 5'-P-terminated segment of unpaired bases, as shown below.

G

J Af l

G

o

"/

,5,~

G

lil

i

!

AI oI A!

T

T

I

~"~

C

I

T

Hydrolysis site

This type of endonucleolytic activity is unimportant in normal replication but is important in excision repair.

551

SECTION 24.2 Enzymology of DNA Replication

Product

Substrate

(a)

5'

3'

3'

5'

3'

3'

5', .

3'

5'

(b)

5' ""

3'

5'

5'

=

3' 5'

.

3' .

5'

.

3'

3'

5'

Nick (c)

3 ' ~ 5'

5'

'

3'

3' 5'

~-

3'

5'

5'

'

3'

3' 5'

RNA 3'

5'

(d)

~

3'

5. (e)

5'

.

N i c k 3 " ~ , 5, # R N A ,-. .

3'

,....

3'

3'

5'

5'

3 ............

3'

5'

5'

3'

5'

3'

5' 3'

5'

'

3' 5'

3'il~5' 5'

(f)

3'

~

3'

5'

N o exonucleolytic activity

FIGURE 24-7

Several substrates and products alter activity of the polymerase 15' --~ 3' exonuclease. The 5' terminus attackedby the enzyme is shown by the small straight arrow. (a) 5'-Terminalnucleotides are removed from each end of the molecule. (b) A gap is enlarged by cleavage at a 5' terminus. (c) A gap is formed by removal of a 5'-terminal nucleotide at a nick. The gap is then enlarged as in (b). (d) RNA (colored line) at the boundary of a gap is removed by cleavage at the 5' terminus of the RNA. (e) A gap is formed from a nick at a DNA-RNAboundary by removal of 5'-P-terminal ribonucleotides. (f) A non-hydrogen-bonded 5' terminus is resistant to the exonuclease; there is an endonucleolytic activity.

Polymerase 111 Polymerase I plays an essential role in the replication process in E. coli, but it is not responsible for the overall polymerization of the replicating strands. The enzyme that accomplishes this is a less abundant enzyme, polymerase III (pol III). (A D N A polymerase II has also been isolated from E. coli, but it probably plays no role in DNA synthesis.) Pol III catalyzes the same polymerization reaction as pot I but has certain distinguishing features. It is a very complex enzyme and is associated with eight other proteins to form the pol III holoenzyme. (The term holoenzyme refers to an enzyme that contains several different subunits and retains some activity even when one or more subunits is missing.) Pol III is similar to pol I in that it has a requirement for a template and a primer but its substrate specificity is much more limited. For a template pol III cannot act at a nick nor can it unwind a helix and carry out strand displacement. The latter deficiency means that an auxiliary system is needed to unwind the helix ahead of a replication fork. Pol III, like pol I, possesses a 3' --+ 5' exonuclease activity, which performs the major editing function in DNA replication. Polymerase III also has a 3' exonuclease activity, but this activity does not seem to play a role in replication.

Pol I and pol III holoenzyme are both essential for

E. coli replication. The need for two polymerases seems to be characteristic of all cellular organisms but not all viruses, e.g., E. coli. Phage T4 synthesizes its own D N A polymerase, which is capable of carrying out all functions necessary for synthesizing phage DNA. In the usual polymerization reaction, the activation energy for phosphodiester bond formation comes from cleaving of the triphosphate. Since DNA ligase can use a monophosphate, another source of energy is needed. This energy is obtained by hydrolyzing either ATP or NAD; the energy source depends on the organism from which the DNA ligase is obtained. Ligases have two major functions: the sealing of singlestrand breaks produced randomly in DNA molecules by nucleases and the joining of fragments during a particular stage of replication. DNA ligases are enzymes that can form a phosphodiester bond at a single-strand break in DNA, a reaction between a 3'-OH group and a 5'-monophosphate. These groups must be termini of adjacent base-paired deoxynucleotides (Figure 24-4). Bacteria usually contain a single species of ligase. Mammalian cells possess two DNA ligases (I and II) present in very small amounts compared with bacteria. Both

552

CHAPTER 24

DNA Replication, Repair, and Mutagenesis

TABLE 24-3

Properties of Eukaryotic DNA Polymerases Property

Polymerase a

Location in cell

Associated primase 3' exonuclease Sensitivity to aphidicolin Biological activity

Nucleus

Nucleus

Mitochondria

Nucleus

Nucleus

Yes No High Replication (lagging strand)

No No Low DNA repair

No Yes Low Replication

No Yes High Replication (leading strand)

No Yes Low Replication

eukaryotic ligases are located in the nucleus. Ligase I is predominant in proliferating cells and presumably plays a role in DNA replication; ligase II predominates in resting cells.

Eukaryotic Polymerases Five polymerizing enzymes have been isolated from many mammalian cells (Table 24-3). Threempol or, pol/3, and pol ymfunction in replication. Pol ot is the major polymerase of mammalian cells; it is found in the nuclei and is analogous to E. coli pol III. It is a multisubunit enzyme with a core (4-5 subunits) responsible for polymerization and a holoenzyme form possessing additional subunits and activities. It lacks the 3' --+ 5' exonuclease editing function. An intriguing protein subunit in the holoenzyme enables it to bind AppppA (diadenosine tetraphosphate), a small molecule that stimulates replication of resting mammalian cells and is hypothesized to be a growth signal. The pol ot holoenzyme of rat liver also possesses a DNA primase activity (see below), a feature not found in prokaryotic enzymes. Pol/3 is a nuclear polymerase, probably analogous in function to E. coli pol I. Pol y is found in mitochondria and is responsible for replication of mitochondrial DNA. It functions in the same way as pol III but is a single polypeptide.

24.3 The Replication Fork DNA replication requires not only an enzymatic mechanism for adding nucleotides to the growing chains but also a means of unwinding the parental double helix. These are distinct processes, and the unwinding of the helix is closely related to the initiation of synthesis of precursor fragments. The pol III holoenzyme cannot unwind the helix. In order for unwinding to occur, hydrogen bonds and

hydrophobic interactions must be eliminated, which requires energy. Pol I utilizes the free energy of hydrolysis of the triphosphate for unwinding as it synthesizes a DNA strand in a way that other polymerases cannot; instead, helix unwinding is accomplished by enzymes called helieases. These enzymes hydrolyze ATP and utilize the free energy of hydrolysis for unwinding. Unwinding of the helix by a helicase is not sufficient in itself for advance of the replication fork. Accessory proteins called single-stranded DNA-binding proteins (SSB proteins) are usually needed. As a helicase advances, it leaves in its wake two single-stranded regions: a longer one that is copied discontinuously and a shorter one just ahead of the leading strand. In order to prevent the single-stranded regions from reannealing or from forming intrastrand hydrogen bonds, the single-stranded DNA is protected with SSB proteins (Figure 24-8). SSB proteins bind tightly to both single-stranded DNA and to one another and hence are able to cover extended regions. As the polymerase advances, it must displace the SSB proteins so that base pairing of the added nucleotide can occur. Some phage replication systems utilize a single protein that functions as both a helicase and an SSB protein; the gene-32 protein of phage T4 is the prototype. It binds very tightly to single-stranded DNA and exceedingly tightly to itself, and its binding energy is great enough to unwind the helix. As a replication fork moves along a circular helix, rotation of the daughter molecules around one another causes the individual polynucleotide strands of the unreplicated portion of the molecule to become wound more tightly, i.e., overwound. (This may be difficult to visualize but can be seen by taking two interwound circular strings and pulling them apart at any point.) Thus, advance of the replication forks causes positive supercoiling (Chapter 23) of the unreplicated portion. This supercoiling obviously cannot increase indefinitely because soon the unreplicated portion becomes coiled so tightly that further advance of the

SECTION24.3 The Replication Fork

553

F I G U R E 24-8 Simplified overview of a bacterial DNA replication fork. The leading strand is initiated with an RNA primer and then elongated by a DNA polymerase, pol III (replicase). The lagging strand is synthesized in a series of fragments, each initiated by primase which inserts short sequences of RNA primers that will be elongated by a replicase. The RNA fragments are removed by another DNA polymerase, pol I, and the breaks in the sugar-phosphate backbone are sealed by DNA ligase.

replication fork is impossible. This additional coiling can be removed by atopoisomerase that can introduce negative superhelicity. In E. coli, the enzyme DNA gyrase produces negative superhelicity in nonsupercoiled covalent circles and is also responsible for removing the positive superhelicity generated during replication. The evidence for this comes from in vivo experiments using drugs (discussed later) that inhibit DNA gyrase; addition of any of these drugs to a growing bacterial culture or raising the temperature of cells with a temperature-sensitive gyrase protein inhibits DNA synthesis. Furthermore, in vitro replication of circular DNA can proceed only if DNA gyrase or a similar topoisomerase is present in the reaction mixture.

Inhibitors of DNA Replication Inhibitors of DNA synthesis are used in the laboratory and in treatment of bacterial, viral, and neoplastic diseases. Successful treatment of these conditions requires careful attention to dosage and the fine difference between drug effectiveness and toxicity. Inhibitors of DNA synthesis can be divided into three main classes: 1. Those that prevent or reduce the synthesis of precursors (bases, nucleotides), 2. Those that affect either the template or the priming ability of the growing strand, and 3. Those that act directly on polymerases or other enzymes needed for replication. A variety of inhibitors bind to DNA and thereby eliminate its template activity. Notable among these are the intercalating agents, which slip in between base pairs (e.g., the acridines, phenanthridium derivatives), and the anthracyclines (daunorubicin, doxorubicin, and plicamycin).

Other agents bind to DNA covalently and cause chain breakage (bleomycin, zinostatin) and interstrand crosslinks (alkyl sulfonates, anthramycin, mitomycin, nitrogen mustards). Several of these compounds are also useful antitumor agents. A variety of platinum coordination compounds bind to DNA and inhibit its template activity, apparently by binding to guanine. Substances that prevent extension of the growing chain (2',3'-dideoxyribonucleosides, cordycepin) are incorporated into the growing chain, but since they lack a 3'-OH group, further extension is not possible. Only a few substances act directly on DNA polymerases; they often are effective only on one or a small number of polymerases. For example, acyclovir inhibits the DNA polymerase of herpes simplex. Aphidicolin inhibits pol o~, pol 3 (but not pol/~ or pol V), many viral polymerases, and pol I and pol II of yeast; this compound is extremely valuable in laboratory research on DNA replication. Other components of the replication complex can also be inhibited; for example, 2'-dideoxyazidocytidine is an inhibitor of bacterial primase, and cournermycin, novobiocin, oxolinic acid, and nalidixic acid are effective inhibitors of DNA gyrase in bacteria.

Topoisomerase I lnhibitors A variety of antibiotics and antineoplastic drugs exert their therapeutic effects by interaction with topoisomerase I and disruption of DNA synthesis during phase of dividing cells. Topoisomerase I is essential for DNA replication and cell growth. The enzyme relieves torsional stress in DNA by inducing reversible single-strand breaks. The interaction of topoisomerase I and certain drugs produces double-strand breaks in DNA that are irreversible and can lead to cell death.

55~

CHAPTER24 DNA Replication, Repair, and Mutagenesis

F I G U R E 24-9 The basic structure of camptothecin. Analogues of camptothecin that are used to treat various neoplastic diseases involve substitutions at positions C-7, C-9, and C-11 of the basic molecule. All analogues as well as the parent molecule bind to topoisomerase I and interfere with its functions during DNA replication.

A variety of antibiotics and antineoplastic drugs function as topoisomerase I inhibitors; these include the quinolone antibiotics, anthracyclines (doxorubicin), epipodophylotoxins (etoposide), and the camptothecins, which are active in treating lung, ovarian, and colorectal cancers. The camptothecins also are used in the treatment of myelomonocytic syndromes, chronic myelomonocytic leukemia (CMML), acute leukemia, and multiple myeloma. A variety of synthetic analogues of natural camptothecins are being tested clinically for efficacy and safety in the treatment of these aggressive cancers (Figure 24-9). The camptothecins were discovered in extracts from the Chinese tree Camptotheca acuminata. Initial studies showed that camptothecins had antitumor activity, but clinical trials demonstrated severe side effects and toxicity. Numerous camptothecin analogues have been synthesized; several have received FDA approval (irinotecan and topotecan), whereas others are still in clinical trials.

24.4 Chromosome Replication DNA in mammalian cells is organized in complex structures called chromosomes (prokaryotes do not have a nucleus, do not divide by mitosis, and do not, strictly speaking, have chromosomes). The DNA in the chromosomes of human and other eukaryotic cells is intimately associated with two classes of proteins called histones and nonhistones. Collectively, DNA, histones, and nonhistones constitute chromatin, from which the name chromosome is derived. The DNA in a chromosome is an extremely long, linear molecule that must be condensed and organized to fit into the chromosomes in the nucleus. (The DNA in the 46 human chromosomes would be about 1 m long if fully extended.) Histones are responsible for the structural organization of DNA in chromosomes; the nonhistone proteins

F I G U R E 24-10 Structure of a nucleosome. DNA is looped around a core of eight histone proteins (pairs of four different histone proteins) and connected to adjacent nucleosomes by linker DNA and another histone (H 1).

regulate the functions of DNA including replication and gene expression. The positive charge of histones, due to the presence of numerous lysine and arginine residues, is a major feature of the molecules, enabling them to bind to the negatively charged phosphate groups in DNA. The electrostatic attraction is an important stabilizing force in chromatin. If chromosomes are placed in solutions of high salt that break down electrostatic interactions, chromatin dissociates into free histones and DNA. Chromatin also can be reconstituted by mixing purified histones and DNA in concentrated salt solutions and gradually removing the salt by dialysis. Histones share a similar primary structure among eukaryotic species. However, they undergo various posttranslational modifications such as phosphorylation, acetylation, methylation, and ADP ribosylation.The chemical modifications of histones can alter their net charge, shape, and other properties affecting DNA binding. Pairs of four different histones (H2A, H2B, H3, and H4) combine to form an eight-protein bead around which DNA is wound; this bead-like structure is called a nucleosome (Figure 24-10). A nucleosome has a diameter of 10 nm and contains approximately 200 base pairs. Each nucleosome is linked to an adjacent one by a short segment of DNA (linker) and another histone (H1). The DNA in nucleosomes is further condensed by the formation of thicker structures called chromatin fibers, and ultimately DNA must be condensed to fit into the metaphase chromosome that is observed at mitosis (Figure 24-11).

SECTION24.4 Chromosome Replication

555

Replication of DNA in Chromosomes The rate of movement of a replication fork in E. coli is about 105 nucleotides per minute; in eukaryotes the DNA polymerases move only about one-tenth as fast. To replicate all of the DNA in a human cell in a few hours means that replication must be initiated at thousands of origins of replication in each chromosome and move bidirectionally. To accomplish this, mammalian cells have thousands of times more DNA polymerase available than is found in bacteria. As replication proceeds through each nucleosome, the histones must dissociate to allow the replication fork to proceed; after replication, the nucleosome re-forms. The separation of parental and daughter DNA also requires the synthesis of new nucleosomes. Recent studies suggest that DNA is replicated in "replication factories," i.e., fixed sites within the nucleus consisting of the numerous proteins needed for replication. The DNA is replicated by being drawn through the replication factories rather than having the replication proteins move along the DNA.

Replication at the Ends of Chromosomes

F I G U R E 24-11 Hypothetical stages in the condensation of DNA with chromatin to form a chromosome. (a) Double-stranded DNA. (b)-(e) Formation of nucleosome beads and fibers consisting of histones and condensed DNA to form (f) a metaphase chromosome. These proposed intermediates are derived from dissociation reactions with intact chromosomes.

Despite the dense packing of DNA in chromosomes, it must be accessible to regulatory proteins during replication and gene expression. At a higher level of organization, chromosomes are divided into regions called euchromatin and heterochromatin. Transcription of genes seems to be confined mainly to euchromatic regions while DNA in heterochromatic regions is genetically inactive. When DNA is replicated during the S phase of the cell cycle, the histone and nonhistone proteins also are duplicated and combine with the daughter DNA molecules.

Since DNA in chromosomes is a linear molecule, problems arise when replication comes to the ends of the DNA. Synthesis of the lagging strand at each end of the DNA requires a primer so that replication can proceed in a 5' to 3' direction. This becomes impossible at the ends of the DNA and 50-100 bp is lost each time a chromosome replicates. Thus, at each mitosis of a somatic cell, the DNA in chromosomes becomes shorter and shorter. Ultimately, after a limited number of divisions, a cell enters a nondividing state, called replicative senescence, which may play an important role in biological aging. To prevent the loss of essential genetic information during replication, the ends of DNA in chromosomes contain special structures called telomeres that are synthesized by a specific enzyme called telomerase. Intact telomerase consists of an RNA primer and associated proteins so telomerase is actually a reverse transcriptase. The activity of telomerase in replenishing telomeres is regulated by a number of telomere-specific DNA-binding proteins, TRF1 and TRF2. TRF1 regulates the length of telomeres and TRF2 protects the ends. Overexpression of TRFI results in progressive shortening of telomeres; underexpression results in lengthening. Another telomere regulatory protein is tankrase (TRFl-interacting ankyrin-related ADPribose polymerase), which alters the activity of TRF1. Telomeres in human chromosomes consist of tandem repeats of the sequence TTAGGG. In most adult somatic cells, telomerase activity is very low or absent. Even in

CHAPTER24 DNA Replication, Repair, and Mutagenesis

556

hematopoietic stem cells that do have residual telomerase activity, telomere shortening is observed at the level of granulocyte and mononuclear cell fractions. Accelerated telomere shortening has been observed in cells from patients with aplastic anemia, suggesting that abnormal telomere shortening is associated with disease and aging. A characteristic of malignant tumor cells is that they can replicate indefinitely. The immortality of tumor cells appears to result, at least in part, from enhanced levels of telomerase that allow them to repair and elongate telomeres at the ends of DNA. This hypothesis is supported by the observation that ectopic expression of the catalytic subunit of telomerase (a product of the hTERT gene) enabled human retinal pigmented epithelial cells and fibroblasts to avoid senescence and to maintain their differentiated state when grown in vitro. Current research is focused on drugs that can promote or inhibit the action of telomerase or telomere-associated regulatory proteins. It is hoped that increasing telomerase activity in cells approaching senescence will retard aging or that decreasing telomerase activity in tumor cells will result in arrested tumor growth.

24.5 DNA Repair DNA can be damaged by external agents and by replication errors. Since maintenance of the correct base sequence of DNA and of daughter DNA molecules is essential for hereditary fidelity, repair systems have evolved that restore the correct base sequence.

5'

3' Template strand

3'4

GAGTCGAATC CTCAGTTTAG

--

5' Newly synthesized strand

Mismatch

excision

GAGTCGAATC CTC AG 9

Replacement correct base

ofI ~ ~

GAGTCGAATC CTCAG CTTAG

(a)

(b)

F I G U R E 24-12 Mismatch repair. (a) Excision of a short segment of a newly synthesized strand and repair synthesis. (b) Methylated bases in the template strand direct the excision mechanism to the newly synthesized strand containing the incorrect nucleotide. The regions in which methylation is complete are black lines; the regions in which methylation may not be complete are shown in color.

an enzyme, DNA methylase, methylates adenine in the sequence GATC. Methylation occurs soon (but not immediately) after the replication fork has synthesized such a sequence in the daughter strand. Thus, the nucleotides in the daughter strand are usually not methylated near the fork, whereas those in the parental strand are always completely methylated (Figure 24-12b). The mismatch repair system recognizes the degree of methylation of a strand and preferentially excises nucleotides from the undermethylated strand. The daughter strand is always the undermethylated strand, so that parental information is retained.

Mismatch Repair and Methylation of DNA DNA polymerases occasionally catalyze incorporation of an "incorrect" base that cannot form a hydrogen bond with the template base in the parental strand; such errors usually are corrected by the editing function of these enzymes. The editing process occasionally fails, so a second system, called mismatch repair, exists for correcting the errors that are not edited out. In mismatch repair, a pair of non-hydrogen-bonded bases (e.g., G . . . T; Figure 24-12a) within a helix are recognized as aberrant and a polynucleotide segment of the daughter strand is excised, thereby removing one member of the unmatched pair. The resulting gap is filled in by pol I, which presumably uses this "second chance" to form correct base pairs; then the final seal is made by DNA ligase. If it is only to correct and not create errors, the mismatch repair system must be capable of distinguishing the correct base in the parental strand from the incorrect base in the daughter strand. Rare methylated bases (methyl-A and methyl-C) provide the basis for this distinction. In E. coli,

Glycosylases Occasionally, uracil or other incorrect bases may become incorporated into new DNA strands. These bases are usually removed by a pathway that starts with the cleavage of the N-glycosidic bond by an enzyme called a glycosylase (Figure 24-13). Many glycosylases are known, and each is base-specific (e.g., uracil N-glycosylase). This enzyme cleaves the N-glycosidic bond and leaves the deoxyribose in the backbone. A second enzyme (AP endonuclease) makes a single cut, freeing one end of the deoxyribose. (AP stands for apurinic acid, a polynucleotide from which purines have been removed by hydrolysis of the N-glycosidic bonds.) This step is followed by removal of the deoxyribose and several adjacent nucleotides (probably by a second enzyme that acts at the other side of the apurinic site), after which pol I fills the gap with correct nucleotides. This sequence, endonuclease-enlargementpolymerase, is an example of a general repair mechanism called excision repair (see below).

557

SECTION24.5 DNA Repair

AGTGACTTAG TCACTGAATC

Ligase

weakened, causing inhibition of advance of the replication fork.

AGTGACTTAG TCACTGAATC I

I Deamination

AGTGACTTAG TCAUTGAATC i

Uracil

j l n

I~,-~

Polymerase

TGA

General Mechanisms for Repair of DNA

AGTGACTTAG TCA TGAATC

tl

N-glycosylase~ AGTGACTTAG TCA TGAATC

_~AP

endonuclease

i

U

FIGURE 24-13 Scheme for repair of cytosine deamination. The same mechanism could remove a uracil that is accidentally incorporated.

Alterations of DNA Molecules Several agents can break phosphodiester bonds. Among the more common are peroxides and various metal ions (e.g., Fe z+, Cu2+). Ionizing radiation also efficiently produces strand breaks. DNases present in cells probably also occasionally break phosphodiester bonds. Double-strand breakage, i.e., two single-strand breaks opposite one another, results from exposure to all forms of ionizing radiation. Single-strand breaks can be repaired by DNA ligases, although sometimes additional enzymes are required. Double-strand breaks rarely are repaired. Bases can be changed into different compounds by a variety of chemical and physical agents. For instance, ionizing radiation can break purine and pyrimidine rings and can cause several types of chemical substitutions, the most common being in guanine and thymine. The best studied altered base is the dimer formed by covalent linkage of two adjacent pyrimidine rings; these are produced by ultraviolet (UV) irradiation. The most prevalent of these dimers is the thymine dimer. The significant effects of the presence of thymine dimers are the following: 1. The DNA helix becomes distorted as the thymines, which are in the same strand, are pulled toward one another (Figure 24-14). 2. As a result of the distortion, hydrogen bonding to adenines in the opposite strand is significantly

CGAT

I I I

GC

I

AAC

I

I

T AG

I I

I

I

T A T T G A T C

UV

'C G A

~

G C T

/ rm \

I

I

I

T

A G

I I

I

A T C

FIGURE 24-14

Ultraviolet radiation damage of two adjacent thymines of DNA. Distortion of the DNA helix caused by two thymines moving closer together when joined in a dimer. The dimer is shown as two joined lines.

Repair of damaged bases was first observed and is best understood in bacteria. It is a widespread and probably universal phenomenon in both prokaryotes and eukaryotes. Some systems that repair dimers repair other types of DNA damage also. Four major pathways for DNA repair exist that can be subdivided into two classes: light-induced repair (photoreactivation) and light-independent repair (dark repair). The latter can be accomplished by three distinct mechanisms: 1. Excision of the damaged nucleotides (excision repair), 2. Reconstruction of a functional DNA molecule from undamaged fragments (recombinational repair), and 3. Disregard of the damage (SOS repair). Pho to reac tivatio n

Photoreactivation is a light-induced (300-600 nm) enzymatic cleavage of a thymine dimer to yield two thymine monomers. It is accomplished by photolyase, an enzyme that acts on dimers contained in single- and doublestranded DNA. The enzyme-DNA complex absorbs light and uses the photon energy to cleave specific C-C bonds of the cyclobutylthymidine dimer. Photolyase is also active against cytosine dimers and cytosinethymine dimers, which are also formed by UV irradiation but much less frequently. Excision Repair

Excision repair is a multistep enzymatic process. Several mechanisms are known, but only two will be described (Figure 24-15). All require an early incision step, in which a nuclease recognizes the distortion produced by a thymine dimer and makes a cut in the sugar-phosphate backbone. Following this, a DNA polymerase mediates a strand displacement step. In E. coli, two cleavages occur; the first is 12 nucleotides from the 5' side of the dimer and the second is 4-5 nucleotides from the 3' side. Each cut produces a 3'-OH and a 5'-P group. The T-OH group of the first cut is recognized by pol I, which then synthesizes a new strand, displacing the dimer-containing DNA strand. When the second cut is reached, the displaced fragment falls away and DNA ligasejoins the 3'-OH and 5'-P groups. In Micrococcus luteus, the first step is breakage of the N-glycosidic bond of the thymine at the 5' end of the dimer (by a dimer-specific glycosylase), leaving a free deoxyribose which is removed, leaving a free 3'-OH group. As

5~8

CHAPTER24 DNA Replication, Repair, and Mutagenesis

F I G U R E 24-15 Two modes of excision repair. (a) Escherichia coli mechanism. Two incision steps are followed by gap-filling and displacement by polymerase I. (b) Micrococcus luteus mechanism. A pyrimidine dimer glycosylase breaks an N-glycosidic bond and makes a single incision. Poi I displaces the strand, which is removed by an exonucleolytic event. In both mechanisms, the final step is ligation.

in E. coli, pol I polymerizes from this end and displaces the dimer-containing strand. After the dimer has been displaced, a second cut is made, the displaced strand falls away, and ligase forms the final phosphodiester bond. Repair of dimers in mammalian cells is more complicated and poorly understood, and requires a larger collection of enzymes.

Recombination Repair Recombination repair is a mechanism for generating a functional DNA molecule from two damaged molecules. It is an essential repair process for dividing cells because a replication fork may arrive at a damaged site, such as a thymine dimer, before the excision repair system has eliminated damage. When pol III reaches a thymine dimer, an adenine is added to the growing strand. However, the distortion of the helix caused by the dimer weakens the hydrogen bond and activates the polymerase editing function, and the adenine

is removed. The cycle begins againman adenine is added and then removed--the net result of which is that the replication fork fails to advance. A cell in which DNA synthesis is permanently stalled cannot complete a round of replication and does not divide. However, in a way that is not understood, after a pause of --~5 seconds per dimer, chain growth begins again beyond the thymine dimer block. The result of this process is that the daughter strands have large gaps, one for each unexcised thymine dimer. Viable daughter cells cannot be produced by continued replication alone because the strands having the thymine dimer will continue to turn out defective daughter strands and the first set of daughter strands would be fragmented when the growing fork enters a gap. However, by a recombination mechanism called sister-strand exchange proper doublestranded molecules can be made. The essence of sister-strand exchange is that a singlestranded segment free of any defects is excised from an undamaged strand on the homologous DNA segment at the replication fork and somehow inserted into the gap

559

SECTION24.6 DNA Mutation

created by the thymine dimer. The combined action of polymerase I and DNA ligase joins this inserted piece to adjacent regions, thus filling in the gap. The gap formed in the donor molecule is also repaired. If this exchange and gap filling are done for each thymine dimer, two complete single daughter strands can be formed, and each can serve in the next round of replication as a template for synthesis of normal DNA molecules. The system fails if two dimers in opposite strands are very near one another because no undamaged segments are available. Since recombinational repair occurs after DNA replication, in contrast with excision repair, it is often called postreplicational repair.

SOS Repair SOS repair includes a bypass system that allows DNA chain growth across damaged segments at the cost of fidelity of replication. It is an error-prone process; even though intact DNA strands are formed, the strands are often altered. As described above, activation of the editing system stalls replication at a thymine dimer. In SOS repair, the editing system is relaxed to allow polymerization to proceed across a dimer. Relaxation of the editing system means a loss of the ability to remove "incorrect" bases added to the growing strand. Most of the time, pol III inserts two adenines at a dimer site. However, the distortion increases the error frequency, allowing other nucleotides to be added to the chain. This error-prone repair is the major cause of UV-induced mutagenesis. An important nuclear protein, conserved from yeast to mammals, is the Ku heterodimer. This protein binds to DNA and repairs double strand breaks caused by x-rays and other agents. The Ku heterodimer is essential in maintaining chromosome integrity.

As predicted, nontumor cells cultured from these patients are hypersensitive to x-rays. Fanconi's syndrome, a lethal aplastic anemia, is also due to defective DNA repair. Cells from affected persons cannot repair interstrand cross-links or damage induced by x-rays. Two premature aging disorders (HutchinsonGilford syndrome and Bloom's syndrome) and several other disorders (Cockayne's syndrome and retinoblastoma) are also associated with defects in DNA repair. Cells from patients with some chromosome abnormalities (e.g., Down syndrome) may also show aberrant DNA repair. Several human DNA mismatch repair genes are associated with hereditary nonpolyposis colon cancer (HNPCC). One of these mismatch repair genes (hMSH2) is located on the short arm of chromosome 2; others are located on chromosome 3. Defects in any of these DNA repair genes predispose individuals to colon cancer as well as to other cancers.

24.6 DNA Mutation Mutation refers to any change in the base sequence of DNA. The most common change is a substitution, addition, rearrangement, or deletion of one or more bases. A mutation need not give rise to a mutant phenotype. A mutagen is a physical agent or chemical reagent that causes mutations. For example, nitrous acid reacts with some DNA bases, changing their chemistry and hydrogen bonding properties, and is a mutagen. Mutagenesis is the process of producing a mutation. If it occurs in nature without the action of a known mutagen, it is called spontaneous mutagenesis and the resulting mutations are spontaneous mutations. If a mutagen is used, the process is called induced mutagenesis.

Human Diseases and DNA Repair Deficiency Human disease may result from inability to carry out certain stages of DNA repair. The best studied disease, xeroderma pigmentosum, is a result of mutations in genes that encode the UV excision system. Cells cultured from tissue obtained from affected individuals are killed by much smaller doses of UV light than are normal cells. Furthermore, the removal of thymine dimers in DNA from these cells is very inefficient. People with this disease develop skin lesions when exposed to sunlight and commonly develop one of several kinds of skin cancer. Ataxia telangiectasia is characterized by severe abnormalities in various organ systems and a high incidence of lymphoreticular cancer. Defective DNA repair was suspected when patients developed an unexpected severe or fatal reaction while undergoing radiotherapy for cancer.

Types of Mutations Mutations can be categorized in several ways. One system is based on the nature of the change, specifically on the number of bases changed. Thus, we distinguish a point mutation, in which a single base pair is changed from a multiple mutation, in which two or more base pairs differ from the wild-type sequence. A point mutation may be a base substitution, a base insertion, or a base deletion, but the term most frequently refers to a base substitution. A second system is based on the consequence of the change in terms of the amino acid sequence that is affected. For example, if there is an amino acid substitution, the mutation is a missense mutation. If the substitution produces a protein that is active at one temperature (typically 30~ and inactive at a higher temperature (usually

560

CHAPTER24 DNA Replication, Repair, and Mutagenesis

i

~~:~.~i . !:.-~i~ !i Acridine

(a)

....y...~ ...

Tyr

i~;~i~i~=:~:,~~,:~ '1=":"~" C.G

Glu

Thr

Gly

lie

TACGAATCGGGTATT ATGCTTAGCCCATAA

rq+ /

~

c. G

/

\

r Replication in the

\

presence of an acridine iii

TACGAGATCGGGTATT ATGCT CTAGCCCATAA (a)

E r r o r in r e p l i c a t i o n : A . T --~ G 9C

(b)

Error in incorporation:

G 9C --, A 9T

F I G U R E 24-16 Two mechanisms of 5-bromouracil (BU)-induced mutagenesis. (a) During replication, BU in its usual keto form substitutes for T, and the replica of an initial A-T pair becomes an A-BU pair. In the first mutagenic round of replication, BU in its rare enol form pairs with G. In the next round of replication, the G pairs with a C, completing the transition from an A-T pair to a G-C pair. (b) During replication of a G-C pair, a BU in its rare enol form pairs with a G. In the next round of replication, the BU is again in the common keto form and it pairs with A, so the initial G-C pair becomes all A-T pair. The replica of the A-BU pair produced in the next round of replication is another A-T pair.

40-42~

the mutation is called a temperature-sensitive (Ts) mutation. Sometimes no amino acid corresponds to a new base sequence (Chapter 25); in that case, termination of synthesis of the protein occurs at that point, and the mutation is called a chain termination mutation or nonsense mutation. These mutations generate one of the three nonsense codons: UAA, UAG, or UGA (Chapter 25). Replication errors can be introduced by base analogues that can be added to a replicating DNA molecule. Such a base analogue must be able to pair with the base on the complementary strand being copied, or the 3' --+ 5' editing function would remove it. However, if it can tautomerize (Chapter 23), or if it has two modes of hydrogen bonding, it will be mutagenic. The substituted base 5-bromouracil (BU) is an analogue of thymine inasmuch as the bromine atom has about the same van der Waals radius as the methyl group of thymine. BU forms a nucleoside triphosphate, and DNA polymerases will add BU to a DNA strand opposite an adenine. However, BU tautomerizes at high frequency to a form that can pair with guanine. Thus, if B U replaces a thymine, in subsequent rounds of replication it occasionally generates a guanine in the complementary strand, which in turn specifies cytosine, resulting in formation of a GC pair (Figure 24-16). A chemical mutagen is a substance that can alter a base that has already been incorporated into DNA and thereby change its hydrogen bonding specificity. Nitrous acid, a

(b)

Tyr

Glu

lie

Gly

Tyr

FIGURE 24-17 Mutagenesis by intercalating substances (e.g., acridine). (a) Separation of two base pairs (shown as a box) by an intercalating agent. (b) A base addition resulting from replication in the presence of an acridine. The change in amino acid sequence read from the upper strand in groups of three bases is also shown.

powerful mutagen, converts amino groups to keto groups by oxidative deamination. Thus, cytosine, adenine, and guanine are converted to uracil (U), hypoxanthine (H), and xanthine (X), respectively, which can form the base pairs UA, HC, and XC, respectively. As a consequence, in a later round of replication, GC-to-AT and AT-to-GC mutations can occur. The chemical mutagen hydroxylamine reacts specifically with cytosine and converts it to a modified base that pairs only with adenine; thus, a GC pair ultimately becomes an AT pair. An interesting class of mutagen are intercalating agents, e.g., the acridines. These planar molecules insert between base pairs. When replication occurs in the region of an intercalated molecule, one or both daughter strands are synthesized that either lack one or more nucleotides or have additional ones. These changes alter the reading frame of the base sequence of a gone and hence are called frameshift mutations (Figure 24-17).

Triplet Repeats and Fragile Sites Inherited and acquired (sporadic) mutations account for an enormous diversity of human diseases and disorders. At least 300 genetic disorders include mental retardation as one of the symptoms, implying that many genes can affect brain development and mental functions. In the 1970s, a defect at the tip of the long arm of X chromosomes, called a fragile site, was discovered in a large number of patients with mental retardation. It is now known that a specific mutation is responsible for the fragile site on

SECTION24.6 DNA Mutation

561

TABLE 24-4

Inherited Diseases Involving Repeated Trinucleotides That Are Present in an Excessive Number of Copies* Disease Fragile X syndrome

Huntington's disease

Myotonic dystrophy Spinocerebellar ataxia Type 1

Fredreich's ataxia

Machado-Joseph disease

Trinucleotide repeat CGG 5-54 unaffected 60-230 carriers 230- 4000 affected CAG 11-30 unaffected 36-121 affected CTG CAG 19-36 unaffected 40-81 affected GAA 7-22 unaffected 200-900 affected CAG 12-37 unaffected 61-84 affected

Symptoms Mental retardation

Chorea, progressive dementia, death

Muscle weakness, myotonia Ataxia, dysarthria, rigidity, abnormal eye movements

Ataxia, dysarthria, hypertropic cardiomyopathy, diabetes

Spasticity, dystonia

*The extent of the repeated triplet nucleotide usually determines the severity of the disease and the age of onset. These diseases may be caused by slippage of the DNA polymerase during replication that increases the length of the repeated triplet from generation to generation. The patterns of inheritance are: Fragile X syndrome (X-linked); Huntington's disease, myotonic dystrophy, Spinocerebellar ataxia Type I, MachadoJoseph disease (autosomal dominant); Fredreich's ataxia (autosomal recessive).

the X chromosome and the associated inherited disorder

fragile X syndrome. This particular mutation accounts for about 20% of all mental retardation; about 1 in 1500 boys and 1 in 2500 girls are born with fragile X syndrome. Although fragile X syndrome is classified a Mendelian dominant sex-linked disorder, it does not behave as a classically dominant gene inherited disease. Only about 80% of males and 35% of females who carry the mutation and a fragile site suffer from mental retardation. For many years it remained a mystery how both men and women could carry a fragile X chromosome and be unaffected. In the 1990s, the molecular explanation for fragile X syndrome was revealed. The gene responsible for fragile X syndrome (FMR1) was found to contain a repeat of the trinucleotide CGG. In unaffected individuals, the number of CGG repeats ranges from about 5 to 54 copies. In mentally retarded individuals, the number of CGG repeats is dramatically increased to hundreds or thousands of copies. When the CGG repeat exceeds 230, the DNA becomes abnormally methylated in that region and the gene becomes nonfunctional. Presumably the amplification of the CGG repeat occurs during DNA replication in germ cells prior to meiosis perhaps due to "stuttering" of the

DNA polymerase as it reads the repeating trinucleotides. Why amplification of the CGG repeats occurs in some X chromosomes but not others and in some individuals and not others is unresolved. Triplet repeats account for a growing list of muscularneurologic diseases including Huntington's disease, myotonic dystrophy, Friedreich's ataxia, and others (Table 24-4). Triplet repeats represent an entirely new class of mutations. They are not caused by any external mutagenic agent but are generated during normal DNA replication. Fragile sites are chromosomal regions that are prone to breakage during chromosome movement or recombination; fragile sites can be visualized by a variety of cytological techniques and dyes that bind to metaphase chromosomes. Fragile sites are relatively common in chromosomes and there is a growing suspicion that they may play a greater role in disease, especially cancer, than previously thought. The chromosomes in many tumor cells exhibit fragile sites that are not present in normal cells. What is not yet clear is whether a fragile site initiates the conversion of a normal cell to a cancer cell or is merely the consequence of the altered growth properties of the

562 tumor cell. One possibility is that fragile sites are more susceptible to carcinogenic chemicals and that mutations in fragile sites contribute to the transformation of a normal cell to a tumor cell.

Supplemental Readings and References S. G. Arbuck and C. H. Takimoto: An overview of topoisomerase I-targeting agents. Seminar in Hematogy 3g(Suppl. 4), 3 (1998). S. E. Ball, E M. Gibson, S. Rizzo, et al.: Progressive telomere shortening in aplastic anemia. Blood 91, 3582 (1998). V. A. Bohr: Gene specific DNA repair. Carcinogenesis 12, 1983 (1991). D. Bootsma: The genetic defect in DNA-repair deficiency syndromes. European Journal of Cancer 29, 1482 (1993). C. E. Bronner, S. M. Baker, P. T. Morrison, et al.: Mutation in the DNA mismatch repair gene homologue hMLH1 is associated with hereditary non-polyposis colon cancer. Nature 368, 258 (1994). C. H. C. M. Buys: Telomeres, telomerase, and cancer. New England Journal of Medicine 342 (2000). J. E. Cleaver: Stopping DNA replication in its tracks. Science 285, 212 (1999). A. Durr, M. Cossee, Y. Agid, et al.: Clinical and genetic abnormalities in patients with Friedreich's ataxia. New England Journal of Medicine 335, 1169 (1996).

CHAPTER24 DNA Replication, Repair, and Mutagenesis M. Engelhardt, R. Kumar, J. Albanell, et al.: Telomerase regulation, cell cycle, and telomere stability in primitive hematopoietic cells. Blood 90, 183 (1997). M. Fessel: Telomerase and the aging cell. Journal of the American Medical Association 279, 1732 (1998). E. C. Friedberg, G. C. Walker, and W. Siede: DNA Repair and Mutagenesis. Washington, DC: ASM Press (1995). M. Grunstein: Histone acetylation in chromatin structure and transcription. Nature 389, 349 (1007). J. H. J. Hoeijmakers: Nucleotide excision repair II: from yeast to mammals. Trends in Genetics 9, 211 (1993). B. Kremer, E Goldberg, S. E. Andrew, et al.: A worldwide study of the Huntington disease mutation. New England Journal of Medicine 330, 1401 (1994). C. Lengauer, K. W. Kinzler, and B. Vogelstein: Genetic instabilities in human cancers. Nature 396, 543 (1998). T. Lindahl and R. D. Wood: Quuality conntrol by DNA rpair. Science 286, 1897 (1999). J.-L. Mandel: Breaking the rule of three. Nature 386, 767 (1997). D. W. Ross: Cancer: The emerging molecular biology. Hospital Practice 35, 63 (2000). S. Smith, I. Giriat, A. Schmitt, et al.: Tankyrase, a poly (ADPribose) polymerase at human telomeres. Science 282, 1484 (1998). T. A. Steitz: DNA polymerises: Structural diversity and common mechanisms. Journal of Biological Chemistry 274, 17395 (1999). S. T. Warren: Trinucleotide repetition and fragile X syndrome. Hospital Practices 32, 73 (1997).

CHAPTER

25

RNA and Protein Synthesis

Most proteins are enzymes that catalyze the myriad of chemical reactions in cells that are necessary for life; other proteins form structural functions as in bone and muscle. The information for making proteins resides in the sequence of bases in DNA in chromosomes and in organelles such as mitochondria. Converting the information contained in genes into proteins involves two complex processes. Transcription is the first step in which the sequence of bases in a gene is converted into a complementary sequence of bases in a molecule of RNA. Three chemically identical but functionally quite different molecules of RNA are transcribed from DNA: messenger RNA (mRNA) carries the genetic information contained in a gene; transfer RNA (tRNA) and ribosomal RNA (rRNA) are also transcribed from genes but are used to convert the information in the sequence of bases in mRNA into the corresponding sequence of amino acids in a protein. Translation is the process by which an mRNA is "read" by tRNAs, ribosomes (complex structures consisting of rRNAs and ribosomal proteins), and numerous other enzymes. Each type of cell is programmed to synthesize only those proteins necessary for its particular cellular functions (Chapter 26). The difference between a neuron and a liver cell is the kind of proteins that are synthesized even though both cells contain exactly the same genetic information. Cellular differentiation is due to differential gene expression; a tumor cell invariably is a cell that has lost the ability to regulate and express its genetic information

correctly and generally grows in an unregulated manner, as opposed to normal cells whose growth is regulated. The flow of information in all cells is from DNA to RNA to protein, which is known as the central dogma of molecular biology; it was formulated by Francis Crick shortly after the discovery of the structure of DNA. Information also can flow from DNA to DNA in both cells and among viruses that infect cells. Information also flows from RNA to RNA during the replication of RNA viruses such as the polio virus. The final permitted information transfer is from RNA to DNA, which only occurs in the case of retroviruses such as human immunodeficiency virus (HIV). The only information transfer that is prohibited by the central dogma is from protein to RNA or to DNA. The permitted information transfers in cells (infected or uninfected) is summarized below.

@

A~

>. @ A

~, Protein

The synthesis of a protein in a human cell can be broadly outlined as follows. A mRNA molecule is transcribed from a single strand of DNA (the "sense" strand) in the nucleus. The mRNA is processed by splicing out nontranslatable segments of nucleotides (introns) and rejoining the translatable segments (exons). Additional chemical modifications are made to both ends of the mRNA molecule and it is transported into the cytoplasm. The mRNA is translated

563

564 in the cytoplasm where groups of three bases in the mRNA (codons) are recognized by anticodons in specific tRNAs that carry a particular amino acid. The genetic code consists of 64 different codons that specify all 20 amino acids as well as codons that function to initiate and terminate translation. More than one codon may specify the same amino acid, which is called degeneracy of the genetic code. Finally, every organism from bacteria to human uses the same codons to specify the same amino acids; this is why the genetic code is said to be universal. Amino acids are joined together in a specific order determined by tRNAs, ribosomes, and associated enzymes that translate the mRNA. Each amino acid is joined to its neighbor by a peptide bond. The specific amino acid sequence of a protein specifies its three-dimensional structure. Some proteins require the help of chaperonins to fold into a functional configuration. When synthesis of a polypeptide chain is completed on a ribosome, it is released from the ribosome and may join with one or more similar or different polypeptides to constitute a functional protein.

25.1 Structure of RNA RNA is a single-stranded polynucleotide containing the nucleosides adenosine, guanosine, cytosine, and uridine. Roughly one-third to one-half of the nucleotides are engaged in intrastrand hydrogen bonds, with single-stranded segments interspersed between double-stranded regions that may contain up to about 30 base pairs. The base pairing produces conformations that are important to the function of the particular RNA molecules.

Ribosomal RNA (rRNA) Ribosomes contain several different RNA molecules, three in prokaryotic ribosomes and four in eukaryotic ribosomes. For historical reasons, each class is characterized by its sedimentation coefficient, which represents a typical size. For prokaryotes, the three Escherichia coli rRNA molecules are used as size standards; they have sedimentation coefficients of 5S, 16S, and 23S. The E. coli rRNA molecules have been sequenced and contain 120, 1541, and 2904 nucleotides, respectively. The sizes of the prokaryotic rRNA molecules vary very little from one species of bacterium to another. Eukaryotic rRNA molecules are generally larger and there are four eukaryotic rRNA molecules. Rat liver rRNA molecules are used as standards; the S values and the average number of nucleotides are 5S (120), 5.8S (150), 18S (2100), and 28S (5050), respectively. The eukaryotic 5.8S species corresponds functionally to the prokaryotic

CHAPTER 25

RNA and Protein Synthesis

5S species; no prokaryotic rRNA molecule corresponds to the eukaryotic 5S rRNA.

Transfer RNA (tRNA) Transfer RNA molecules range in size from 73 to 93 nucleotides. Since they function as amino acid carriers, they are named by adding a superscript that designates the amino acid carried, e.g., tRNA Ala for alanine tRNA. All tRNA molecules studied contain extensive doublestranded regions and form a cloverleaf structure in which open loops are connected by double-stranded stems. By careful comparison of the sequences of more than 200 different tRNA molecules, common features have been found and a "consensus" tRNA molecule consisting of 76 nucleotides arranged in a cloverleaf form has been defined (Figure 25-1). By convention, the nucleotides are numbered 1 through 76 starting from the 5'-P terminus. The standard tRNA molecule has the following features: 1. The 5'-P terminus always is base-paired, which probably contributes to the stability of tRNA. 2. The 3'-OH terminus always is a four-base single-stranded region having the base sequence XCCA-3'-OH, in which X can be any base. This is called the CCA or acceptor stem. The adenine in the CCA sequence is the site of attachment of the amino acid by the cognate synthetase. 3. tRNA has many "modified" bases. A few of these, dihydrouridine (DHU), ribosylthymine (rT), pseudouridine (~p), and inosine (I), occur in particular regions. 4. tRNA has three large single-stranded loops. The anticodon loop contains seven bases. The loop containing bases 14-21 is called the DHU loop; it is not constant in size in different tRNA molecules. The loop containing bases 54-60 almost always contains the sequence T ~ C and is called the T~pC loop. 5. Four double-stranded regions called stems (or arms) often contain GU base pairs. The names of the stems match the corresponding loop. 6. Another loop, containing bases 44-48, is also present. In the smallest tRNAs it contains four bases, whereas in the largest tRNA molecule it contains 21 bases. This highly variable loop is called the extra arm.

25.2 Messenger RNA Messenger RNA molecules in prokaryotic and eukaryotic cells are similar in some structural aspects but also differ significantly. All messenger RNAs contain the same four nucleotides, A, C, G, and U, and utilize the codon AUG to

565

SECTION25.2 Messenger RNA

F I G U R E 25-1 (a) The currently accepted "standard" tRNA cloverleaf with its bases numbered. A few bases present in almost all tRNA molecules are indicated. (b) Schematic diagram of the three-dimensional structure of yeast tRNA Phe. (Courtesy of Dr. Sung-Hou Kim.)

initiate translation of a polypeptide and the codons UAG, UGG, and UAA to terminate translation. Prokaryotic mRNAs are polycistronic (polygenic) and usually carry information for the synthesis of several polypeptides from a single mRNA. The triplet codons in prokaryotic mRNA are transcribed from the sense strand of DNA and subsequently are translated continuously from the 5 t - P O 4 end of the mRNA to the 3'-OH end. Since prokaryotic DNA is not separated from the cytoplasm by a nuclear membrane, translation begins on mRNA molecules before transcription is completed. Thus, transcription and translation are coupled in prokaryotes. Synthesis of each polypeptide chain in a polycistronic mRNA is determined by an AUG initiation codon and one or more nonsense codons that release the finished polypeptide from the ribosome. Eukaryotic mRNAs differ from prokaryotic mRNAs in several respects (Figure 25-2). Eukaryotic genes invariably contain information for only a single polypeptide but each gene may consist of millions of nucleotides because eukaryotic genes contain introns and exons. The mRNA that is transcribed (primary transcript) is processed in several ways: 1. The introns (intervening sequences) are spliced out of the primary transcript and the exons (expressed

sequences) are joined together. The splicing reactions and removal of introns from the primary transcript are carried out by small nuclear ribonucleoproteins (snRNPs). 2. While transcription is in process, the 5' end of the mRNA is capped with a methyl guanine nucleotide (m y Gppp).

3. After the primary transcript is complete, a polyA tail (-AAAn AoH) is added to the 3' terminus.

Prokaryotic mRNA 5' @

A Gene 1

~

XS~O ~/&G~ Gene 2

~ G : 3'

Gene 3

Eukaryotic mRNA (Primary transcript)

Cap I = intron

AAAn A-3' poly A tail One G e n e

E = oxon

F I G U R E 25-2 Structures of prokaryotic and eukaryotic primary transcripts (mRNAs). Prokaryotic mRNAs are polygenic, do not contain introns or exons, and are short lived in the cell. Eukaryotic mRNAs are monogenic, contain introns and exons, and usually are long lived in the cell.

566

4. Other modifications of the primary transcript are possible such as alternative splicing which produces mRNAs with different sets of exons and RNA editing in which bases are modified or changed in the original transcript. 5. The functional mRNA is transported to the cytoplasm where translation occurs on ribosomes bound to the endoplasmic reticulum of the cell.

25.3 Enzymatic Synthesis of RNA The basic chemical features of the synthesis of RNA are the following (Figure 25-3): 1. The precursors of RNA synthesis are the four ribonucleoside 5'-triphosphates (rNTPs): ATE GTP, CTP, and UTE The ribose portion of each NTP has an -OH group on both the 2' and the 3' carbon atoms. 2. In the polymerization reaction, a 3'-OH group of one nucleotide reacts with the 5'-triphosphate of a second nucleotide; a pyrophosphate is removed and a phosphodiester bond is formed. This same reaction occurs in the polymerization of DNA. 3. The sequence of bases in an RNA molecule is determined by the base sequence of the DNA template strand. Each base added to the growing end of an RNA chain is chosen by base pairing with the appropriate base in the template strand; thus, the bases C, T, G, and A in a DNA strand cause incorporation of G, A, C, and U, respectively, in the newly synthesized RNA molecule. The RNA is complementary to the template DNA strand, which is called the coding (sense) strand or template strand. 4. The RNA chain grows in the 5' ~ 3' direction, which is the same as the direction of chain growth in DNA

CHAPTER25 RNA and Protein Synthesis

synthesis. The RNA strand and the DNA template strand are also antiparallel. 5. RNA polymerases, in contrast with DNA polymerases, can initiate RNA synthesis, i.e., no primer is needed. 6. Only ribonucleoside 5'-triphosphates participate in RNA synthesis, and the first base to be laid down in the initiation event is a triphosphate. Its 3'-OH group is the point of attachment of the subsequent nucleotide. Thus, the 5' end of a growing RNA molecule terminates with a triphosphate. In tRNAs and rRNAs, and in eukaryotic mRNAs, the triphosphate group is removed.

E. coli RNA polymerase consists of five subunits--two identical ot subunits and one each of/3, ~', and o - - h a v i n g a total molecular weight of 465,000; it is one the largest enzymes known. The ot subunit is easily dissociated from the enzyme and, in fact, does so shortly after polymerization is initiated. The term core enzyme describes the o-free unit (Otzflfl'); the complete enzyme is called the holoenzyme (ot2/3/~'~). In this chapter, the name RNA polymerase is used when the holoenzyme is meant. Several different RNA polymerases exist in eukaryotes and are described below. An E. coli cell contains 3000-6000 RNA polymerase molecules; the number is greater when cells are growing rapidly. In eukaryotes, the number of RNA polymerase molecules varies significantly with cell type and is greatest in cells that actively make large quantities of protein, e.g., secretory cells.

25.4 Prokaryotic Transcription The first step in prokaryotic transcription is the binding of RNA polymerase to DNA at a particular region called

FIGURE 25-3 Model of RNA synthesisby RNA polymerasefrom the sense strand of DNA. See text for details.

SECTION25.4 Prokaryotic Transcription

567

F I G U R E 25-4 Segments of the coding strand of conserved regions from various genes showing the common sequence of six bases. The start point for mRNA synthesis is indicated by the heavy letters. The "conserved T" is underlined.

a promoterma specific nucleotide sequence, which for different RNA classes range in length from 20 to 200 nucleotides. In bacteria, a promoter is divided into subregions called t h e - 3 5 sequence (8-10 bases long) and t h e - 1 0 or Pribnow box (6 bases). The bases in t h e - 3 5 sequence are quite variable but the Pribnow boxes of all promoters are similar (Figure 25-4). Both the-35 sequence and the Pribnow box interact with RNA polymerase to initiate RNA synthesis. Following RNA polymerase binding to a promoter, a conformational change occurs such that a segment of the DNA is unwound and RNA polymerase is positioned at the polymerization start site. Transcription begins as soon as the RNA polymerase-promoter complex forms and an appropriate nucleotide binds to the enzyme. RNA polymerase contains two nucleotide binding sites called the initiation site and the elongation site. The initiation site binds only purine triphosphates (ATP and GTP), and one of these (usually ATP) becomes the first nucleotide in the RNA chain. Thus, the first DNA base that is copied from the DNA is usually thymine. The initiating nucleoside triphosphate binds to the enzyme and forms a hydrogen bond with the complementary DNA base. The elongation site is then filled with a nucleoside triphosphate that is selected strictly by its ability to form a hydrogen bond with the next base in the DNA strand. The two nucleotides are then joined together, the first base is released from the initiation site, and initiation is complete. The dinucleotide remains hydrogen-bonded to the DNA. The elongation phase begins when the polymerase releases the base and then moves along the DNA chain. The drug rifampin binds to bacterial RNA polymerases and is a useful experimental inhibitor of initiation of transcription. It binds to the fl subunit of RNA polymerase, blocking the transition from the chain initiation phase to the elongation phase; it is an inhibitor of chain initiation but not of elongation. Actinomycin D also inhibits initiation but does so by binding to DNA. These drugs have limited clinical use because of their toxicity.

F I G U R E 25-5 Base sequence of (a) the DNA of the E. coli trp operon at which transcription termination occurs and of (b) the 3' terminus of the mRNA molecule. The inverted-repeat sequence is indicated by reversed arrows. The mRNA molecule is folded to form a stem-and-loop structure.

After several nucleotides have been added to the growing chain, RNA polymerase holoenzyme changes its structure and loses the cr subunit. Thus, most elongation is carried out by the core enzyme, which moves along the DNA, binding a nucleoside triphosphate that can pair with the next DNA base and opening the DNA helix as it moves. The open region extends over about 30 base pairs. Chain elongation does not occur at a constant rate but slows down or stops at various points along the DNA molecule and, in some cases, may have a regulatory function. Termination of RNA synthesis occurs at specific base sequences within the DNA molecule. Many prokaryotic termination sequences have been determined and most have the following three characteristics (Figure 25-5): 1. An inverted-repeat base sequence containing a central nonrepeating segment; the sequence in one DNA strand would read ABCDEF-XYZ-F'E'D'C'B'A', in which A and A', B and B', and so on, are complementary bases. The RNA transcribed from this segment is capable of intrastrand base pairing, forming a stem and loop. 2. A sequence having a high G + C content. 3. A sequence of AT pairs in DNA (which may begin in the stem) that results in a sequence of 6-8 U's in the RNA. Termination of transcription includes the following steps:

568

CHAPTER 25

RNA and Protein Synthesis

F I G U R E 25-6 Transcription cycle of E. coli RNA polymerase showing dissociation of the c~ subunit shortly after chain elongation begins, dissociation of the core enzyme during termination, and re-formation of the holoenzyme from the core enzyme and the o- subunit. A previously joined core enzyme and a subunit will rarely become rejoined; instead, reassociation occurs at random.

1. Cessation of RNA elongation, 2. Release of newly formed RNA, and 3. Release of the RNA polymerase from the DNA. There are two kinds of termination events in prokaryotes: those that are self-terminating (dependent on the DNA base sequence only) and those that require the presence of a termination protein called Rho. Both types of events occur at specific but distinct base sequences. Rho, an oligomeric protein, does not bind to the core polymerase or to the holoenzyme and binds only very weakly to DNA. The action of Rho is poorly understood. Some microorganisms regulate transcription of certain genes by inhibiting Rho thereby allowing transcription to continue into adjacent genes, a process called antitermination. The transcription cycle of E. coli RNA polymerase is shown in Figure 25-6. Various drugs inhibit chain elongation. Cordycepin is converted to a 5'-triphosphate form and then acts as a substrate analogue, blocking chain elongation.

Lifetime of Prokaryotic mRNA All mRNA molecules are subject to attack by RNases, and this degradation is an essential aspect of the regulation of gene expression. Proteins are not made when they are not needed, and the rate of protein synthesis is determined by

a balance between the rates of RNA synthesis and RNA degradation. The half-life of a typical prokaryotic mRNA molecule is only a few minutes, so constant production of a bacterial protein requires continued transcription. In contrast, eukaryotic mRNA molecules have a lifetime of hours to days. Presumably, the reason for the difference is that bacteria must adapt to rapidly changing environments, whereas eukaryotic cells receive a constant supply of nutrients that maintain a uniform environment.

25.5 Transcription in Eukaryotes The chemistry of transcription in eukaryotes is the same as in prokaryotes. However, the promoter structure and the mechanism for initiation are strikingly different.

Eukaryotic RNA Polymerases Eukaryotic cells contain three classes of RNA polymerases, denoted I, II, and III, which are distinguished by their requirements for particular ions and by their sensitivity to various toxins. All are found in the nucleus. Minor RNA polymerases are found in mitochondria and chloroplasts. Polymerase I molecules are located in the nucleolus and are responsible for synthesis of 5.8S, 18S, and 28S rRNA molecules. Polymerase II synthesizes all

SECTION 25.5

Transcription in Eukaryotes

569

F I G U R E 25-7 Sequences found in and near some RNA polymerase II promoters. Only the TATA box is represented in many promoters (some promotors lack this sequence). The CAAT box occurs much less frequently, and the GC box has only occasionally been observed. Upstream sites are very common but are not considered to be part of the promoter.

RNA molecules destined to become mRNA. Polymerase III synthesizes 5S rRNA and the tRNAs. Polymerases II and III are inhibited by ot-amanitin, the toxic product of Amanita phalloides mushrooms, and are identified by their sensitivity to this substance. Rifampin, a powerful inhibitor of bacterial RNA polymerase, is inactive against the eukaryotic nuclear polymerases but inhibits mitochondrial RNA polymerases, although at higher concentrations than needed to inhibit bacterial polymerases. Thus, rifampin is used with other drugs (e.g., isoniazid) in treating tuberculosis. Actinomycin D is a general inhibitor of eukaryotic transcription by virtue of its binding to DNA. It has been useful in the treatment of childhood neoplasms (e.g., Wilms' tumor) and choriocarcinoma. However, it inhibits rapidly proliferating cells of both normal and neoplastic origin and hence produces toxic side effects.

RNA Polymerase II Promoters The structure of eukaryotic promoters is more complex than that of prokaryotic promoters. DNA sequences, hundreds of base pairs (bp) upstream from the transcription start site, control the rate of initiation. Furthermore, initiation requires numerous specific proteins (transcription factors) that bind to particular DNA sequences. Without the transcription factors, RNA polymerase II cannot bind to a promoter. However, KNH polymerase II itself is not a transcription factor. The complexity of initiation may derive, in part, from the fact that eukaryotic DNA is in the form of chromatin, which is inaccessible to RNA polymerases. Many RNA polymerase II promoters have the following features: 1. A sequence, TATAAAT, about 25 bp upstream from the transcription start site, known as the TATA or Hogness box. (Note its similarity to the Pribnow box.) The TATA box probably determines the base that is first transcribed. 2. A common sequence in the-75 region, GG(T/C)CAATCT, in which T and C appear with equal frequency at the third position. This sequence is

called the CAAT box. A third element, recognized ina few promoters, is the GC box, GGGCGG (Figure 25-7). Note: The term "upstream" refers to regions in the DNA that are to the left (or 5') of the start of transcription of a gene; the term "downstream" refers to regions to the right (or 3') of the gene. Regulatory regions affecting gene expression may be located close to the transcriptional start site, thousands of bases or more upstream or downstream from the gene, or in introns.

RNA Polymerase III Promoters RNA polymerase III promoters differ significantly from RNA polymerase II promoters in that they are located downstream from the transcription start site and within the transcribed segment of the DNA. For example, in the 5S RNA gene of the South African toad (Xenopus laevis) the promoter is between 45 and 95 nucleotides downstream from the start point. Thus, the binding sites on RNA polymerase III are reversed with respect to the transcription direction, as compared with RNA polymerase II. That is, RNA polymerase II reaches forward to find the start point, and RNA polymerase III reaches backward. In fact, RNA polymerases can slide in either direction along a DNA template; however, they can only synthesize RNA molecules in a 5' --+ 3' direction.

Eukaryotic mRNA Synthesis Eukaryotic mRNA used in protein synthesis is usually about one-tenth the size of the primary transcript. This size reduction results from excision of noncoding sequences called intervening sequences or introns. After excision, the coding fragments are rejoined by RNA splicing enzymes (Figure 25-8).

Capping and Polyadenylylation Capping occurs shortly after initiation of synthesis of the mRNA and precedes other modifications that protect the mRNA from degradation by nucleases. The poly(A)

5~0

CHAPTER25

RNA and Protein Synthesis

FIGURE 25-8

Schematic drawing showing production of eukaryotic mRNA.The primarytranscript is capped before it is released. Then its T-OH end is modified, and finally the interveningregions are excised. MeG, 7-methylguanosine. **Two nucleotides whose riboses may be methylated.

terminus is synthesized by a nuclear enzyme, poly(A) polymerase. However, adenylate residues are not added directly to the 3' terminus of the primary transcript. Instead, RNA polymerase transcribes past the recognition site for addition of poly(A). After synthesis of the complete RNA, endonucleolytic cleavage occurs at the poly(A) recognition site in the RNA, and poly(A) is added. A sequence, AAUAAA, located 10-25 bases upstream from the poly(A) addition site, is also required for enzyme recognition of the site of polyadenylylation.

Splicing of RNA in Eukaryotes A distinguishing feature of most primary transcripts of higher eukaryotes is the presence of untranslated intervening sequences (introns) that interrupt the coding sequence and are excised from the primary RNA transcript. In the processing of RNA in higher eukaryotes, the amount of discarded RNA ranges from 30% to nearly 90% of the primary transcript. The remaining coding segments (exons) are joined together by splicing enzymes to form translatable mRNA molecules. The excision of the introns and the formation of the final mRNA molecule by joining of the exons is called RNA splicing. The 5' segment (the cap) of the primary transcript is never discarded and hence is always present in the completely processed mRNA molecule; the 3' segment is also usually retained. Thus, the number of exons is usually one more than the number of introns. The number of introns per gene varies considerably (Table 25-1). Furthermore, within different genes the introns are distributed differently and have many sizes (Figure 25-9), and introns are usually longer than exons. The splicing reaction is remarkably precise: cuts are made at unique positions in transcripts that contain

thousands of bases. The fidelity of the excision and splicing reaction is extraordinary, for if an error of even one base were made, the correct reading frame would be destroyed. Such fidelity is achieved by recognition of particular base sequences by splicing enzymes. Base sequence studies of the regions adjacent to several hundred different introns indicate that common sequences can be found at each end of an intron. The sites at which cutting occurs are always 5' to GU and 3' to AG. The rule is that the base sequence of an intron begins with GU and ends with AG. Introns are excised one by one, and ligation occurs before the next intron is excised; thus, the number of different nuclear RNA molecules present at any instant is huge. Translation does not occur until processing is complete.

TABI,E 25-1

Translated Eukaryotic Genes in Which Introns Have Been Demonstrated Gene

a-Globulin Immunoglobulin L chain Immunoglobulin H chain Yeast mitochondrial cytochrome b Ovomucoid Ovalbumin Ovotransferrin Conalbumin a-Collagen

N u m b e r of lntrons

2 2 4 6 6 7 16 17 52

*At present the histone and interferon genes are the only known translated genes in the higher organisms that do not contain introns.

571

SECTION25.6 6enetic Code

F I G U R E 25-9 Diagram of the conalbumin primary transcript and the processed mRNA. The 16 introns, which are excised from the primary transcript, are shown in color.

Before RNA splicing was discovered, the nucleus was observed to contain a significant amount of seemingly untranslated RNA. The collection of RNA molecules of widely varied sizes was given the name heterogeneous nuclear RNA (hnRNAL a term that is still sometimes used for nuclear RNA.

Splicing and Ribozymes Different snRNPs are found in eukaryotic cells which function in removing introns from primary RNA transcripts. The association of small RNAs, nuclear proteins, and the introns that they attach to is referred to as a spliceosome. Small nuclear RNAs (designated U1 through U6) provide specificity to different spliceosomes so that they recognize different classes of introns. Introns are distinguished according to their three-dimensional structures and each class of introns is spliced out by a different mechanism. Class I introns were originally discovered in ciliated protozoa and subsequently were found in fungi, bacteriophages, and other organisms. The RNA itself in a class I intron has catalytic activity and class I introns remove themselves from primary RNA transcripts by a self-splicing reaction. Class I introns are not true enzymes in that they function only once. The nucleotides in the intron that is spliced out are recycled in the cell. Class H introns are removed from RNA by a selfsplicing reaction that proceeds through an intermediate structure called a lariat. The removal of class II introns from RNAs also results in the splicing together of exons on either side of the intron. The ability of class II introns to specifically bind to a 5' exon has led to their use as reagents to construct novel RNA molecules. Chemical derivatives of class II introns have been constructed that can carry out the reverse of the splicing reaction. When these introns insert themselves into RNA, they can be used to shuffle sequences or to link one RNA molecule to another. Class II introns, which are found in bacteria, plant organelles, yeast, and fungi, also are capable of reintegrating into DNA after being excised from an RNA molecule. Class II introns encode a multifunctional intron-encoded

protein (IEP) that has reverse transcriptase activity, RNA splicing activity, and DNA endonuclease activity. These three enzymatic activities allow the intron to be excised from RNA, copied into an RNA-DNA heteroduplex, and inserted into DNA at specific target sites recognized by the IEP-intron complex. Recently, class II introns have been modified so that they can be targeted to specific genes in any DNA molecule. This novel molecular mechanism raises the possibility that suitably engineered class II introns can be used therapeutically in gene therapy and possibly to inactivate viruses such as HIV by integration and disruption of essential genes. Ribonuclease P (RNase P) consists of both protein and an RNA component that has catalytic activity. RNase P functions in eukaryotic cells to process the 5' end of precursor tRNA molecules. RNase P also can be directed to cleave any RNA molecule when the target is complexed with a short complementary oligonucleotide called an external guide. Ribozymes refer to catalytic RNA molecules that recognize specific target sequences in other RNA molecules. This activity and specificity makes ribozymes potentially useful therapeutic agents. For example, a ribozyme could be directed to silence the expression of a deleterious gene by destroying the mRNA transcript before it can be expressed. Or ribozymes might be constructed that would inactivate the mRNA or oncogenes or the expression of genes in RNA viruses such as HIV. Laboratory studies have supported the feasibility of ribozyme therapy but its application in clinical practice is still far in the future.

25.6 Genetic Code "Universal" Genetic Code Production of proteins from mRNA requires translation of the base sequence into an amino acid sequence. The collection of base sequences (codons) that correspond to each amino acid and to signals for termination of translation is the genetic code. The code consists of 64 triplets of bases (Table 25-2). The codons are written with the 5' terminus

CHAPTER25 RNA and Protein Synthesis

57~

TABLE 25-2 "Universal" Genetic Code*

First Position (5' end)

Third Position (3' end)

Second Position U

C

A

G

the exception of serine, leucine, and arginine, all synonyms (codons corresponding to the same amino acid) are in the same box and differ by only the third base. For example, GGU, GGC, GGA, and GGG all code for glycine. The fidelity of translation is determined by two features of the system:

U

Phe Phe Leu Leu

Ser Ser Ser Ser

Tyr Tyr Stop Stop

Cys Cys Stop Trp

U C A G

C

Leu Leu Leu Leu

Pro Pro Pro Pro

His His Gln Gln

Arg Arg Arg Arg

U C A G

A

Ile Ile Ile IMetl

Thr Thr Thr Thr

Asn Asn Lys Lys

Ser Ser Arg Arg

U C A G

G

Val Val Val [Val[

Ala Ala Ala Ala

Asp Asp Glu Glu

Gly Gly Gly Gly

U C A G

*The boxed codons are used for initiation. GUG is very rare.

of the codon at the left. The following features of the code should be noted: 1. Sixty-one codons correspond to 20 different amino acids. 2. The codon AUG has two functions. It corresponds to the amino acid methionine when AUG occurs within a coding sequence in the mRNA, i.e., within a polypeptide chain. It also serves as a signal to initiate polypeptide synthesismwith methionine for eukaryotic cells but with N-formylmethionine for prokaryotic cells. How the protein-synthesizing system distinguishes an initiating AUG from an internal AUG is discussed below. The codon GUG also has both functions, but it is only rarely used in initiation. Once initiation has occurred at an AUG codon, the reading frame is established and the subsequent codons are translated in order. 3. Three codonsmUAA, UAG, and U G A m d o not represent any amino acid but serve as signals to terminate the growing polypeptide chain. 4. Except for methionine (AUG) and tryptophan (UGG), most amino acids are represented by more than one codon. The assignment of codons is not random; with

1. Attachment of the correct amino acid to a particular tRNA molecule by the corresponding aminoacyl synthetase, and 2. Correct codon-anticodon pairing. The former results from the specificity of interaction of the enzyme, the amino acid, and the tRNA molecule. The latter is assured by base pairing. A striking aspect of the code is that, with few exceptions, the third base in a codon appears to be unimportant, i.e., XYA, XYB, XYC, and XYD are usually synonymous. This finding has been explained by the base pairing properties of the anticodon-codon interaction. In DNA, no base pairs other than GC and AT are possible because the regular helical structure of double-stranded DNA imposes steric constraints. However, since the anticodon is located within a single-stranded RNA loop, the codon-anticodon interaction does not require formation of a structure with the usual dimensions of a double helix. Model building indicates that the steric requirements are less stringent at the third position of the codon, a feature called wobble. The wobble hypothesis allows for inosine (I), a nucleoside in which the base is hypoxanthine and which is often found in anticodons, to base-pair with A, U, or C. In addition, the wobble hypothesis allows for U to base-pair with G. This explains how a tRNA molecule carrying a particular amino acid can respond to several different codons. For example, two major species of yeast tRNA Ala are known; one responds to the codons GCU, GCC, and GCA and has the anticodon IGC. Recall that the convention for naming the codon and the anticodon always has the 5' end at the left. Thus, the codon 5'-GCU-3' is matched by the anticodon 5'-IGC-3'. The other tRNA Ala has the anticodon CGC and responds only to GCG. The function of three stop codons derives from the fact that no tRNA exists that has an anticodon that can pair with the stop codons. Genetic Code of Mitochondria The genetic code of mitochondria is not the same as the "universal" code, which has implications for the evolution of mitochondria. Human mitochondria contain a set of tRNA molecules that are not found elsewhere in the

SECTION25.6 Genetic Code

573

TABLE 25-3

Genetic Code of Human Mitochondria

First Position (5' end)

Third Position (3' end)

Second Position U

C

A

G

U

Phe Phe Leu Leu

Ser Ser Ser Ser

Tyr Tyr Stop Stop

Cys Cys Trp Trp*

U C A G

C

Leu Leu Leu Leu

Pro Pro Pro Pro

His His Gln Gln

Arg Arg Arg Arg

U C A G

A

IIle I Ile IMet~ IMetl

Thr Thr Thr Thr

Asn Asn Lys Lys

Ser Ser Stop* Stop*

U C A G

G

Val Val Val Val

Ala Ala Ala Ala

Asp Asp Glu Glu

Gly Gly Gly Gly

U C A G

*These entries are found in mitochondriabut not in the univeral code. Boxed codons are used as start codons. The mitochondrial codes of other organisms exhibit further differences.

cell and a circular DNA molecule containing 16,569 base pairs. This DNA molecule encodes some mitochondrial enzymes (Chapter 14) and is the template for synthesis of all mitochondrial tRNA and rRNA molecules. Human mitochondrial DNA sequences contain the genes for 12S and 16S ribosomal RNA, 22 different tRNA molecules, three subunits of the enzyme cytochrome oxidase (whose amino acid sequence is known), cytochrome b, and several other enzymes. The human mitochondrial code is shown in Table 25-3; entries shown with an asterisk differ from the universal code (cf. Table 25-2). The differences are striking in that most are in the initiation and termination codons. That is, in mammalian mitochondria, 1. UGA codes for tryptophan and not for termination. 2. AGA and AGG are termination codons rather than codons for arginine. 3. AUA and AUU are initiation codons, as is AUG. Both AUA and AUG also code for methionine. AUU also codes for isoleucine, as in the universal code. 4. AUA codes for methionine (and initiation, as shown in item 3) instead of isoleucine.

Maize mitochondria use CGG for tryptophan rather than for arginine, and CGU, CGC, and CGA for arginine. Yeast mitochondria use CUX, where X is any base, for threonine rather than for leucine. Both maize and yeast use AGA and AGG for arginine. Evidently, various mitochondrial codes can differ from each other as well as from the universal code. The number of mammalian mitochondrial tRNA molecules is 22, which is less than the minimum number (32) needed to translate the universal code. This is possible because in each of the fourfold redundant sets--e.g., the four alanine codons GCU, GCC, GCA, and GCGNonly one tRNA molecule (rather than two, as explained above) is used. In each set of four tRNA molecules, the base in the wobble position of the anticodon is U or a modified U (not I). It is not yet known whether this U is base-paired in the codon-anticodon interaction or manages to pair weakly with each of the four possible bases. For those codon sets that are doubly redundant--e.g., the two histidine codons CAU and CAC--the wobble base always forms, a G-U pair, as in the universal code. The structure of the human mitrochondrial tRNA molecule is also different from that of the standard tRNA molecule (except for mitochondrial tRNALeuuux). (X = any nucleotide.) The most notable differences are the following: 1. The universal sequence CTgzCXA is lacking in mitochondrial tRNA. 2. The "constant" 7-bp sequence of the TgzC loop varies from three to nine bases. 3. The invariant bases U8, A14, G15, G18, G19, and U48 of the standard tRNA molecule are not invariant in mitochondrial tRNA. In standard tRNA molecules, each of these bases participates in bonds that produce the folded L-shaped molecule. Thus, the mitochondrial tRNA molecule seems to be stabilized by fewer interactions. The threedimensional configurations of these molecules are not known with certainty; possibly they differ from the standard L-shape, and mitochondrial tRNA engages in a different type of interaction with the ribosome than standard tRNA molecules do. Most DNA molecules contain long noncoding segments (spacers) between genes. In mitochondrial DNA, there are either a few bases or none. In each case, there is a start codon (AUG, AUA, or AUU) at the 5' end of the mRNA molecule or within a few bases of it. This arrangement differs from the usual arrangement for eukaryotes in that ordinarily eukaryotic mRNA molecules commence with a short leader sequence thought to be responsible for binding to the ribosome. Such a leader is not present in the mitochondrial mRNA molecules, which suggests that

574

CHAPTER25 RNA and Protein Synthesis

the 5' terminus itself of a mitochondrial mRNA molecule binds to the ribosome. The plethora of diseases and clinical abnormalities caused by mitochondrial mutations discussed in Chapter 14.

H tRNA fMet + Met

O

II

I (CH2)2 O

\\

p

25.7 A t t a c h m e n t of Amino Acid to t R N A M o l e c u l e

I

Methionyl-tRNAsynthetase~ H 3 N ~ C ~ C ~ O ~ t R N A f M e t

H

H

I

~N~

H

I

S

O

i,

C~C~O~tRNA

I

(CH2)2

tMet

Transformylase

THF

J

fTHF

I

S

I

CH3

When an amino acid is covalently linked to a tRNA molecule, the tRNA is said to be aminoacylated or charged. The notation for a tRNA charged with serine is seryl tRNA. The term uncharged tRNA refers to a tRNA molecule lacking an amino acid. Acylation is accomplished in two steps, both of which are catalyzed by an aminoaeyl tRNA synthetase. In the first, or activation, step an aminoacyl AMP is generated in a reaction between an amino acid and ATP: H

I

O

II

H3N + ~ C ~ C ~ O -

I

R

H + ATP ~

I

O

O

II

II

H3N + ~ C ~ C ~ O ~ P ~ R i b o s e ~ A d e n i n e

I

R

I

+ PP

O-

In the second, or transfer, step, the aminoacyl AMP reacts with tRNA to form an aminoacylated tRNA and AMP: Aminoacyl AMP + tRNA ~ aminoacyl tRNA + AMP As in DNA synthesis, the reaction is driven to the right by hydrolysis of pyrophosphate, so the overall reaction is Amino acid + ATP § tRNA + H20 -+ aminoacyl tRNA § AMP § 2Pi Usually only one aminoacyl synthetase exists for each amino acid. However, for a few amino acids specified by more than one codon, more than one synthetase does exist.

25.8 Initiator t R N A M o l e c u l e s Initiation Codon

and Selection of

The initiator tRNA molecule in prokaryotes--tRNA tMetm has several properties that distinguish it from all other tRNA molecules. One feature is that the tRNA is first acylated with methionine, and then the methionine is modified. Acylation is by methionyl tRNA synthetase, which also charges tRNA Met. However, the methionine of charged tRNA fMet is immediately recognized by another enzyme, tRNA methionyl transformylase, which transfers a formyl group from Nl~ (fTHF) to the amino group of the methionine to form

N-formylmethionine (fMet): Transformylase does not formylate methionyl tRNA Met because tRNA Met and tRNA fMet are structurally different. A second feature is its ability to initiate polypeptide synthesis. Eukaryotic initiator tRNA molecules differ from the prokaryotic initiator molecule in several ways. The most striking difference is that whereas eukaryotic organisms produce both a normal tRNA Met and an initiator tRNA, which is also charged with methionine, the methionine does not undergo formylation. In eukaryotes, the first amino acid in a growing polypeptide chain is Met and not fMet. The codon for both kinds of tRNA molecules in eukaryotes is AUG, just as for prokaryotes. The sequence AUG serves as both an initiation codon and the codon for methionine. Since methionine occurs within protein chains, some signal in the base sequence of the mRNA must identify particular AUG triplets as start codons. In eukaryotes, initiation usually begins at the AUG triplet nearest to the 5' terminus of the mRNA molecule, i.e., no particular signal is used (although sequences do play a role in the efficiency of initiation). Presumably, the ribosome and the mRNA interact at the 5' terminus and slide with respect to one another until an AUG is encountered. This AUG determines the reading frame. When a stop codon is encountered, the ribosome and the mRNA dissociate. Only a unique polypeptide is translated from a particular eukaryotic mRNA molecule. In prokaryotes, the situation is quite different. At a fixed distance upstream from each AUG codon used for initiation is the sequence AGGAGGU (or a single-base variant). This sequence, known as the Shine-Dalgarno sequence, is complementary to a portion of the 3'-terminal sequence of one of the RNA molecules in the ribosome (in particular, the 16S rRNA in the 30S particle; see below). Base pairing between these complementary sequences orients the initiating AUG codon on the ribosome. Thus, initiation in prokaryotes is restricted to an AUG on the 3' side of the Shine-Dalgarno sequence. Many prokaryotic mRNA molecules are polycistronic and contain coding sequences for several polypeptides. Thus, a polycistronic mRNA molecule must possess a

575

SECTION25.9 Ribosomes

series of start and stop codons for each polypeptide. The mechanism for initiating synthesis of the first protein molecule in a polycistronic mRNA is the same as that in a monocistronic mRNA. However, if a second protein in a polycistronic mRNA is to be made, protein synthesis must reinitiate after termination of the first protein. This is usually accomplished by the start codon of the second protein being so near the preceding stop codon that reinitiation occurs before the ribosome and the mRNA dissociate. Otherwise, a second initiation signal is needed.

25.9 Ribosomes A ribosome is a multicomponent structure that serves to bring together a single mRNA molecule and charged tRNA molecules so that the base sequence of the mRNA molecule is translated into an amino acid sequence. The ribosome also contains several enzymatic activities needed for protein synthesis. The protein composition of the prokaryotic ribosome is well known, but less is known about the composition of eukaryotic ribosomes. However, both share the same general structure and organization. The proper ties of the bacterial ribosomes are constant over a wide range of species, and the E. coli ribosome has been analyzed in great detail and serves as a model for all ribosomes.

Chemical Composition of Prokaryotic Ribosomes A prokaryotic ribosome consists of two subunits. The intact particle is called a 70S ribosome because of its S (sedimentation) value of 70 Svedberg units. The subunits, which are unequal in size and composition, are termed 30S

and 50S subunits. The 70S ribosome is the form active in protein synthesis. At low concentrations of Mg 2+, ribosomes dissociate into ribosomal subunits. At even lower concentrations, the subunits dissociate, releasing indi vidual RNA and protein molecules. The composition of each subunit is as follows: 30S subunit : one 16S r R N A molecule + 21 different proteins 50S subunit : one 5S r R N A molecule + one 23S r R N A molecule + 34 different proteins

The proteins of the 30S subunit are termed S 1, $2 . . . . . $21 (in this context, S is for "small" subunit); one copy of each is contained in the 30S subunit. The proteins of the 50S subunit are denoted by L (for "large" subunit). Each 50S subunit contains one copy of each protein molecule, except for proteins L7 and L12, of which there are four copies, and L26, which is not considered to be a true component of the 50S subunit. Most ribosomal proteins are very basic proteins, containing up to 34% basic amino acids. This basicity probably accounts in part for their strong association with the RNA, which is acidic. The amino acid sequences of most of the E. coli ribosomal proteins are known. A summary of the structure of the E. coli 70S ribosome is shown in Figure 25-10.

Ribosomes Are Ribozymes Ever since the discovery of the role of ribosomes in protein synthesis there has been uncertainty as to the functional roles of the rRNAs and the many ribosomal proteins. Conventional wisdom held that the rRNAs provided the scafolding (structural role) that oriented the ribosomal proteins so that one or more could carry out the enzymatic

FIGURE 25-10 Dissociation of a prokaryoticribosome.The configurationof two overlappingcircles is used throughoutthis chapter, for the sake of simplicity.

576

CHAPTER25 RNA and Protein

reactions necessary for protein synthesis. It was not until crystallization of the 50S subunit provided an atomic-level view of its structure that it was finally proved that the opposite is actually correct. The ribosomal proteins provide the structural framework for the 23S which actually carries out the peptidyltransferase reaction. Thus, the 23S rRNA is an enzyme and the ribosome is a ribozyme. Although the activity of the 23S rRNA is dependent on the ribosomal proteins, the 50S subunit that is substantially deproteinized still can perform the peptidyltransferase reaction. A crucial feature of the peptidyltransferase reaction is a particular adenine that is conserved in rRNAs extracted from the large ribosomal subunits of thousands of different organisms from all three kingdoms. Sequence analysis shows that this adenine base (2451A) always is present in the active site of the ribosome and acts as a general acidbase catalyst by deprotonating the nucleophilic amine as shown below. ...

2451A

\

25.10 Protein Synthesis Protein synthesis has three stages" 1. Polypeptide chain initiation, 2. Chain elongation, and 3. Chain termination. The main features of the initiation step are binding of mRNA to the ribosome, selection of the initiation codon, and binding of the charged tRNA bearing the first amino acid. In the elongation stage, there are two steps: joining of adjacent amino acids by peptide bond formation, and moving the mRNA and the ribosome with respect to one another so that codons are translated successively (translocation). In the termination stage, the completed polypeptide dissociate from the ribosomes, which are released to begin another cycle of synthesis.

l

.,.

i

,H aa-tRNA--=N:/""~==O

Synthesis

aa-tRNA----N HO 2451A ~tRNA

tRNA m

..,

tetrahedml

intermediate

The positive charge on 2451A in the tetrahedral intermediate is probably transferred to adjacent tRNA nucleotides that undergo tautomeric shifts. The evidence that ribosomal RNAs perform enzymatic functions critical to protein synthesis lends weight to the evolutionary significance of RNA in primordial chemical reactions that eventually led to the development of cells.

Every polypeptide has an amino terminus and a carboxyl terminus. In both prokaryotes and eukaryotes, synthesis begins at the amino terminus. For a protein having the sequence H 2 N - M e t - T r p - A s p . . . Pro-Val-COOH, the fMet (or Met) is the initiating amino acid and Val is the last amino acid added to the chain. Translation of mRNA molecules occurs in the 5' --+ 3' direction.

Chemical Composition of Eukaryotic Ribosomes

Stages of Protein Synthesis

The basic features of eukaryotic ribosomes are similar to those of bacterial ribosomes, except for their larger size. They contain a greater number of proteins (about 80) and an additional RNA molecule. A typical eukaryotic ribosome has a sedimentation coefficient of about 80 and consists of two subunits: 40S and 60S. These sizes may vary by as much as 10% from one organism to another in contrast with the homogeneity of bacteria ribosomes. The components of the subunits of eukaryotic ribosomes are as follows:

The mechanismsof protein synthesis in prokaryotes and eukaryotes differ slightly in detail, but the prokaryotic mechanism is used as a general model"

40S subunit 9one 18S rRNA molecule + 30-35 proteins 60S subunit 9one 5S, one 5.8S, and one 28S rRNA molecule + 45-50 proteins

1. Initiation. Protein synthesis in bacteria begins by the association of one 30S subunit (not the 70S ribosome), an mRNA, a charged tRNA fMet, three protein initiation factors, and guanosine 5'-triphosphate (GTP). These molecules make up the 30S preinitiation complex. Association occurs at an initiator AUG codon, whose selection was described above. A 50S subunit joins to the 30S subunit to form a 70S initiation complex (Figure 25-11). This joining process requires hydrolysis of the GTP contained in the 30S preinitiation complex. There are two tRNA

577

SECTION25.10 Protein Synthesis mRNA + 30S subunit + fMet-tRNA fMet + GTP + initiation factors

1 mRNA

I

fMet

50S subunit

L

P site

A site

GDP + P, + initiation factors 70S initiation complex

F I G U R E 25-11 Early steps in protein synthesis: in prokaryotes: formation of the 30S preinitiation complex and 70S initiation complex.

binding sites on the 50S subunit: the aminoacyl(A) site and the peptidyl(P) site; each site consists of several L proteins and 23S rRNA. The 50S subunit is positioned in the 70S initiation complex such that the tRNA fMet, which was previously bound to the 30S preinitiation complex, occupies the P site of the 50S subunit. Positioning tRNA fMet in the P site fixes the position of the anticodon of tRNA fMet so that it can pair with the initiator codon in the mRNA. in this way, the reading frame is unambiguously defined. 2. Binding of a second tRNA to the A site. The A site of the 70S initiation complex is available to any tRNA molecule whose anticodon can pair with the codon adjacent to the AUG complex initiation codon. However, entry to the A site by the tRNA requires the activity of a protein called elongation factor EF-Tu (Figure 25-12). EF-Tu binds GTP and an aminoacyl tRAA, forming a ternary complex, aminoacyl tRNA [EF-Tu-GTP]. This ternary complex enters the A site for the codon-dependent placement of the aminoacyl tRNA at the A site. During this process of placement, GTP is hydrolyzed to GDP and Pi, and EF-Tu is released. EF-Tu-GTP is regenerated from EF-Tu-GDP through the inter action of another protein, EF-Ts, and GTP. 3. Chain elongation (formation of the first peptide bond). After a charged tRNA is positioned in the A site, a peptide bond between fMet and the adjacent amino acid forms. The peptide bond is formed by an enzyme complex called peptidyltransferase, whose active site resides in 23SrRNA of 50S subunit as previously discussed (Figure 25-13). As the peptide bond is formed, the Met is cleaved from the tRNA fMet

in the P site by another ribosomal protein, tRNA deacylase. 4. Translocation of the ribosome. After the peptide bond forms, an uncharged tRNA occupies the P site and a dipeptidyl tRNA is in the A site. At this point three events, which together make up the translocation step, occur: the deacylated tRNA fMet leaves the P site, the peptidyl tRNA moves from the A site to the P site, and the ribosome moves a distance of three bases on the mRNA to position the next codon at the A site. The translocation step requires the activity of another elongation protein, EF-G, and hydrolysis of GTP to provide the energy to move the ribosome. Thus, the total amount of energy expended in the synthesis of one peptide bond comes from the hydrolysis of four high-energy phosphate bonds (equal to about 30 kcal = 7.5 • 4). Recall that the synthesis of each molecule of ammoacyl tRNA consumes two high-energy phosphate bonds (ATP --+ AMP + PPi; PPi + 2Pi). (In these calculations, the one GTP molecule expended in the formation of the initiation complex is not included because of its small contribution to the overall synthesis of a polypeptide.) 5. Refilling of the A site. After translocation has occurred, the A site is again available for a charged tRNA molecule with a correct anticodon. When this occurs, the series of elongation reactions is repeated. 6. Termination. When a chain termination codon is reached, no aminoacyl tRNA is available that can fill the A site, so chain elongation stops. Since the polypeptide chain is still attached to the tRNA occupying the P site, release of the protein is

5~8

CHAPTER25 RNA and Protein Synthesis

FIGURE 25-12 Elongation phase of protein synthesis: binding of charged tRNA, peptide bond formation, and translocation. Note that this phase consumes 2 GTP during each cycle of chain elongation.

accomplished by release factors (RFs), which are proteins that, in part, respond to chain termination codons. In the presence of release factors, peptidyltransferase catalyzes the reaction of the bound peptidyl moiety with water rather than with the free aminoacyl tRNA (Figure 25-14). Thus, the

polypeptide chain, which has been held in the ribosome solely by the interaction with the tRNA in the P site, is released from the ribosome. 7. Dissociation of the 70S ribosome. The 70S ribosome dissociates into 30S and 50S subunits, which may start synthesis of another polypeptide chain.

FIGURE 25-13 Peptide bond formation in prokaryotes. In eukaryotes, the chemistryis the same, although the initiating tRNA is not tRNAfMetbut tRNAinit Met 9

SECTION25.10 Protein Synthesis

579

Peptidyltransferase

OH +

O II H2N~Ala~Phe~His~Thr~C~OH

+ Free 70S ribosome

I Rapid dissociation

30S subunit + 50S subunit

FIGURE 25-14

The steps of termination of protein synthesis. Role of GTP GTR like ATE is an energy-rich molecule. Generally, when such molecules are hydrolyzed, the free energy of hydrolysis is used to drive reactions that otherwise are energetically unfavorable. This does not seem to be the case in protein synthesis. The reaction sequence indicates that GTP facilitates binding of protein factors either to tRNA or to the ribosome. Furthermore, hydrolysis of GTP to GDP always precedes dissociation of the bound factor. Comparison of the structure of the free factor and the factor-GTP complex indicates that the factor undergoes a slight change in conformation when GTP is bound. The function of GTP is to induce a conformational change in a macromolecule by binding to it. Since it is easily hydrolyzed by various GTPases, the use of GTP as a controlling element allows cyclic variation in macromolecular shape. When GTP is bound, the macromolecule has an active conformation, and when the GTP is hydrolyzed or removed, the molecule resumes its inactive form. GTP plays a similar role in hormone activation systems (Chapter 30).

Posttranslational Modification of Proteins The protein molecule ultimately needed by a cell often differs from the polypeptide chain synthesized. Modification of the synthesized chain occurs in several ways: 1. In prokaryotes, fMet is never retained as the NH2, terminal amino acid. In roughly half of all proteins, the formyl group is removed by the enzyme deformylase, leaving methionine as the NH2 terminal amino acid. In both prokaryotes and eukaryotes, the fMet, methionine, and possibly a few more amino

acids, are often removed; their removal is catalyzed by a hydrolytic enzyme called aminopeptidase. This hydrolysis may occur as the chain is being synthesized or after the chain is released from the ribosome. The choice of deformylation versus removal of fMet usually depends on the identity of the adjacent amino acids. Deformylation predominates if the second amino acid is arginine, asparagine, aspartic acid, glutamic acid, isoleucine, or lysine, whereas f Met is usually removed if the adjacent amino acid is alanine, glycine, proline, threonine, or valine. 2. Newly created NH2 terminal amino acids are sometimes acetylated, and amino acid side chains may also be modified. For example, in collagen a large fraction of the prolines and lysines are hydroxylated (see below). Phosphorylation of serine, tyrosine, and threonine occurs in many organisms. Various sugars may be attached to the free hydroxyl group of serine or threonine to form glycoproteins. Finally, a variety of prosthetic groups such as heme and biotin are covalently attached to some proteins. 3. Two distant sulfhydryl groups in two cysteines may be oxidized to form a disulfide bond. 4. Polypeptide chains may be cleaved at specific sites. For instance, chymotrypsinogen is converted to the digestive enzyme chymotrypsin by removal of four amino acids from two different sites. In some cases, the uncleaved chain represents a storage form of the protein that can be cleaved to generate the active protein when needed. This is true of many mammalian digestive enzymes, e.g., pepsin is formed by cleavage of pepsinogen. An interesting precursor is a huge protein synthesized in animal cells infected

CHAPTER25 RNA and Protein Synthesis

580

F I G U R E 25-15 Polysomes. (a) Electron micrograph of an E. coli polysome. (Courtesy of Barbara Hamkalo.) (b) Diagram showing relative movement of the 70S ribosome and the mRNA and growth of the protein chain.

with some viruses; this molecule is cleaved at several sites to yield different active proteins and hence is called a polyprotein. Some cellular enzymatic systems are also formed by cleavage of polyproteins. C o u p l e d Transcription and Translation After about 25 amino acids have been joined together in a polypeptide chain in prokaryotes, the AUG initiation site of the mRNA molecule becomes exposed and a second initiation complex then forms. The overall configuration is of two 70S ribosomes moving along the mRNA at the same rate. When the second ribosome has moved a distance similar to that traversed by the first, a third ribosome is able to attach. This processmmovement and reinitiationmcontinues until the mRNA is covered with ribosomes separated by about 80 nucleotides. This large translation unit is called a polyribosome or polysome (Figure 25-15). Polysomes greatly increase the overall rate of protein synthesis, since 10 ribosomes traversing a single segment of mRNA clearly can make 10 times as many polypeptides per unit of time as can a single ribosome. In prokaryotes, transcription and translation are coupled; this can not occur in eukaryotes since transcription occurs in the nucleus and translation in the cytoplasm.

Endoplasmic Reticulum In most eukaryotic cells two major classes of ribosomes exist: attached ribosomes and free ribosomes. The attached ribosomes are bound to an extensive cytoplasmic network of lipoprotein membranes called the endoplasmic reticulum. The rough endoplasmic reticulum consists of bound ribosomes; the smooth endoplasmic reticulum is devoid of ribosomes. There is no structural difference between a

free and an attached ribosome, and attachment to the membrane occurs after synthesis of particular proteins begins. Most endoplasmic reticulum membranes enclose large, irregularly shaped, discrete regions of the cell called cisternae. In this sense, the membrane system has an inside and an outside, and ribosomes are bound only to the outside. Cells responsible for secreting large amounts of a particular protein (e.g., hormone-secreting cells) have an extensive endoplasmic reticulum. Most proteins destined to be secreted by the cell or to be stored in intracellular vesicles such as lysosomes (which contain degradative enzymes) and peroxisomes (which contain enzymes for eliminating hydrogen peroxide) are synthesized by attached ribosomes. These proteins are primarily found in the cisternae of the endoplasmic reticulum. In contrast, most proteins destined to be free in the cytoplasm are made on free ribosomes. The formation of the rough endoplasmic reticulum and the secretion of newly synthesized proteins through membrane is explained by the signal hypothesis (Figure 25-16). The basic idea is that the signal for attachment of the ribosome to the membrane is a sequence of very hydrophobic amino acids near the amino terminus of the growing polypeptide chain. When protein synthesis begins, the ribosome is free. Then the amino terminal hydrophobic amino acids interact with lipophilic membrane components and somehow direct the association of the large ribosomal subunit to a ribosomal receptor protein on the membrane surface. As protein synthesis continues, the protein moves through the membrane to the cisternal side of the endoplasmic reticulum. A specific protease termed the signalpeptidase cleaves the amino terminal signal sequence. Intracellular compartmentation of the newly synthesized proteins is a complex process, and disorders in this process lead to severe abnormalities (see below).

SECTION25.10 Protein Synthesis

581

F I G U R E 25-16 Signal hypothesis for the synthesis of secretory and membrane proteins. Shortly after initiation of protein synthesis, the amino-terminal sequence of the polypeptide chain binds a signal recognition protein, which then binds to a docking protein. The signal peptide is released from the signal recognition protein as the ribosome binds to a ribosome receptor, which is adjacent to a pore. Translation continues with the signal peptide passing through the pore. Once through the endoplasmic reticulum, the signal sequence is excised by the signal peptidase within the vesicle. When protein synthesis is completed, the protein remains within the vesicle and the ribosome is released.

In summary, the following events take place. When the predominant portion of the protein is synthesized and migrates in the endoplasmic reticulum, the signal sequence is removed by proteolytic cleavage. The finished protein, which is sequestered on the cisternal side of the endoplasmic reticulum, undergoes many posttranslational modifications, one of which is the addition of a series of carbohydrate residues to form glycoproteins (Chapter 16). Glycoproteins are transported to the Golgi complex, where further modification of the carbohydrate residues occurs. The finished glycoproteins are then packaged into lysosomes, peroxisomes, or secretory vesicles. The last fuses with the plasma membrane, discharging its contents into the extracellular fluid. Defects in compartmentation processes may lead to mislocation of the proteins and cause deleterious effects. Many proteins that are destined for various organelles (e.g., mitochondria) are not routed through endoplasmic reticulum and the Golgi complex. These proteins are made on cytosolic polysomes. They possess presequences at amino terminal ends that target them to receptors on appropriate organelles. Import of proteins into cell organelles is aided by chaperone proteins (Chapter 4).

Compartment Disorders Two defects of compartmentation are known. One, mucolipidosis or I-cell disease (Chapter 16), is characterized by mislocalization of lysosomal enzymes (acid hydrolases).

These glycoproteins lack mannose-6-phosphate residues, and consequently they are secreted into extracellular fluids instead of being sequestered into lysosomal vesicles. The mannose-6-phosphate serves as a marker that allows the enzymes to bind with mannose-6-phosphate receptors that direct the enzymes into the vesicles to be packaged into lysosomes. The lysosomal enzymes in the extracellular fluid bring about indiscriminate destruction of tissues. The second example of the compartmentation defect is ~ 1-antitrypsin deficiency (also known as o~1-proteinase inhibitor deficiency). The defect in this disorder is almost exactly the opposite of that in I-cell disease, i.e., I-cell disease is a disorder of a defect in (intracellular) retention of proteins and o~1-antitrypsin deficiency is a defect in the secretion of a protein, ot1-Antitrypsin (ol 1-AT) consists of a single polypeptide chain of 394 amino acid residues with three oligosaccharide side chains (Figure 25-17), all of which are attached to asparagine residues. It contains three/~ sheets and eight o~ helices. It has an overall molecular weight of 51,000. It is a polar molecule and readily migrates into tissue fluids. It is synthesized in hepatocytes and secreted into the plasma, where it has a half-life of 6 days. Its normal serum concentration is 150-200 mg/dL. Although the presence of O~l-AT has been noted in other cell types, the primary source in plasma is the hepatocyte. O~l-AT does not possess a propeptide but does contain a 24-residue signal peptide, which is eliminated during its passage through the endoplasmic reticulum, ot 1-AT is a defense protein, and its synthesis and release into circulation

582

CHAPTER25 RNA and Protein Synthesis

F I G U R E 25-17 Schematic representation of the amino acid sequence of c~l-antitrypsin (PiMM) and some of its variants. The amino acid residues shown are not drawn to scale; CHO represents the oligosaccharide units.

increase in response to trauma and inflammatory stimulus. Thus, it is known as an acute-phase reactant, ot I-AT functions by forming a tight complex with the active site of the target enzyme that inhibits its proteolytic activity. The enzyme-inhibitor complexes are rapidly cleared by the reticuloendothelial cells. Thus, or l-AT is a suicide protein. Many genetic variants of ot ~-AT have been identified by isoelectric focusing. The ot I-AT gene is on chromosome 14, is 10.2 kb long, and contains four introns. The phenotype is based on a codominant pattern with both alleles expressed equally. Variants are classified by a system known as Pi (protease inhibitor). The most common form is PiMM (carried by 95% of the U.S. population). Most polymorphism observed in ot i -AT are due to single-aminoacid substitutions. o~m-AT is a broad-spectrum serine proteinase inhibitor (Chapter 6). Its principal action is to inhibit leukocyte (neutrophil) elastase. This function appears to be most important in maintaining the integrity of the elastic fibers for the elastic recoil of normal lung tissue. Recall that elastic fibers contain an amorphous core of elastin surrounded by an envelope of microfibrils (Chapter 16). The target substrate for neutrophil elastases is elastin. The turnover rate of mature elastin is extremely low, and if it is destroyed, replacement is severely limited. Thus, the risk of development of a lung disease (emphysema) characterized by dilatation of air spaces distal to the terminal bronchiole and destruction of bronchiole walls is high in c~l-AT deficiency. A human phenotype in which a severe deficiency of ot 1-AT (10-15% of normal serum concentration) has been observed is designated PiZ. The risk of development of emphysema in PiZZ (homozygotic ZZ individuals) is 20 times that in PiMM (a normal genotype). In addition, cigarette smoking by PiZ individuals accelerates development of destructive lung disease for several reasons.

1. The levels of c~l-AT are very low, and smoking causes an increase in elastolytic load owing to change in the phagocytic population. 2. A reduction of the proteinase inhibitory activity is due to oxidation of the reactive methionine residue of the active center by the cigarette smoke as well as by oxygen radicals produced by leukocytes and macrophages. Emphysema is a common disease, and its most common cause is cigarette smoking. Only 1-2% of cases are due to genetic deficiency of ot1-AT. The substitution of a lysine for a glutamic acid at position 342 leads to the deficiency of c~I-AT in PiZ individuals (Figure 25-17). This substitution of a basic for an acidic amino acid is consistent with a base change of cytosine to thymine in DNA. The amino acid change prevents normal processing of oligosaccharide side chains of the protein and therefore its secretion. As a result, Z-or ~-AT accumulates in hepatocytes. An association of hepatic disease with severe c~l-AT deficiency has been observed; however, the relationship is not understood. The sequestered a ~-AT may cause damage to hepatocytes or render them more susceptible to injury, toxins, or viruses. Liver transplantation has been attempted with limited success in patients with hepatocellular failure. The therapy for destructive lung disease is administration of an anabolic steroid (danazol), which increases the serum levels of c~l-AT by about 40%, or direct replacement of c~I-AT by transfusion. Another structural variant of ot ~-AT that has been studied in detail is S-or I-AT (PISS). This protein differs from M-or i-AT at position 264, where a glutamic acid residue is substituted by valine (Figure 25-17). The S-ort-AT undergoes normal addition of oligosaccharides, and the protein is secreted into the blood. Affected individuals have sufficiently high levels of S-o~1-AT to protect against neutrophil elastase and so are not at higher risk for developing

583

SECTION25.10 Protein Synthesis C

NH

NH

II

.o

I/c",

II

H2NCNH

-o

NHCNH2 I

CH3

HO

|

O

H3C " ~ H3C\ /OH3

HO

N

I

CH ' 30

0

I

HO ~ 1 / 0 \ 1 CONH2 OH

O

OH

R ~--- C H 3 " " N H --"

O OH Binds to a specific site on the 23S RNA and blocks elongation by interfering with the translocation step.

OH Interferes with pairing between aminoacyl tRNAs and RNA codons, causing misreading.

Erythromycin-

Streptomycin:

Inhibits the binding of aminoacyl tRNAs to the ribosome in bacteria.

Tetracycline:

H3C\ /OH3

e

N

HN

k

OH

H \ /C-----O /C\. H3C--'O~CH

2

NH 3

Puromycin" Causes premature chain termination. The molecule resembles the 3' end of the aminoacylated tRNA and will enter the A site. It transfers to the growing chain causing premature termination. FIGURE 25-18 Antibiotic inhibitors of protein synthesis.

emphysema. Prenatal diagnosis of o~1-AT can be done by the use of either synthetic oligonucleotide probes or restriction fragment length polymorphism analysis. The reactive site of o~1-AT is at the methionine residue, position 358. The sequence around the reactive center of several proteinase inhibitors, including o~~-AT, antithrombin III, and several unrelated plant proteinase inhibitors, is strikingly similar. In fact, ot 1-AT, antithrombin III, and ovalbumin (a protein without known inhibitory function)

share extensive homologies and may all have evolved from a common ancestral protease inhibitor more than 500 million years ago. A fatal bleeding disorder due to substitution of a methionine for an arginine residue at position 358 of o~1-AT has been reported. This particular alteration brought a change in the specificity of the inhibitor. This altered protein (known as O~l-antitrypsin Pittsburgh) exhibits antithrombin activity while losing antielastase activity. Antithrombin III, which has an important regulatory

58~

role in hemostasis (Chapter 36), has a reactive arginine at its active center. Since the o~I-AT and antithrombin III are structurally similar, the substitution of methionine of ot 1-AT for arginine at the active center alters its specificity. This bleeding disorder is further complicated by the fact that the concentration of normal antithrombin III does not increase in response to inflammatory stress as does the level of ot 1-AT. Therefore, with each hemolytic episode, the abnormal o~I-AT increases, creating a cycle resulting in uncontrolled bleeding.

Inhibitors of Protein Synthesis and Related Disorders Many antibacterial agents (antibiotics) have been isolated from fungi. Antibiotics are used both clinically and as reagents for unraveling the details of protein, RNA, and DNA synthesis. Antibiotics that have no effect on eukaryotic translation either fail to penetrate the cell membrane (which is quite common) or do not bind to eukaryotic ribosomes. The differences in effectiveness of antibiotics on the two classes of cells in vivo is the basis of their usefulness as therapeutic agents for bacterial infections (Figure 25-18). Some antibiotics are active against both bacterial and mammalian cells. One example is chloramphenicol, which inhibits peptidyltransferase in both bacterial and mitochondrial ribosomes, although eukaryotic cytoplasmic ribosomes are unaffected. Such a drug may be clinically useful if a concentration range can be maintained in the patient in which the antibacterial action is substantial but toxic effects on host cells are minimal. However, because of the potential for toxicity, such antibiotics are used only in serious infections when other drugs fail. Three antibiotic inhibitors that act only on prokaryotes are streptomycin, tetracycline, and erythromycin. Other antibiotics act primarily on eukaryotic cells (Table 25-4). Eukaryotic protein synthesis can also be inhibited by toxins of bacterial origin. An example is the toxin produced by Corynebacterium diphtheriae bacteria carrying a lysogenic bacteriophage. Uninfected bacteria do not elaborate the toxin, and therefore acquisition of the phage is essential for the toxic effect. Diphtheria is an acute infectious disease usually localized in the pharynx, larynx, and nostrils and occasionally in the skin. Initially, the cytotoxic effects are restricted to those tissues immediately adjacent to the bacterial growth. With increased bacterial growth, production of toxin increases and is disseminated to the blood, lymphatics, and other organ systems (e.g., cardiovascular and nervous systems), where it brings about destructive changes.

CHAPTER25 RNA and Protein Synthesis

The pathogenesis of diphtheria is due entirely to the toxin's inhibition of protein synthesis in the host cells. Diphtheria toxin consists of a single polypeptide chain (M.W. 63,000) with two intramolecular disulfide linkages. Toxin binds to the outer surface of susceptible cells at specific sites and enters by receptor-mediated endocytosis. The toxin is cleaved proteolytically into a small fragment, A (M.W. 21,000), and a larger fragment, B (M.W. 42,000), during its internalization. Fragment A is the biologically active part of the molecule and presumably penetrates the cell membrane with the aid of fragment B. Upon entry into the cytoplasm, fragment A catalyzes the ADP ribosylation of the transfer factor, EF-2, leading to its inactivation and the interruption of protein synthesis. The ADP-ribose group is donated by NAD +. The ADP ribosylation reaction, catalyzed by the toxin, is specific for EF-2 of eukaryotic cells; other proteins of eukaryotic and bacterial cells are not substrates. This specificity is due to an unusual amino acid residue in EF-2, diphthamide, which is the acceptor of the ADP ribosyl group. Diphthamide derives from the posttranslational modification of histidine. The acute symptoms are treated with antitoxin. The bacteria, which are gram-positive, succumb to a variety of antibiotics, including penicillin. Diphtheria is effectively prevented by immunization with toxoid (inactivated toxin) preparations. ADP ribosylation is also involved in the action of cholera toxin and in certain pathogenic strains of E. coli (Chapter 12). Cholera toxin catalyzes the ADP ribosylation of the guanidinium, group of a specific arginine residue in the guanine nucleotide-binding protein of the adenylate cyclase. This activates adenylate cyclase, which catalyzes the formation of cAMP from ATE The cAMP formed stimulates secretion of water and electrolytes from intestinal epithelial cells. Thus, patients infected with Vibrio cholerae secrete enormous quantities (up to 20 L/d) of water and electrolytes. Without adequate and prompt replacement, death can ensue owing to dehydration and electrolyte imbalance. Interestingly, in the case of diphtheria toxin, ADPribosylation leads to inactivation of a protein, whereas with cholera toxin, ADP ribosylation causes activation of the protein. A group of plant lectins, such as abrin, ricin, and modeccin, are highly toxic to eukaryotic cells. Their mode of action consists of inhibition of protein synthesis by enzymatically inactivating the EF-2 binding region of the 60S ribosomal subunit, whereas the diphtheria toxin inactivates the EF-2 protein itself. Ricin is isolated from castor beans and has a molecular weight of 66,000. Like most plant and bacterial toxic proteins, ricin contains two

SECTION 25.11

Collagen Biosynthesis and Its Disorders

585

25.11 Collagen Biosynthesis and Its Disorders

TABLE 25-4

Inhibitors of Protein Synthesis in Eukaryotes Inhibitor

Abrin, ricin Diphtheria toxin

*Chloramphenicol

*Puromycin

*Fusidic acid Cycloheximide Pactamycin Showdomycin Sparsomycin

Action

Inhibits binding of aminoacyl tRNA Catalyzes a reaction between NAD* and EF2 to yield an inactive factor. Inhibits translocation. Inhibits peptydyltransferase of mitochondrial ribosomes. Is inactive against cytoplasmic ribosomes. Causes premature chain termination by acting as an analogue of charged tRNA. Inhibits translocation by altering an elongation factor. Inhibits peptidyltransferase. Inhibits positioning of tRNA Met on the 40S ribosome. Inhibits formation of the elF2-tRNAMet-GTP complex. Inhibits translocation.

*Also active on prokaryotic ribosomes.

polypeptide chains with two different but complementary functions. The A chain possesses enzymatic activity and is responsible for toxicity, and the B chain, which is a lectin, binds to galactose-containing glycoproteins or glycolipids on the cell surface. The A and B chains are linked by a labile disulfide linkage. Upon binding of the ricin molecule to the carbohydrate receptors of the cell surface via the B chain, the A chain enters the cytoplasm, presumably by receptor-mediated endocytosis, where it inhibits protein synthesis by irreversible inactivation of the 60S ribosomal subunit. Toxins have been used to develop highly selective cytotoxic agents targeted against specific cells. For example, ricin A has been coupled to agents that selectively bind to cell surface membrane components. The selective agents may be monoclonal antibodies (Chapter 35), hormones, or other cell surface ligands. These conjugates act as selective cytotoxic agents and may have potential therapeutic applications (e.g., in the treatment of cancer). Potential clinical application for diphtheria toxin may be impossible because of the prevalence of the diphtheria antitoxin in human populations.

Collagen occurs in several genetically distinct forms and is the most abundant body protein; most of the body scaffolding is composed of collagen (Chapter 11). Its structure is uniquely suited for this structural role. It is a fibrillar protein but also exists in a nonfibrillar form in the basement membrane. The basic structural unit of collagen, tropocollagen, consists of three polypeptide chains. Each polypeptide chain has the general formula (-Gly-X-Y-)333. Some of the amino acid residues at the X and Y positions are proline, hydroxyproline, lysine, hydroxylysine, and alanine. Collagen is a glycoprotein and contains only two types of carbohydrate residues, namely, glucose and galactose linked in O-glycosidic bonds to hydroxylysyl residues. In collagen, each polypeptide chain is coiled into a special type of a rigid, kinked, left-handed helix, with about three amino acid residues per turn. The three helical polypeptides in turn are wrapped around each other to form a right-handed triple-stranded superhelix that is stabilized by hydrogen bonding. The collagen molecules are aggregated in an ordered quarter-staggered array to give rise to microfibrils, which in turn combine to give fibrils. Covalent cross-linkages occur at various levels of collagen fiber organization and provide great mechanical strength. Collagen biosynthesis is unusual in that it consists of many posttranslational modifications. The unique posttranslational reactions of collagen biosynthesis are 1. Hydroxylation of selected prolyl and lysyl residues. 2. Glycosylation of certain hydroxylysyl residues. 3. Folding of procollagen polypeptides into a triple helix. 4. Conversion of procollagen to collagen. 5. Self-assembly into fibrils. 6. Oxidative deamination of s-amino groups of strategically located lysyl and hydroxylysyl residues to provide reactive aldehydes. These form cross-linkages between polypeptide chains of the same molecule as well as between the adjacent molecules that give strength and stability to the fibrils. The first three processes take place inside the cell, whereas the last three are extracellular modifications. Collagen disorders can result from primary defects in the structure of procollagen or collagen or from secondary changes that affect collagen metabolism. Collagen disorders are both acquired and inherited. Ehlers-Danlos syndrome and osteogenesis imperfecta are examples of inherited primary collagen diseases; scurvy and various fibrotic processes (e.g., pulmonary fibrosis and cirrhosis)

CHAPTER25 RNA and Protein Synthesis

586

are examples of secondary collagen diseases. The mechanisms that lead to collagen diseases are: 1. Aberration in the control mechanisms that alter the balance between synthesis and degradation. This imbalance can lead either to excessive deposition or depletion of collagen. 2. Synthesis of structurally altered collagen due to defects at the level of DNA transcription, RNA processing, translation, or posttranslational modifications. 3. Imbalance in the relative rates of synthesis of genetically distinct collagens. 4. Abnormalities in the packing of collagen molecules into a fiber or in the interaction of collagen fibers with other extracellular components of the connective tissue.

Transcription and Translation of Collagen Polypeptides In general, the transcription and translation of collagen polypeptides resemble other proteins. The primary RNA transcript is capped and polyadenylated, introns are removed, and the functional mRNA is transported to the cytoplasm. The procollagen mRNA, in addition to being one of the largest mRNAs in eukaryotic cells, contains unusually high amounts of G and C, because these bases are overrepresented in the codons for glycine and proline. As is the case for other secretory proteins, the procollagen mRNA encodes a signal sequence coding for hydrophobic

amino acids at the amino terminal ends. The signal sequences of proot 1 chain are different from the proot2 chain and are unusually long (100 amino acid residues) in comparison with other secreted proteins (15-30 residues). Because of their hydrophobicity, these signal sequences are thought to facilitate binding and transport of the nascent polypeptide into cisternae of the endoplasmic reticulum. The signal sequences are removed by proteolysis within the membrane during the translation. The synthetic rate of proot chain is rather slow and is probably related to the unfolding of the secondary structure of procollagen mRNA, which is rich in GC content. Molecular defects at the levels of transcription and translation of collagen polypeptides have not yet been clearly established but may cause one type of EhlersDanlos syndrome (type IV). This disorder is characterized by decreased synthesis of type III collagen in the aorta, intestinal tract, skin, and probably other tissues. Among the Ehlers-Danlos disorders, type IV is the most severe because of the threat of arterial rupture or gastrointestinal perforation. Ehlers-Danlos syndrome is a group of inherited disorders that share similar clinical symptoms; many have been attributed to specific errors in the structure or metabolism of collagen (Table 25-5). The major phenotypic features of Ehlers-Danlos syndrome include hyperextensible skin, hypermobile joints, easy bruisability, and friability of tissues. The clinical severity of these disorders is highly variable. Defects in splicing, transcription, or translation may also cause osteogenesis imperfecta. In this disease, fibroblasts or tissue samples have a markedly diminished capacity for type I collagen

TABLE 25-5

Ehlers-Danlos Syndrome Type I

Inheritance

VI VII

AD AD AD ADorAR XL XL AR AD

VII IX X

AD SL AR

II III IV V

Biochemical Defect

Collagen Fibril Diameter

Unknown Unknown Unknown Decreased type III collagen synthesis and its intracellular accumulation Unknown Unknown Lysyl hydroxylase deficiency Deletion of exons from the genes of proal(I) and pro a 2 (I) that encode the amino-terminal cleavage site; required for the action of procollagen aminoprotease Unknown Lysyl oxidase deficiency Fibronectin deficiency

AD= Autosomal dominant;AR= autosomal recessive; XL= X-linked.

Increased Increased Increased Heterogeneous Heterogeneous Increased Decreased Heterogeneous

Not determined Increased Not determined

SECTION 25.11

Collagen Biosynthesis and Its Disorders

TABLE 25-6

Osteogenesis Imperfecta Type

Inheritance

I

AD

II

AR

III

AR

IV

AD

Biochemical Defect

Decreased synthesis of pro a 1(I), leading to less production of type I collagen because of several defects in the gene. Markedly diminished synthesis of type I collagen; presence of hydroxylysine-enriched collagens associated with delayed helix formation. Delayed secretion of type I procollagen associated with excess mannose residues in the carboxyterminal propeptide region; absence of a 2 (I) synthesis. Point mutations in or ) gene, resulting in substitution for glycine residues.

AD= Autosomal dominant; AR= autosomal recessive.

synthesis. Osteogenesis imperfecta is a genetically and clinically heterogeneous disorder (Table 25-6). It is one of the most common inherited connective tissue disorders and has an incidence of 1" 15,000. It is characterized clinically by defects in bone, tendon, dentin, ligament, and skin. Intracellular Posttranslational Modifications

Hydroxylations of Selected Prolyl and Lysyl Residues The hydroxylation reactions are catalyzed by three enzymes: prolyl 4-hydroxylase (usually known as prolyl hydroxylase), prolyl 3-hydroxylase, and lysyl hydroxylase. These enzymes are located within the cisternae or rough endoplasmic reticulum; as the procollagen chains enter this compartment, the hydroxylations begin. All three enzymes have the same cofactor requirements: ferrous ion, oe-ketoglutarate, molecular oxygen, and ascorbate (vitamin C). The reducing equivalents required for the hydroxylation reaction are provided by the decarboxylation of equimolar amounts of o~-ketoglutarate to succinate and carbon dioxide. One atom of the 02 molecule is incorporated into succinate while the other is incorporated into the hydroxyl group. The data on enzyme kinetics and mechanism of reaction are consistent with the ordered binding of Fe 2+, o~-ketoglutarate, 02, and the peptide

587

substrate and an ordered release of hydroxylated peptide, CO2, succinate, and Fe z+. The Fe z+ does not necessarily dissociate from the enzyme during each catalytic cycle. The activation of oxygen is required and may involve the formation of superoxide (O 2 ). The requirement for ascorbate is specific with purified enzyme preparations. It is not consumed stoichiometrically during the hydroxylation reaction. The exact role of ascorbate is not known, but presumably it reduces either the enzyme iron complex or the free enzyme. Humans, other primates, and guinea pigs lack the enzyme required for the conversion of gluconate to ascorbate (Chapter 15). However, most mammals do possess this enzyme and are able to synthesize ascorbate from glucose by way of glucuronic acid. Other reducing agents, such as dithiothreitol, L-cysteine, and some reduced pteridines, in high concentrations, can partially replace ascorbate in in vitro assays. The generalized hydroxylation reaction is shown below. COOl COOCH2 I I CH2 Enzyme ~ Substrate-OH + I + CO2 CH2 Substrate-H + 02 + I Fe2* CH2 C ~ - O Ascorbate I I COOCOO 0r Succinate

The substrates for all three hydroxylases are highly specific. Free proline and lysine are not substrates. The residue to be hydroxylated must be in a peptide linkage (the minimum is a tripeptide) and in the correct X or Y position in the chain. The proline hydroxylase and the lysyl hydroxylase catalyze the hydroxylation of only prolyl or lysyl residues in the Y positions of peptides with the s e q u e n c e - X - Y - G l y - , whereas the prolyl 3-hydroxylase catalyzes the hydroxylation of prolyl residues at the X position only if Y is 4-hydroxyproline. The conformation of the substrate is also important for catalytic activity. The substrates have to be nonhelical; triple-helical polypeptides do not function as substrates. Thus, the hydroxylation reactions must occur before triplehelix formation. Many important functions are associated with the hydroxylation of prolyl and lysyl residues. The 4-hydroxyprolyl residues participate in interchain hydrogen bonding that aids in the maintenance of triplehelical structures. For example, nonhydroxylated polypeptide chains cannot form stable triple-helical structures at body temperature. The biological role of 3-hydroxyprolyl residues is not known. The hydroxyl groups of hydroxylysyl residues are sites of attachment of carbohydrate

588

and also participate in the intermolecular collagen crosslinks. The importance of lysyl hydroxylation is seen in patients with the type VI variant of Ehlers-Danlos syndrome (Table 25-5). The collagen in these individuals has a decreased fibril diameter and profound changes in mechanical properties. Skin fibroblasts show virtually no lysyl hydroxylase activity. Furthermore, hydroxylysine formation can be severely affected in some tissues, mildly affected in others, and unaffected in still others (e.g., cartilage). These observations suggest the presence of tissue-specific lysyl hydroxylases.

Glycosylation of Hydroxylysyl Residues The glycosylation occurs as the N-terminal ends of the polypeptide chains move into cisternae at specific hydroxylysyl residues. The carbohydrates found are galactose and the disaccharide, glucosylgalactose. These reactions are catalyzed by two specific enzymes: hydroxylysyl galactosyltransferase and galactosylhydroxylysyl glucosyltransferase. The first enzyme catalyzes the transfer of galactose from UDP-galactose to hydroxylylsyl residues, and the second enzyme transfers a glucose from UDP-glucose to galactosylhydroxylysyl residues. Both enzymes require a bivalent cation, preferably manganese. The substrate has to be in the nonhelical conformation, and the glycosylation ceases when the collagen propeptides fold into a triple helix. Thus, both hydroxylations and glycosylation must occur before triple-helix formation, which is an intracellular process. The exact function of the carbohydrate unit in the collagen molecule is not clear, but it may have some role in the organization of fibrils. For example, an inverse relationship between carbohydrate content and fibril diameter has been observed. Deficiency of galactosylhydroxylysyl glucosyltransferase has been seen in the members of kindred with a dominantly inherited disease known as epidermolysis buflosa simplex. This disease belongs to a group of inherited disorders characterized by blister formation in response to minor skin trauma. Further investigation of this disorder may provide information about the role of glycosylation of collagen. Collagen propeptides contain oligosaccharide units typical of glycoproteins in the proregions of the polypeptide chain (Figure 25-19). These oligosaccharide units may be present in either the N- or C-terminal proregions of the peptide or both. In type I procollagen, the oligosaccharide units are at the C-terminal, whereas in type II procollagen, the units are found at both ends, These oligosaccharicles do not appear in the finished collagen molecule because the proregions are excised. The oligosaccharide units probably are synthesized through

CHAPTER25 RNA and Protein Synthesis _ (Man)n ~' ( G I c N * c ) 2

N-Te rmi nal propeptides Occur in part in globular conformation and in short triplehelical structure

Collagen

C-Terminal propeptides Occur in globular conformation

F I G U R E 25-19 Structure of type I procollagen molecule, which consists of two proot l (I) and one proot2(I) chains. Note the presence of oligosaccharide units at the C-terminal propeptide region. In other collagen types, the oligosaccharide units may be present in either one of the regions or both. The disulfide linkages are also present in these regions. These linkages may be either intra- or interchain linkages and may be involved in the triple-helical formation. In the finished collagen molecule, the N- and C-propeptide regions are absent because they are excised (shown by vertical dotted lines). Glc, Glucose; Gal, galactose; Man, mannose; GlcNAc, N-acetylglucosamine. [Reproduced with permission from D. J. Prockop and K. I. Kivirikko, Heritable diseases of collagen. N Engl J Med 311:376 (1984).1

dolichol intermediates (Chapter 16) and linked to the nascent peptide chains within the rough endoplasmic reticulum. The function of these asparaginyl-linked oligosaccharide units is not known. Since ascorbate is required for hydroxylation reactions, its deficiency causes accumulation of the collagen polypeptides in the endoplasmic reticulum and their eventual secretion. However, these proteins lack the proper modifications and therefore cannot be used for assembly of collagen fibrils. Thus, the role of ascorbate in the hydroxylation of collagen is one of its major physiological roles (see also Chapter 38). Scurvy, a connective tissue disorder, is due to vitamin C deficiency in the diet. Most pathological changes of scurvy can be attributed to failure to synthesize collagen. Scurvy patients have defective blood vessels and poor intravascular support, leading to frequent hemorrhages, defective formation of bone and teeth, and poor wound healing. All of these manifestations can be corrected by administration of vitamin C.

Formation of Disulfide Linkages and Assembly of Procollagen Polypeptides into a Triple Helix The propeptide regions of procollagen polypeptides contain cysteine residues that can form both intra- and interchain disulfide linkages (Figure 25-19). As is the case for other proteins that contain disulfide linkages, it is not known whether synthesis of these linkages requires an enzyme or whether it occurs spontaneously. The assembly of procollagen polypeptides into a triple helix appears to have two requirements:

SECTION25.11 Collagen Biosynthesis and Its Disorders

1. The 4-hydroxylation of at least 100 prolyl residues per prod chain; 2. The formation of interchain disulfide linkages in the C-terminal propeptide. The latter cannot be formed until translation is nearly finished. The rate of disulfide bond and triple-helix formation varies greatly from one cell type to anothermonly minutes in tendon cells that synthesize type I collagen but an hour in cells that synthesize basement membrane collagen. These differences in synthesis time may account for the variations in hydroxylation and glycosylation. Thus, the extent of posttranslational modifications depends not only on levels of enzyme and cofactors but also on the time available.

Translocation and Secretion of Procollagen After the procollagen polypeptides are assembled into a triple helix, they are secreted by the classical route. They pass through the smooth endoplasmic reticulurn and the Golgi complex, where they are packaged into membranous vesicles and secreted into the extracellular space by exocytosis. This process requires ATP and may involve microtubules and microfilaments. The conformation of procollagen markedly affects the rate of secretion. Prevention of the formation of a triple helix (e.g., lack of 4-hydroxylation due to vitamin C deficiency) leads to the accumulation of nonhelical propolypeptides within the cisternae of the rough endoplasmic reticulum and a delayed rate in its secretion.

Extracellular Posttranslational Modification Conversion of procollagen to collagen requires at least two proteases: a procollagen aminoprotease and a procollagen carboxyprotease. The former catalyzes removal of the N-terminal propeptide and the latter removal of the C-terminal propeptide. The two enzymes are endopeptidases, function at neutral pH, require a bivalent cation such as Ca 2+, and show a preference for the helical conformation. There appears to be no preferential order for the cleavage of the propeptides. The conversion of procollagen to collagen by removal of propeptides seems to be essential for the formation of collagen fibrils. This supposition is supported by studies of two heritable diseases, one found in humans and the other in cattle, sheep, and cats. In both, the defect lies in the removal of N-terminal propeptides and results in impaired fibril formation. The human disorder is the type VII variant of Ehlers-Danlos syndrome (Table 25-5). Affected individuals exhibit marked joint hypermobility, dislocation of joints, short stature, and minor changes in skin elasticity. Their skin fibroblasts show normal

589

N-terminal procollagen protease activity; however, the defect resides in the deletions of the exon that encodes the protease cleavage sites, thus preventing normal cleavage of the N-terminal propeptide. In animals, however, the defect is the N-terminal procollagen protease deficiency, which produces skin that is easily torn; thus, the disease is known as dermatesparaxis.

Formation of Collagen Fibrils from Ordered Aggregation of Collagen Molecules The collagen molecules formed by removal of the propeptides spontaneously assemble into fibrils. At this stage, the fibrils are still immature and lack tensile strength, which is acquired by cross-linking. The initial step in cross-link formation is the oxidative deamination of or-amino groups in certain lysyl and hydroxylysyl residues catalyzed by lysyl oxidase. The enzyme is a copper-dependent (probably cupric) protein, and the reaction requires molecular oxygen and pyridoxal phosphate for full activity. Only native collagen fibrils function as substrates.

/ HN \ HC--(CH2)4--NH3 + + 02 c/ /%

Lysyl residue

/ HN

\

NH 3

H

HC--(CH2)3--C-'-O + H20

o/ /% Allysine residue

In a similar reaction, the hydroxyallysine residue is formed from hydroxylysine residues. The aldehyde groups react spontaneously with each other or with amino groups, with formation of intra- and intermolecular linkages. Lysyl oxidase is also involved in the formation of elastin crosslinkages. Several collagen disorders result from defects in the formation of cross-links (Chapter 11). The cross-linking disorders can be due to a hereditary cleficiency of lysyl oxidase, inhibition of lysyl oxidase, deficiency of copper, defects in the formation of cross-links, or defects in their stabilization. The genetic deficiency of lysyl oxidase is characterized by Ehlers-Danlos syndrome type IX (Table 25-5) and some forms ofcutis laxa. Type IX EhlersDanlos syndrome patients exhibit extreme extensibility of the skin, "cigarette-paper" scarring, and easy bruisability. In cutis laxa, the skin is loose and inelastic and appears to be too large for the surface it covers. Some affected individuals exhibit deficiency of lysyl oxidase, presumably

590

CHAPTER25 RNA and Protein Synthesis

limited to skin. The animal counterpart of lysyl oxidase deficiency is seen in several variants of aneurysm-prone mice. All of these disorders are X-linked. Lysyl oxidase undergoes irreversible inactivation when exposed to certain nitriles, fl-Aminopropionitrile (HzN-CHz-CHz-CN), which is found in certain peas (e.g., Lathyrus odoratus), inhibits the lysyl oxidase presumably by binding covalently to the enzyme. Ingestion of these peas results in lathyrism, which is characterized by multiple defects in collagen- and elastin-containing tissues. In experimental animals, lathyrism is produced by administering/3-aminopropionitrile during their active growth period. Lysyl oxidase is also irreversibly inhibited by carbonyl reagents (e.g., hydroxylamine) and copperchelating agents. Impaired cross-linking can also occur as a result of copper deficiency, since lysyl oxidase is a copper-dependent enzyme. In experimental animals, copper deprivation causes skeletal and cardiovascular abnormalities. These abnormalities are similar to a human disorder known as Marfan's syndrome, an autosomal dominant trait whose molecular defect is unknown. A genetic defect affecting the gastrointestinal absorption of copper, known as Menkes' (kinky-hair) syndrome, exhibits neurological, connective tissue, pigmentary, and hair abnormalities. These defects can be explained by deficient activities of various copper-requiring enzymes (Chapter 37). The connective tissue changes are attributed to the deficiency of lysyl oxidase. Cross-linking can be inhibited by agents that can react with the aldehyde groups of allysyl and hydroxyllysine residues. Penicillamine reacts with the aldehyde groups, forming a thiazolidine complex and rendering the aldehyde groups unavailable for cross-link formation.

Penicillamine is an effective chelator of copper, thereby inhibiting lysyl oxidase. Penicillamine is used therapeutically in the treatment of disorders involving copper accumulation (Wilson's disease) or in lead and mercury poisoning. Several inherited disorders of methionine metabolism (Chapter 17) give rise to excessive production ofhomocysteine, HS-CHz-CHzCH(NH~-)COO-, and its excretion in urine. The most common form of homoeystinuria is due to a deficiency of cystathionine synthase (Chapter 17). A major clinical manifestation of homocystinuria is connective tissue abnormalities that are probably due to the accumulation of homocysteine, which either inactivates the reactive aldehyde groups or impedes the formation of polyfunctional cross-links. The structure or the metabolism of collagen is abnormal in a variety of other disorders. For example, in diabetes mellitus (Chapter 22), poor wound healing, atrophy of the skin, and thickening of the basement membranes have been attributed to changes in collagen metabolism. Fibrotic tissue and scars consist primarily of collagen fibrils. The fibrosis is a normal process of wound healing in response to trauma and injury. However, excessive fibrosis in parenchymal tissues (e.g., liver and lungs) can severely limit their function. Thus, attempts are being made to develop specific agents that would inhibit collagen synthesis or increase its catabolism. Both growth and aging involve changes in collagen metabolism. These changes occur in the type, quantity, and quality of collagen. Growth of connective tissues requires the appropriate type and amounts of collagen. It is not known to what extent changes in collagen metabolism are responsible for aging.

Supplemental Readings and References

H,\ /OH,

N. Ban, P. Nissen, J. Hansen, et al.: The complete atomic structure of the large ribosomal subunit at 2.4 /~ Resolution. Science 289, 905

(2OOO). +

\H

I

H2NmCH

I

COOH

Penicillamine

H,\/OH, S

~

C

CH H

I

COOH Thiazolidine complex

P. Cramer, D. A. Bushnell, J. Fu, et al.: Architecture of RNA polymerase II and implications for the transcription mechanism. Science 288, 640 (2000). D. E. Draper: Protein-RNA recognition. Annual Reviews in Biochemistry 64, 593 (1995). L. Ellgaard, M. Molinari, and A. Helenius: Setting the standards: quality control in the secretory pathway. Science 286, 1882 (1999). R. Green and H. Noller: Ribosomes and translation. Annual Reviews in Biochemistry 66, 40 (1997). H. Guo, M. Karberg, M. Long, et al.: Group II introns designed to insert into therapeutically relevant DNA target sites in human cells. Science 289, 452 (2000). V. Hatzimanikatis, L. H. Choe, and K. H. Lee: Proteomics: theoretical and experimental considerations. Biotechnology Progress 15, 312 (1999). J. W. B. Hershey, M. B. Mathews, and N. Sonnenberg, eds.: Translational Control, Cold Spring Harbor Laboratory Press, 1996.

Supplemental Readings and References E H. von Hippel: An integrated model of the transcription complex in elongation, termination, and editing. Science 281, 660 (1998). M. Ibba and D. $611: Quality control mechanisms during translation. Science 286, 1893 (1999). H. A. James and I. Gibson: The therapeutic potential of ribozymes. Blood 91, 371 1998. G. Kuznetsov and S. K. Nigam: Folding of secretory and membrane proteins. New England Journal of Medicine 339, 1688 (1998). R. Lund and J. E. Dahlberg: Proofreading and aminoacylation of tRNAs before export from the nucleus. Science 282, 2082 (1998). G. W. Muth, L. O-Donnelly, and S. A. Strobel: A single adenosine with a neutral pKa in the ribosomal peptidyl transferase center. Science 289, 947 (2OOO). W. Neuport: Protein transport into mitochondria. Annual Reviews in Biochemistry 66, 863 (1997). V. M. Pain: Initiation of protein synthesis in eukaryotic cells. European Journal of Biochemistry 236, 747 (1996). T. V. Pestova, S. I. Borukhov, and C. U. T. Hellen: Eukaryotic ribosomes require initiation factors I and IA to locate initiation codons. Nature 394, 854 (1998).

591 A. M. Pyle: Ribozymes: a distinct class of metalloenzymes. Science 261, 709 (1993). H. Riezman: The ins and outs of protein translocation. Science 278, 1728 (1997). C. H. Schilling, J. S. Edwards, and B. O. Palsson: Toward metabolic phenomics: analysis of genomic data using flux balances. Biotechnology Progress 15, 288 (1999). E Schimmel and E. Schmidt: Making connections: RNA-dependent amino acid recognition. Trends in Biomedical Science 20, 1 (1995). H. Stark, E. V. Orlova, J. Rinke-Appel, et al.: Arrangement of tRNAs in preand posttranslational ribosomes revealed by electron cryomicroscopy. Cell 88, 19 (1997). M. D. Wang, M. J Schnitzer, H. Yin, et al.: Force and velocity measured for single molecules of RNA polymerase, Science 282, 902 (1998). S. Wikner, M. Maurizi, and S. Gottesman: Posttranslational quality control: folding, refolding, and degrading proteins. Science 286, 1888 (1999). C. L. Will, C. Schnaider, R. Read, et al.: Identification of both shared and distinct proteins in the major and minor spliceosomes. Science 284, 2003 (1999).

CHAPTER

26

Regulation of Gene Expression

The number of molecules of a protein produced per unit time from a particular gene differs from gene to gene, sometimes greatly. This variation can occur in several ways. For some genes, the strength of a promoter or a ribosome binding site is sufficient to regulate the required concentration of a particular protein. However, other gene products are needed only occasionally, e.g., when a particular nutrient is present in the surrounding medium; still other gene products are required only during particular stages of differentiation or in specific cell types. For such genes, the products are usually not present in significant amounts except when gene expression is activated, and the level of gene expression is determined by an on-off switch of some sort. In this chapter, numerous mechanisms are described by which differences in gene expression are achieved.

26.1 Regulation of mRNA Regulation of gene expression is achieved by 1. Regulation of the number of mRNA molecules produced per unit time, 2. Regulation of the translation of mRNA, 3. Regulation of the number of copies of a gene, and 4. Posttranslational modification.

In prokaryotes, mRNA synthesis can be controlled simply by regulating initiation of transcription. In eukaryotes, mRNA is formed from a primary transcript followed by a series of processing events (e.g., intron excision, polyadenylylation). Eukaryotes regulate not only transcription initiation but the various stages of processing as well. An important aspect ofmRNA regulation is determined by the turnover of mRNA molecules, i.e., translation can occur only as long as the mRNA remains intact. In bacteria, mRNA molecules have a lifetime of only a few minutes, and continued synthesis of mRNA molecules is needed to maintain synthesis of the proteins encoded in the mRNAs. In eukaryotes, the lifetime of mRNA is generally quite long (hours or days), thereby enabling a small number of transcription initiation events to produce proteins over a long period of time. Metabolic pathways normally consist of a large number of enzymes; in some cases, the individual enzymes are used in a particular pathway and nowhere else. In these cases, it is efficient to regulate expression of all or none of the enzymes. In bacterial systems, all enzymes of the pathway are encoded in a single polycistronic mRNA molecule, and synthesis of the mRNA produces all the enzymes. In eukaryotes, common signals for transcription of different genes may be used or the primary transcript can be differentially processed to yield a set of mRNA

593

594

CHAPTER26

Regulation of Gene Expression

molecules each of which encodes one protein. Regulation of synthesis of the primary transcripts simultaneously regulates synthesis of all the gene products. In prokaryotes, gene expression fluctuates because bacteria must be able to respond rapidly to a changing environment. However, in the differentiation of cells of the higher eukaryotes, changes in gene expression are usually irreversible, as in the differentiation of a muscle cell from a precursor cell. By changing the number of copies of a gene during differentiation, eukaryotic cells can regulate levels of gene expression.

26.2 Gene Regulation in Prokaryotes Several common patterns of transcription regulation have been observed in bacteria. The particular pattern depends on the type of metabolic activity of the system being regulated. For example, in a catabolic (degradative) pathway, the concentration of the substrate for the first enzyme in the pathway often determines whether all the enzymes in the pathway are synthesized. In contrast, the final product is often the regulatory substance in a biosynthetic pathway. In the simplest mode, absence of an end product stimulates transcription and presence of an end product inhibits it. Even for a gene in which a single type of protein is synthesized from monocistronic mRNAs, the protein may "autoregulate" itself in the sense that the transcriptional activity of the promoter is determined directly by the concentration of the protein. The molecular mechanisms for each regulatory system vary considerably but usually fall in one of two major categories--negative regulation and positive regulation. In negative regulation, an inhibitor, which keeps transcription turned off, is present in the cell and an antiinhibitor--an inducer--is needed to turn the system on. In positive regulation, an effector molecule (which may be a protein, small molecule, or molecular complex) activates a promoter. Negative and positive regulation are not mutually exclusive, and some degradative pathways, such as the utilization of lactose in E. coli, are both positively and negatively regulated.

Lactose (lac) Operon The prototype for negative regulation is the system in

E. coli for metabolizing lactose (lac) (Figure 26-1). The key chemical reaction carried out by the lac system is a cleavage of lactose to galactose and glucose. This reaction enables bacteria to utilize lactose as a carbon source when glucose is not available. The regulatory mechanism of the lac system, known as the operon model, was the

F I ( ; U R E 26-1 (a) Genetic map of the lac operon, not drawn to scale; the p and o sites are actually much smaller than the genes. (b) Diagram of the lac operon in (I) repressed and (II) induced states. The inducer alters the shape of the repressor, so the repressor can no longer bind to the operator.

first system described in detail, and it defines most of the terminology and concepts in current use. The principal regulatory features of the lac operon are as follows: 1. The products of two genes are required for lactose utilization. The lacz gene encodes an enzyme, /3-galactosidase, that degrades lactose; the lacy gene encodes a protein, lac permease, needed to transport lactose and concentrate it within the cell. A third gene, laca, encodes an enzyme that participates in lactose metabolism only in certain circumstances; some lactose-fermenting bacteria do not have the laca gene, and it is not essential for lactose utilization in bacteria. 2. Lac enzymes are encoded in a single polycistronic mRNA molecule (lac mRNA), and enzyme levels are regulated by controlling synthesis of that mRNA. 3. Synthesis of lac mRNA is determined by the activity of a specific regulatory proteinmthe lac repressor. This protein is encoded in the laci gene, which is transcribed in a monocistronic mRNA molecule distinct from lac mRNA. When the repressor is active,

SECTION 26.2

6ene Regulation in Prokaryotes

lac mRNA is not made. This is the negative regulation of the lac operon. Mutants have been isolated that produce inactive repressor proteins; these cells make lac mRNA continuously (constitutive synthesis), even in the absence of lactose. Actually, some lac mRNA is always made at a level of about one or two transcription events per generation because repression is never complete. This basal level synthesis is responsible for a very small amount of the proteins. 4. A sequence of bases (in the DNA)--the operator lac o--is adjacent to the lac mRNA promoter and binds repressor. When the repressor protein is bound to the operator, attachment of RNA polymerase to the promoter is prevented by steric interference, and initiation of transcription of lac mRNA does not occur. A mutation in the operator, which eliminates repressor binding, also leads to constitutive synthesis. 5. An inducer (a small effector molecule) is needed to initiate transcription of lac mRNA. The inducer of the lac operon, which is allolactose, [/~-D-galactopyranosyl-(1 ~ 6)-/3-D-glucopyranose] a structural isomer of lactose formed by basal synthesis of/~-galactosidase, binds to the repressor and alters its three-dimensional structure such that it is unable to bind to the operator. Thus, in the presence of lactose or other inducers the operator is unoccupied and the promoter is available for initiation of mRNA synthesis. It is common to refer to inactivation of the repressor by an inducer as derepression. Thus, the overall regulatory pattern is as follows: A bacterium growing in a medium without lactose does not make either the lac z or lacy product because the repressor that is made is bound to the operator and prevents synthesis of lac mRNA. The cell grows by utilizing whatever other carbon source is available. If lactose is added (and if glucose is absent; see below), basal level lac y and lac z proteins bring the lactose into the cell and convert it to allolactose. As a result, the repressor is inactivated, RNA polymerase binds to the promoter, and synthesis of lac mRNA begins. Lac mRNA synthesis continues until the lactose is exhausted, in which case the inactive repressor would be reactivated and repression reestablished. The lac operon is also positively regulated, presumably because of the role of glucose in general metabolism. The function of/~-galactosidase is to generate glucose by cleaving lactose, so if both glucose and lactose are available, the cell can use glucose and there is no reason for the lac operon to be induced. (The other cleavage product, galactose, is also converted to glucose by enzymes of the galactose operon.) Indeed, when glucose is present in the medium, little or no lac mRNA is made because glucose

595

TABLE 26-1

Concentration of Cyclic AMP in Cells Growing in Media Having the Indicated Carbon Sources Carbon Source Glucose Glycerol Lactose Lactose + glucose Lactose + glycerol

cAMP Concentration Low High High Low High

indirectly prevents RNA polymerase from binding to the lac promoter. This positive regulation is accomplished by cyclic AMP, a regulatory molecule in both prokaryotes and eukaryotes. Cyclic AMP (cAMP) is synthesized from ATP by the enzyme adenylate cyclase, and its concentration is regulated by glucose metabolism. In a bacterial culture that is starved of glucose, the intracellular concentration of cAMP is very high. If a culture is growing in a medium containing glucose, the cAMP concentration is very low. The observation that in a medium containing a carbon source that cannot enter the glycolytic pathway the cAMP concentration is high (Table 26-1) suggests that some glucose metabolite or derivative is an inhibitor of adenylate cyclase. Cyclic AMP does not act directly as a regulator but is bound to a protein called the cAMP receptor protein (CRP), which forms a complex with cAMP (cAMP-CRP). This complex is active in the lac system and in many other operons involved in catabolic pathways. The cAMPCRP complex binds to a base sequence in the DNA in the lac promoter region and stimulates transcription of lac mRNA; thus, cAMP-CRP functions as a positive regulator. The cAMP-CRP complex is not needed for binding of RNA polymerase to the lac promoter, for a free promoter binds the enzyme. However, an open-promoter complex does not form unless cAMP-CRP is also bound to the appropriate DNA sequence. Thus, when glucose is absent, the cAMP concentration is high and there is enough cAMP-CRP to allow an active transcription complex to form; if glucose is present, cAMP levels are low, no cAMPCRP is available, and transcription cannot begin. Thus, the lac operon is independently regulated both positively and negatively, a feature common to many carbohydrate utilization operons (Figure 26-2).

Tryptophan (trp) O p e r o n The tryptophan operon is responsible for the production of the amino acid tryptophan, whose synthesis occurs in

596

Regulation of Gene Expression

CHAPTER 26

that binds to the operator. When the external supply oftryptophan is depleted (or reduced substantially), the operator becomes exposed, and transcription begins. This type of on-off mechanism--activation of an aporepressor by the product of the biosynthetic pathwaymhas been observed in other biosynthetic pathways. When the trp operon is derepressed, which is usually the case unless the concentration of tryptophan in the medium is very high, the optimal concentration of tryptophan is maintained by a modulating system in which the enzyme concentration varies with the concentration of tryptophan. This modulation is effected by F I G U R E 26-2 Three states of the lac operon showing that lac mRNA is made only if cAMP-CRP is present and repressor is absent.

1. Premature termination of transcription before the first structural gene is reached, and 2. Regulation of the frequency of premature termination by the concentration of tryptophan.

five steps, each requiring a particular enzyme. In E. coli, these enzymes are translated from a single polycistronic mRNA. Adjacent to the enzyme coding sequences in the DNA are a promoter, an operator, and two regions called the leader and the attenuator (Figure 26-3). The leader and attenuator sequences are transcribed. Another gene (trpR) encoding a repressor is located some distance from this gene cluster. Regulation of the trp operon is determined by the concentration of tryptophan: when adequate tryptophan is present in the growth medium, there is no need for tryptophan biosynthesis. Transcription is turned off when a high concentration of tryptophan is present and is turned on when tryptophan is absent. The regulatory signal is the concentration of tryptophan itself and, in contrast with lactose, tryptophan is active in repression rather than induction. The trp operon has two levels of regulation~an on-off mechanism and a modulation system. The protein product of the trpR gene~the trp aporepressor--cannot bind to the operator in contrast with the lac repressor. However, if tryptophan is present, the aporepressor and the tryptophan molecule join together to form an active repressor complex

Located between the 5' end of the trp mRNA molecule and the start codon of the trpE gene is a 162-base segment called the leader (a general term for such regions). Within the leader is a sequence of bases (bases 123 through 150) having regulatory activity. After initiation of mRNA synthesis, most mRNA molecules are terminated in this region (except in the complete absence of tryptophan), yielding a short RNA molecule consisting of only 40 nucleotides and terminating before the structural genes of the operon. This region in which termination occurs is a regulatory region called the attenuator. The base sequence around which termination occurs (Figure 26-4) has the usual features of a transcription termination site~namely, a possible stem-and-loop configuration in the mRNA followed by a sequence of eight AU pairs. The leader sequence has an AUG codon that is in-phase with a UGA stop codon; together these start-stop signals encode a polypeptide of 14 amino acids. The leader sequence has an interesting featuremat positions 10 and 11 are two adjacent tryptophan codons. Premature termination of mRNA synthesis is mediated through translation of the leader peptide. The two tryptophan codons make translation of the leader polypeptide

60 Numberof b a s e pairs

,-

"~

i

162 i

~,-

,~,..-.-..-.~ L ~ . . ~

I

po

~r

~~','-

I

t L

I

trpE

1350

1593

1560

-,--'----"-'~-

-,~

I

I

troD

1196 ~,, ~.___...~

trpC

804

300

_,, , ~ - ~ - . - ~

,-~.~

I

trpB

I

J I

trpA !

Spacer

Spacer

Attenuator sequence

(trpa) i~

Regulation

, I~

Enzyme

production

-

F I G U R E 26-3 Escherichia coli trp operon. For clarity, the regulatory region is enlarged with respect to the coding region. The proper size of each region is indicated by the number of base pairs. L is the leader.

SECTION26.2 Gene Regulation in Prokaryotes

597

F I G U R E 26-4 Terminal region of the trp leader mRNA (right end of L in Figure 26-3). The base sequence given is extended past the termination site at position 140 to show the long stretch of U's. The bases (colored lines) form an inverted repeat sequence that could lead to the stem-and-loop configuration shown (segment 3-4 in Figure 26-5).

sequence quite sensitive to the concentration of charged tRNA Trp. If tryptophan is limiting, there will be insufficient charged tRNA Trp and translation will pause at the tryptophan codons. Thus, regulation depends on two characteristics of gene regulation in bacteria. 1. Transcription and translation are coupled. 2. Base-pair formation in mRNA is eliminated in any segment of the mRNA that is in contact with the ribosome. Figure 26-5 shows that the end of the trp leader peptide is in segment 1 and a ribosom is in contact with about 10 bases

in the mRNA past the codons being translated. When the final codons are being translated, segments 1 and 2 are not paired. In a coupled transcription-translation system, the leading ribosome is not far behind the RNA polymerase molecule. Thus, if the ribosome is in contact with segment 2, when synthesis of segment 4 is being completed, then segments 3 and 4 are free to form the duplex region 3 - 4 without segment 2 competing for segment 3. The presence of the 3 - 4 stem-and-loop configuration allows termination to occur when the terminating sequence of seven U's is reached. If exogenous tryptophan is not present or is present in very small amounts, the concentration of charged tRNATrp will be limiting, and occasionally a translating ribosome will be stalled for an instant at the tryptophan codons. These codons are located 16 bases before the beginning of segment 2. Thus, segment 2 will be free before segment 4 has been translated and the 2-3 duplex will form. In the absence of the 3 - 4 stem and loop, termination will not occur and the complete mRNA molecule will be made, including the coding sequences for the trp genes. Thus, if tryptophan is present in excess, termination occurs and little enzyme is synthesized; if tryptophan is absent, there is no termination and the enzymes are made. At intermediate concentrations, the frequency of ribosome pausing will be such as to maintain the optimal concentrations of enzymes. This tryptophan regulatory mechanism is called attenuation and has been observed for several amino acid biosynthetic operons, e.g., histidine and phenylalanine.

Ribosome ~ ,

"l'rp codons

1 ~: ~ 1~2

i ~

~

~

4

(a) Free mRNA. Base pairs between 1 and 2 and between 3 and 4.

End of transc ription

ontinuationof E ' transcripti~

(b) Low concentration of tryptophan. Ribosome .stalled in region 1 permits formation of 2-3 before transcription of region 4 is completed.

F I G U R E 26-5 Model for the mechanism of attenuation in the E. coli trp operon.

3~

(c) Highconcentration of tryptophan. Ribosome reaches region 2 before region 4 is completed, and permits formation of 3-4.

590

CHAPTER26

Regulation of Gene Expression

Roughly halfway through the life cycle of the phage this promoter enters the cell, transcription occurs by T7 RNA polymerase, and the structural proteins and lysis enzymes are synthesized. Lysis does not occur until phage particles have been assembled. Phage T4 has numerous promoters only a few of which can be recognized by E. coli RNA polymerase. However, unlike T7, the late promoters are made available by successive modification of the E. coli enzyme. These modifications are of two types: addition of phage-encoded protein subunits and chemical modification of preexisting subunits. Temporal regulation occurs because the gene responsible for the first modification is encoded in the first set of mRNAs, that for the second modification in the second set, and so on. To ensure that the late mRNA, which encodes the structural proteins and the lysis enzyme, is not synthesized until adequate DNA has been made by the replication system, the template for this late mRNA cannot be parental T4 DNA but must be a replica. E. coli phage A also uses E. coli RNA polymerase throughout its life cycle. With this phage, regulation of transcription is accomplished in two ways: modification of the positive regulatory elements, such as the cAMPCRP complex, needed for recognition of certain promoters; specific repressors are also used to turn off expression of certain genes. Figure 26-6 shows a portion of the genetic map of ~., which includes four regulatory genes (cro, N, Q, and cll), three promoters (pL, pR, and pR2), and five termination sites (tL1, tRl, tR2, tR3, and tR4). Seven mRNA molecules are also shown; the L and R series are transcribed leftward and rightward respectively from complementary DNA strands. The genes O and P encode proteins required for k DNA replication, and the late genes encode the structural proteins of the phage and the lysis enzyme. The basic transcription sequence is: Two early mRNAs, LI and RI, are made that encode the regulatory proteins N and Cro, respectively. N is an antitermination

Some bacterial operons are regulated solely by attenuation without repressor-operator interactions.

Temporal mRNA Regulation in Phage Systems The lac and trp operons are regulated by molecules that turn gene expression on and off in response to the concentration of nutrients. However, some organisms require regulation of genes in time. For example, in the production of phage (bacteriophage or bacterial viruses) in an infected cell, various biosynthetic activities must occur on schedule. When a bacterium is infected by a phage, the enzymes responsible for breaking open the cell and releasing progeny phage must act late in the life cycle after phage DNA replication and packaging has occurred; otherwise, the cell could burst before any phage formed. Almost all phage systems accomplish this temporal regulation by selective and sequential transcription of particular sets of genes. This transcription gives rise to various classes of mRNA which are usually termed early and late mRNA. Three E. coli phage systems, which accomplish temporal regulation in different ways, are described briefly below. Phage T7 contains several promoters, but only one is recognized by E. coli RNA polymerase. Transcription of T7 DNA begins at this promoter. Two important proteins are encoded there by transcript (early mRNA). One phosphorylates and inactivates the E. coli RNA polymerase, thus preventing E. coli from making any bacterial mRNA. The second protein is a new RNA polymerase that does not recognize any E. coli promoters but is active at the remaining phage promoters. In the absence of E. coli RNA polymerase, the early mRNA is no longer made, but the new T7 RNA polymerase makes the second transcript, which encodes the DNA replication enzymes and other proteins needed early in the life cycle. Transcription at the third promoter is delayed because injection of the phage DNA into the bacterium occurs very slowly.

DNA

att

int

xis

red

N tL1

~ ~ nutL pL oL L1

cl ~ oFt

~ cro pR

c,

nutR tR1

Late genes

o

tR2

tR3

pR2

R1 hr

tR4 (qut)

J

R2 J

R4 v

F I G U R E 26-6 Genetic map of the regulatory genes of phage ~. Genes are listed above the line; sites are below the line. The mRNA molecules are heavy lines. The dashed black arrows with thin lines indicate the sites of action of the N, Cro, and Q proteins.

j

SECTION 26.3

6ene Regulation in Eukaryotes

protein and enables E. coli RNA polymerase to ignore certain transcription termination sites and thereby extend synthesis of these mRNAs. It acts together with a bacterial protein, NusA, by binding to a site called nutR in the DNA. When RNA polymerase, which has initiated transcription at pR, reaches this site, it picks up the N-NusA complex and is thereby modified such that it is able to ignore the tR1 and tR2 terminators. A similar site, nutL, is present downstream from the pL promoter. Because of this antitermination effect, in time RI is extended until the DNA replication proteins and another regulatory protein, Q, are made. Q is also an antitermination protein. A small constitutively synthesized RNA, R4, is made from the outset of the infection. The Q protein binds to a site (qut) downstream from the promoter for R4, causing RNA polymerase to antiterminate, and R4 is extended to form an mRNA that encodes the head, tail, and lysis proteins. From the extended R1 mRNA, the gene-Cro protein is made. The concentration of Cro ultimately reaches a value at which the protein dimerizes, producing the active form. The Cro dimer acts as a repressor at both pL and pR; this activity turns off synthesis of early proteins that are no longer needed and prevents excess synthesis of DNA replication proteins. Lambda, like many phages (but not T4 or T7), can engage in two alternative life cycles. In the lytic cycle, progeny phage particles are produced and the cell ultimately lyses, releasing the phage to the surrounding medium. In the lysogenic cycle, injected phage DNA is repressed and becomes inserted into the bacterial chromosome. At a later time, if the bacterial DNA is damaged sufficiently, the phage DNA is excised and a lytic cycle ensues. DNA damage, in a complicated way, leads indirectly to inactivation of the cell repressor and subsequent excision of the phage DNA and production of progeny phage. The int gene (Figure 26-6) encodes the enzyme that causes insertion of the phage DNA into the bacterial chromosome. Neither the cI repressor nor the Int protein is needed in the lytic cycle; however, and both are needed in the lysogenic cycle and are coordinately regulated. The two products, CI and Int, are encoded in different mRNA molecules but synthesis of the mRNA is initiated by a common signal. The product of the gene clI is a positive regulatory element. Like the cAMP-CRP complex in the lac operon, the cH product must be bound to the DNA adjacent to the promoters for the mRNAs encoding the cI and Int proteins. In this way, the choice of the lytic versus the lysogenic cycle depends on the concentration of the cII protein. If the concentration is low, neither cI repressor nor Int is made and the lytic cycle is followed; if the concentration is high, both cI repressor and Int are made and the lysogenic cycle occurs. The concentration

599

of clI protein is regulated in response to environmental influences.

Regulons One fairly well-studied regulon is the heat-shock or hightemperature protection regulon (htp) which synthesizes a variety of proteins when bacteria are exposed to temperatures above 40~ In E. coli, a single protein, the product of the htpR gene, is a positive effector of all mRNAs of the regulon. The C-terminal end of the HtpR protein is homologous to the RNA polymerase cr subunit, which suggests that the heat-shock response involves a reprogramming of RNA polymerase by this cr-like protein, enabling RNA polymerase to initiate transcription at a class of promoters that is not otherwise recognized. The inducible SOS repair system was described in Chapter 24. This system allows the frequency of replication errors to increase when repair is necessary and is regulated in order to keep the normal error frequency low. Since the repair system is needed only following certain types of DNA damage, some feature of the damage may act as the inducer. Several genes representing functions not directly related to repair are also components of this system, which is called the SOS regulon. Common to all mRNAs of the SOS regulon is an operator region to which a repressor, LexA, binds. When DNA is damaged, a protein called RecA binds to the damaged segment. Binding causes a conformational change in the protein and converts it to a specific protease that is active against only a small number of repressor proteins, LexA among them. Cleavage of LexA prevents it from binding to the SOS operator, so transcription of all mRNAs of the SOS regulon occurs, yielding Uvr enzymes, RecA, and SOS repair proteins. LexA normally represses its own synthesis (it is autoregulated). However, the large amount of RecA protein made after the LexA protein has been cleaved continues to be activated for proteolysis and cleaves LexA; thus, all proteins of the SOS regulon continue to be made. Once DNA repair is completed, RecA loses its proteolysis activity, and LexA is no longer cleaved. Without RecA protease activity, newly made LexA rapidly accumulates, binds to the SOS operators, and turns off the SOS regulon; thus, the state of the cell existing before DNA damage occurred is reestablished.

26.3 Gene Regulation in Eukaryotes Regulationof gene expression in eukaryotes proceeds primarily by control of transcription as in prokaryotes. Some systems are also regulated at the translational level.

600 However, synthesis of mRNA in eukaryotes is not a simple matter of initiation at a promoter, as it is in prokaryotes, but includes several steps in which the primary transcript is converted to mRNA (Chapter 25). Control of these processing steps is also used to regulate gene expression in eukaryotes. A variety of differences between the regulatory mechanisms of prokaryotes and eukaryotes are evident. First, transcription of related genes is initiated in response to a single signal. Second, a single polygenic primary transcript may be differentially processed to yield a set of distinct mRNA molecules each encoding one protein. Third, large proteins can be processed into small, active polypeptides. The second and third mechanisms are unique to eukaryotes. Housekeeping Genes a n d RNases Many proteins in eukaryotic cells such as those involved in glycolysis (Chapter 13) are needed continuously; the genes encoding such proteins are called housekeeping genes. A variety of regulatory mechanisms have evolved to ensure a constant supply of these gene products. In some instances the amount of each protein may be regulated by the strength of the promoter and ribosomal binding site. However, in housekeeping genes with strong promoters, the gene product functions as a repressor and binds to a site adjacent to the gene, thus regulating the level of transcription. This mechanism is called autoregulation. If the gene product is in short supply, transcription is activated; as the concentration of the product increases, the level of transcription is reduced. In bacteria, mRNAs are rapidly degraded, which is essential for an organism that must adapt to rapidly changing environments. The RNase E protein (product of the rne gene) is essential in controlling the stability of mRNAs in E. coli. The RNase E mRNA, in turn, is autoregulated; an amino terminal fragment of RNase E acts as a repressor of the rne gene. In human cells an analogous RNase activity has been purified using antibodies against RNase E and both RNases recognize the same sequence (AUUUA). At least four RNase families can be identified in eukaryotes that have significant homology to pancreatic RNases. Overall, it is estimated that there should be as least 100 different eukaryotic RNases. Because of the crucial roles RNases and their structural homologues play in regulation of gene expression, splicing of primary transcripts, organogenesis, and other cellular activities, RNases are often referred to as housekeeping enzymes. RNase activity in serum and cell extracts is elevated in a variety of cancers and infectious diseases. The level of RNases is regulated by both activators and inhibitors. The

CHAPTER 26

Regulation of Gene Expression

TABLE 26-2 Diseases Associated with Elevated Levels o f RNase Activity in Body Fluids or Cell Extracts Disease

Colorectal adenocaricnoma CML monocytes Chronic fatigue syndrome Liver diseases Chronic myeloid leukemia (CML) Pancreatic necrosis Neurological infectious diseases

RNase

RNase L RNase L RNase L poly-C degrading RNase >200 folds increase in endoribonuclease activity pancreatic RNase poly-C degrading RNase

cellular content of RNase depends on both regulation of endogenous synthesis as well as the uptake of enzymes synthesized in the pancreas. Because of the multiple regulatory roles of RNases in cells, elevated levels are associated with a wide spectrum of diseases (Table 26-2) and serve as useful markers in diagnosis. RNases also are being investigated for antifungal, antiviral, and antitumor activity, and drugs are being developed that are based on specific RNase activities. Gene Families

Eukaryotic genes are not arranged in operons, since each mRNA contains only one gene. However, many systems are grouped into gene families either because of their location or, more commonly, because of their related function. For example, the set of tRNA genes, whose transcription is correlated by some unknown mechanism, is an example of a gene family. Another example, whose regulation is particularly simple, is the family of rRNA genes. Eukaryotic ribosomes contain one copy each of 5S, 5.8S, 18S, and 28S rRNA, and these are synthesized in equal numbers. The 5.8S, 18S, and 28S rRNA molecules are components of a single transcript that is cleaved to yield one copy of each molecule. The 5S rRNA is, however, part of another transcript. How the ratio of the 5S rRNA to the others is maintained is not known; possibly, the promoter strengths of the two transcripts are the same. However, this kind of gene organization is not particularly common, and related genes often reside on different chromosomes. Of particular interest are the developmentally regulated gene families. Similar, but not identical, proteins having the same properties are made at different stages of development. A well-studied example is hemoglobin, a tetrameric protein containing two o~ subunits and two /~ subunits.

SECTION 26.4

Mechanisms of Gene Regulation in Eukaryotes

There are several different forms of both o~ and/3 subunits differing by only one or a few amino acids, and the forms that are present depend on the stage of development of an organism (Chapter 28). For example, the following temporal sequence shows the subunit types present in humans at various times after conception. Subunit Embryonic type (< 8 wk)

Fetal Adult (8-41 wk) (birth--+ henceforth)

or-like /3-like

o/ y G and yA

~2 --+ ~1 ~

o/ /~ and

Both the or- and fl-like genes form distinct gene clusters. A property of each cluster is that the order of the genes is the order in which they are expressed in development.

601

TABLE 26-3

The Role of General Transcription Factors in Initiation of Gene Expression in Human Cells Factor

Subunits*

TFIID

13

TFIIA TFIIB TFIIE TFIIF

3 1 2 2

TFIIH

9

Function

Recognizes TATA box; recruits TFIIB (composed of factors TBP and TAFs) Stabilizes TFIID binding Orients RNAPII to start site Recruits TFIIH (helicase) Destabilizes nonspecific RNAPII-DNA interactions Promotes promoter melting by helicase activity

*RNAPII contains nine subunits.

26.4 Mechanisms of Gene Regulation in Eukaryotes Gene expression in human cells is regulated primarily at the level of transcription as it is in prokaryotic cells. However, because transcription is more complex in eukaryotic cells, gene expression can be regulated at many different stages:

1. Initiation of transcription, 2. Processing the primary transcript (this includes capping, splicing out introns, addition of the polyA tail, and RNA editing in which specific nucleotides are changed), 3. Transport of the processed mRNA from the nucleus to the cytoplasm, 4. Translation of the mRNA (synthesis of protein), and 5. Posttranslational modification of the protein(s). Each of these regulatory steps is crucial to the proper functioning of the cell and the organism. Transcription Factors The synthesis of mRNA molecules by RNA polymerase II (RNAPII) is a multistep process requiring the interaction of numerous proteins with DNA in the region of the promoter (Chapter 25). However, before the expression of a gene can be initiated, the transcription factor TFIID must attach to the promoter. This is followed by the attachment of other proteins, called general transcription factors (GTFs), to DNA in the region of the promoter. A description of the functions of some human GTFs is given in Table 26-3. The various GTFs facilitate attachment of RNAPII to the promoter at the correct nucleotide for initiation, destabilize the DNA at the promoter, and initiate transcription; together the GTFs and RNAPII are called

the preinitiation complex. Once the first phosphodiester bond has formed, transcription has been initiated. The large number of GTFs required to initiate transcription does not completely solve the transcription problem. DNA is organized in chromatin and is wrapped around histone proteins and tightly packaged into nucleosomes (Chapter 24). These structures inhibit transcription as well as DNA replication. A distinct class of transcription factors has been identified that modifies chromatin structure so that transcription can occur. A complex of proteins termed RSF (remodeling and spacing factor) facilitates transcription initiation on chromatin templates in vitro. Another protein complex termed FACT (facilitates chromatin transcription) promotes elongation of RNA chains through nucleosomes. Together, RSF and FACT disassemble nucleosomes and permit transcription initiation to occur. In addition to the GTEs that control the initiation of transcription at the TATA box and the protein factors that disassemble nucleosomes, other transcription factors are required to regulate the expression of particular genes or families of genes. Each transcription factor binds to DNA at a specific sequence, which ensures specificity (Table 26-4). The large number of transcription factors have certain structural motifs that facilitate their binding to DNA and, based on structural similarities, fall into four distinct groups (Figure 26-7).

1. Zinc finger: The structure of the zinc finger motif (discovered in TFIIA) consists of two cysteine residues and two histidine residues separated by 12 amino acids. The two cysteine residues are separated by two amino acids and the two histidine residues are separated by three amino acids. The cysteine and histidine residues are linked by a zinc ion and this

60~

CHAPTER 26

Regulationof Gene Expression

TABLE 26-4 Promoter and Enhancer Controlling Elements and Transcription Factors That Bind to Them Response Element

Consensus Sequence*

Factor

TATAAAA

TATA Box

TFIID

GGCCAATCT

CAAT Box

CTG/NF1

GGGCGG

GC Box

SP1

ATTTGCAT

Octamer

Octl, Oct2 H e a t s h o c k factor

CNNGAANNTCCNNG

HSE

TGGTACAAATGTTCT

GRE

Glucocortoid receptor

CAGGGACGTGACCGCA

TRE

Thyroid receptor

CCATATTAGG

SRE

Steroids

*Consensus sequence is for either a promoter or enhancer element. The letter N stands for any nucleotide (A,G, C,T).

NH 2

NH 2

0-10--0---0

9

0

x 0

."

-.

I

I I

0

0

; o

H2N ~ "

/

0

o Zinc finger

~

X

COOH

COOH

Leucine zipper

9"O~cOOH

NH 2

COO

a- Ihelix

c~- helix ~

Helix - loop - helix COOH

Helix - turn - helix F I G U R E 26-7 Structures of transcription factors that facilitate binding to DNA at specific sequences to regulate gene expression.

SECTION26.4 Mechanisms of Gene Regulation in Eukaryotes

motif is repeated nine times in TFIIA protein. The 12 amino acids between the cysteine and histidine residues form a loop that can interact with the major groove in DNA; this loop is called the "zinc finger." 2. Leucine zipper: The structure of the leucine zipper (first discovered in C/EBP, CCAAT, and enhancer binding protein) consists of four leucine residues in an or-helical segment of the protein. Two polypeptides join to form a Y-shaped dimer whose arms can interact with the major groove of DNA. The stem of the Y-shaped structure is known as the "leucine zipper." 3. Helix-turn-helix (HTH): This structure (first discovered in the )~ bacteriophage repressors, cI and cro) consists of two o~-helical regions of a protein joined by an amino acid sequence that allows the o~ helices to turn. The two o~ helices form a dyad axis of symmetry, a structure that binds tightly to the major groove of DNA. 4. Helix-loop-helix (HLH): This structure is similar to the helix-turn-helix in that it consists of two oe-helical segments joined by a long sequence of amino acids that can form a loop. This gives the two o~-helical segments more flexibility so that the proteins can fit into the large groove of DNA at some distance from one another. This may facilitate looping of the DNA to make enhancer or promoter sites more accessible.

603

The serum albumin gene is one example of a gene that is regulated by several transcription factors. Although this gene is present in all tissues, it is only expressed in liver and spleen. The gene is activated by five different transcription factors that bind DNA in a region located between the CCAAT and TATA boxes (Figure 26-8). The transcription factor NF- 1 actually binds to the right of the CCAAT box. Binding sites for transcription factors are determined by changing bases in the DNA one at a time and observing changes in the binding of transcription factors.

Steroid Receptors Steroid hormones perform many functions in cells, one of which is to activate gene expression by binding to steroid receptors, proteins in the cytoplasm that, when activated, act as factors that initiate transcription. All steroid hormones are derived from cholesterol and, as a result, have similar chemical structures. Steroid hormones differ one from another primarily in hydroxylation of particular carbon atoms and by aromatization of the steroid A ring of the molecule. Once a steroid hormone binds to a steroid receptor protein, the complex undergoes a series of structural changes that result in the complex binding to DNA at a particular sequence called a steroid response element (SRE)

FIGURE 26-8 Expression of the human serum albumingene is regulated by five transcription factors, four of which bind to the promoter region.

60~

CHAPTER 26

located at some distance upstream or downstream from the promoter. Steroid receptor proteins are synthesized from a gene family that shows a high degree of homology in the DNA binding region. All steroid receptors belong to one of five classes: androgen receptor (AR), estrogen receptor (ER), glucocorticoid receptor (GR), mineralocorticoid receptor (MR), and progesterone receptor (PR). Also, all receptors contain a zinc finger motif which, if altered by mutation, destroys the steroid receptor's function.

G-Protein Diseases Mutations that alter the functions of trimeric guanine nucleotide-binding proteins (G-proteins) are responsible for wide range of diseases (Table 26-5). While most of these diseases are quite rare, the identification of an altered G-protein in hypertension is estimated to affect as many as 15% of patients with hypertension. G-proteins relay signals from hormones, neurotransmitters, and other extracellular agents to different intracellular effectors that regulate enzymes and ion channels. G-proteins are responsible for signal transfer from more than 1000 cell receptors. Changes in G-proteins cause excessive or insufficient transmission of signals that are essential for proper cellular functioning. G-proteins consist of three subunits: an ot subunit that is loosely bound to a dimer consisting of a 13 subunit that is tightly bound to a V subunit. Human chromosomes contain genes for 16ot, 6fl, and 12y subunits. Thus, a large

TABLE 26-5 Diseases Caused by Mutations That Alter the Signal Activities of G Proteins

Diseases Excess Signal Pituitary and thyroid adenomas Adrenal and ovarian adenomas McCune-Albright syndrome Cholera

G Proteins

G3ot Gi2o~ Gsot G2o~

Deficient Signal Pseudohypoparathyroidism type Ia Nightblindness

G5oL

Deficient or Excessive Signal Pertussis Essential hypertension

GioL

GtoL

133

Regulation of Gene Expression

variety of Got proteins define different G-protein trimers that regulate signaling pathways such as cAMP synthesis, cGMP breakdown, closing of Ca 2+ channels, and opening of K + channels. The activity of G-proteins is regulated by the binding and hydrolysis of guanosine triphosphate (GTP) by the Got subunit. If guanosine diphosphate (GDP) is bound to the ot subunit, it will associate with the fly subunits, but the trimer is inactive. When a cell receptor is activated, the trimer releases GDP and the ot subunit is able to bind GTE After binding GTP the ot subunit dissociates from the/3 V subunit and from the receptor. Either the ot-GTP complex or the/3 9/subunits can then activate downstream effectors. Thus, defects in any of the numerous G-proteins may alter cellular biochemistry and cause disease.

Regulation of Transcription by Methylation Most cells contain several enzymes that transfer a methyl group from S-adenosylmethionine (Chapter 17) to cytosine or adenine in DNA. These DNA methylases are both base-specific and sequence-specific. Another enzyme that methylates cytosine only in particular CpG sequences also is important for transcription. In a few cases, particularly in vertebrates, methylation of CpG sequences prevents transcription of some genes. Evidence for a regulatory role of methylation is: 1. Certain genes are heavily methylated in cells in which the gene is not expressed and unmethylated in cells in which the gene is expressed. 2. In vitro methylation of the upstream site of a cloned v-globulin gene prevents transcription. Methylation outside the upstream sequence does not inhibit transcription. 3. If the base analogue 5-azacytidine is added to cultures of growing cells, newly made DNA contains the analogue, and methylation of CpG sites containing the analogue fails to take place (5-azacytidine is also a general inhibitor of many methylases). Cells in such cultures gain the ability to make proteins whose synthesis is normally turned off (i.e., in cells growing in medium lacking the base analogue). 4. The housekeeping genes, which provide for general cell function and which are continuously transcribed, are rarely methylated in or near their initiation regions. Undifferentiated and precursor cells often replicate. If methylation actually prevents gene expression in some types of cell, an inhibitory methylated site must be inherited as a methylated site in a daughter strand during DNA

SECTION26.4 Mechanisms of Gene Regulation in Eukaryotes

605

FIGURE 26-9 Mechanism by which the pattern of methylation in parental DNA is inherited in daughter molecules. The rule is that a C in a CpG sequence can be methylated only if the C in the complementary CpG sequence is already methylated. For clarity only, methyl groups have been drawn outside the sugar-phosphate strands.

replication. This requires methylation of a sequence complementary (rather than identical) to a methylated site. A property of certain DNA methylases shows how a methylated parental strand can direct methylation of the appropriate daughter sequence, i.e., these enzymes only methylate CpG (embedded in certain surrounding sequences) and only when the CpG in the opposite strand is already methylated (Figure 26-9). In this way, the methylation pattern of parental DNA strands is inherited by the daughter strands. The sex chromosome composition of human males and females is XY and XX, respectively. If cells contain more than one X chromosome, all except one are inactivated. The cells of XX females contain a structure called a Barr body, which is a condensed, heterochromatic, transcriptionally inactive X chromosome. The cells of XXY males also contain a Barr body. XXY males suffer from Klinefelter's syndrome; they are usually mentally retarded, sterile, and suffer from physiological and developmental abnormalities. It is thought that extra X chromosomes must be prevented from gene expression to preserve the correct gene dosage in both males and females. Evidence for inactivation of one X chromosome in XX females comes from the observation that females are mosaic for X-linked alleles that are heterozygous. For example, a woman who is heterozygous for a gene that controls production of sweat glands has patches of skin that perspire and patches that do not. Cells in the patches of skin

that do not perspire express the mutant allele, and the wild type is silent in the inactive X chromosome. Cells in the patches of skin that do perspire express the wild type allele, and the mutant allele is silent. X chromosome inactivation occurs early in embryonic female development and the X chromosome that is inactivated is selected at random. Once selected, the same X chromosome is inactivated in subsequent cell divisions, which explains why patches of skin and other tissues differ in the expression of X-linked heterozygous genetic loci. Inactivation of the X chromosome is thought to occur in three phases: initiation, spreading, and maintenance. Initiation involves the choice of chromosome to be inactivated and depends on a unique genetic locus on the X chromosome called the X-inactivation center (X/c). Once one X-inactivation center has been triggered in a cell, the other X chromosome is protected from inactivation by a "blocking signal." The inactivation signal spreads from the Xic locus, eventually to inactivating almost all of the genes on that X chromosome. However, at least 19 genes on inactive X chromosomes have been shown to have some transcriptional activity, so inactivation is not complete. Specific maintenance mechanisms ensure that the inactive X chromosome is clonally transmitted and that it remains inactive in subsequent cell divisions. Methylation of CpG islands at the beginning of genes in the inactive X chromosome appears to play an important role in suppressing gene expression; the corresponding CpG islands in the active X chromosome are usually unmethylated.

606

Genomic Imprinting Approximately two dozen autosomal genes in humans are inherited in a silent state from one parent and in a fully active state from the other parent. Such genes are said to be "imprinted" by the parents during gamete formation and the phenomenon is referred to as genomir imprinting, which does not change the nucleotide sequence of DNA. Rather, it is an epigenetic phenomenon in which the DNA at a particular locus is marked according to gender, and this determines whether or not the locus is expressed during embryonic development. Although regarded as a normal epigenetic phenomenon, two clinically different disorders are characterized by genomic imprinting. Prader- Willi and Angelman syndromes are distinct disorders associated with multiple abnormalities and mental retardation. Both disorders are caused by mutations at the proximal end of chromosome 15 that silence one or more genes. In Prader-Willi syndrome, the maternally inherited chromosome is silent; in Angelman syndrome, the paternally inherited chromosome is silent. For both syndromes, a small deletion is usually responsible for the genomic imprinting that silences the relevant gene(s). Both syndromes can also result from having both copies of chromosome 15 derive from only one parent, a condition called uniparental disomy. Angelman syndrome is caused by mutation in a single gene whereas Prader-Willi syndrome is caused by mutations in more than one gene. Thus, Angelman syndrome can also be caused by mutations in the responsible gene itself, which is not the case for PraderWilli syndrome. Diagnosis of both can be confirmed by analysis of DNA methylation in the respective genes since methylation is the mechanism used for imprinting.

Regulation of RNA Processing Initiation of transcription ultimately leads to production of a primary transcript, which in higher eukaryotes is processed to form an mRNA. Alternative processing patterns can yield different mRNAs. One example comes from chicken skeletal muscle in which two forms of the muscle protein myosin, LC1 and LC3, are produced. The myosin gene has two different TATA sequences that yield two different primary transcripts. These two transcripts are processed differently to form mRNA molecules encoding distinct forms of the protein (Figure 26-10). Another example of regulation of RNA processing is found in adenovirus, which has only a few promoters but which makes numerous primary transcripts and a large number of mRNAs. A fairly small amount of DNA is used efficiently because proteins are translated in all reading

CHAPTER26 Regulation of Gene Expression

frames and different regions of the DNA are, by virtue of distinct RNA splicing patterns, used to form different proteins. The amount of each mRNA is regulated with respect to the time after infection. RNAs 1 and 2 are both formed shortly after the primary transcript is made, although more of mRNA-2 is made. Later in the viral life cycle, mRNA- 1 is not made and mRNA-2 is abundant. If cycloheximide, an inhibitor of protein synthesis, is added to the infected cells before the shift in splicing pattern takes place, the shift does not occur. This inhibition implies that a newly synthesized protein is a positive effector of the shift. Cycloheximide has a similar effect on other mRNA species derived from other primary transcripts at late times. Adenovirus has a single promoter for all RNA made late in the cycle of infection. The primary transcripts terminate at five major polyadenylation sites. Each termination site influences the splicing pattern by allowing particular introns or intron termini to be present or not. The five sets are not used with equal frequency, with the result that most mRNAs encoding various genes are not the same. This is the primary mechanism for determining the relative amounts of the different structural proteins synthesized late in the adenovirus life cycle. Another type of regulation of processing involves choice of different sites of polyadenylation. One example is the differential synthesis of the hormone calcitonin in different tissues; another is the synthesis of two forms of the heavy chain of immunoglobulins (Chapter 35). In both cases, the differential processing includes distinct patterns of intron excision (i.e., splicing), but they are necessitated by an earlier event in which differential poly(A) sites are selected from the primary transcript. That is, when the poly(A) site nearer the promoter is selected, a splice site used in the larger primary transcript is not present, so a different splice pattern results. Thus, slightly different proteins are synthesized. The calcitonin gene consists of five exons and uses two alternative polyadenylation sites that respond to different signals in different tissues. In the thyroid calcitonin is produced by a signal that produces a pre-mRNA consisting of exons 1-4. The introns are then spliced out to give the mature mRNA. However, in neural tissue a different polyadenylation site is activated that produces a pre-mRNA consisting of exons 1-5 and the intervening introns. When this larger pre-mRNA is processed, the introns are spliced out but so is exon 4, producing a mature mRNA consisting of exons 1-3 and 5. This is translated into a growth factor called calcitoninrelated gene peptide (CRGP). Mutations in the calcitonin gene can result in both adrenal and thyroid tumors,

SECTION26.4 Mechanisms of Gene Regulation in Eukaryotes

607

F I G U R E 26-10 Chicken LC1/LC3 gene. Two distinct TATA sequences lead to production of different primary transcripts, which contain the same coding sequences. Two modes of intron excision lead to the formation of distinct mRNA molecules that encode proteins having different amino terminal regions and the same carboxy terminal regions.

conditions that are classified as multiple endocrine neo-

plasias. Alternative RNA Splicing and Editing Most primary transcripts in eukaryotic cells derive from complete removal of all introns and complete joining of all exons. This results in only one species of mature mRNA being synthesized from each primary transcript. However, many eukaryotic genes can give rise to many different forms of mature mRNAs using different promoters and different polyadenylation sites (discussed above), as well as by alternative splicing. This form of regulation of RNA processing is especially useful because a single gene can be expressed differently at various developmental stages or in different tissues of the same organism. One example is the troponin T gene that synthesizes the fast skeletal muscle protein. This gene consists of 18 exons; however, only 11 are found in all mature mRNAs. Five of the exons can be included or excluded and two are mutually exclusive; if one is included, the other is excluded. Altogether, 64 different mature mRNAs can be produced by alternative splicing of the primary transcript of the troponin T gene. RNA editing involves processing of RNA in the nucleus by enzymes that change a single nucleotide (insertion, deletion, or substitution). One example is apolipoprotein B (apo B) gene. In the liver this gene produces a 4536-amino-acid protein (apo B100), whereas in the small intestine the same gene produces a 2152-amino-acid protein (apo B48). The truncated protein is identical in

amino acid sequence to the first 2152 amino acids in apo B 100. This occurs because in cells of the small intestine one nucleotide (at position 6666) is edited by deamination of a cytosine residue. The conversion of a cytosine to uracil at this position produces a stop codon that terminates translation and produces the truncated protein. Thus, selective editing of mRNAs prior to translation in specific tissues is used to produce different proteins.

Regulation of Iron Utilization in Cells Iron is an important constituent of human cells and regulates many biochemical functions (Chapter 29). Iron acts as an effector molecule in the translation of several mRNAs by binding to either 3' or 5' stem-loop structures, called iron-response the elements (IREs), that flank the coding sequences for several genes whose expression is regulated by iron. In particular, the inRNAs for the transferrin receptor, light and heavy chains of ferritin, an crythrolytic form of aminolevulinic acid synthetase, and a form of mitochondrial aconitase are regulated by IREs and an IRE-binding protein (IRE-BP). The IRE in the ferritin in RNA is in the 5' flanking sequence. Translation of the mRNA is regulated by binding of IRE-BP whose activity, in turn, is regulated by the concentration of iron in the cell. In contrast, the IRE of transferrin mRNA is in the 3' flanking sequence; binding of IRE-BP to this IRE regulates turnover of the mRNA. This example illustrates the variety of ways that gene expression can be regulated under varying physiological conditions in human cells.

60~

Polyproteins In prokaryotes, coordinated regulation of the synthesis of several gene products is accomplished by regulation of the synthesis of a single polycistronic mRNA molecule encoding all of the products. The analogue to this arrangement in eukaryotes is the synthesis of a polyprotein, a large polypeptide that is cleaved after translation to yield individual proteins. Each protein can be thought of as the product of a single gene. In such a system, the coding sequences of each gene in the polyprotein unit are not separated by stop and start codons but instead by specific amino acid sequences that are recognized as cleavage sites by particular protein-cutting enzymes. Polyproteins have been observed with up to eight cleavage sites; the cleavage sites are cut not simultaneously but rather in a specific order. Use of a polyprotein serves to maintain an equal molar ratio of the constituent proteins; moreover, delay in cutting at certain sites introduces a temporal sequence of production of individual proteins, a mechanism frequently used by animal viruses. Some polyproteins are differentially cleaved in different tissues. An example is proopiomelanocortin, a polyprotein that is the source of several hormones synthesized in the pituitary gland. In the anterior lobe of the pituitary, the polyprotein is cleaved to release/3-1ipotropin and adrenocorticotropic hormone (ACTH). In the intermediate lobe, a different pattern of cleavage forms/~-endorphin and ot-melanotropin (Chapter 31 ).

Translational Regulation Translational regulation refers to the number of times a finished mRNA molecule is translated. The three ways in which translation of a particular mRNA may be regulated are 1. By the lifetime of the mRNA, 2. By the probability of initiation of translation, and 3. By regulation of the rate of overall protein synthesis. The silk gland'of the silkworm Bombyx mori predominantly synthesizes a single type of protein, silkfibroin. Since the worm takes several days to construct its cocoon, it is the amount and not the rate of fibroin synthesis that must be great; hence, the silkworm can manage with a fibroin mRNA molecule that is very long lived. Silk production begins with chromatid amplification in which, over a period of several days, the single cell of which the gland is composed has a ploidy of 106. Each fibroin gene is transcribed from a strong promoter to yield about 104 fibroin mRNA molecules. An "average" eukaryotic mRNA molecule has a lifetime of 3 hours before it is degraded.

CHAPTER26

Regulation of Gene Expression

However, fibroin mRNA survives for several days, during which each mRNA molecule is translated repeatedly to yield 105 fibroin molecules. Thus, the whole unicellular silk gland makes 1015 molecules or 300 mg of fibroin. Production of a large amount of a single type of protein by means of a prolonged mRNA lifetime is common in highly differentiated cells. For example, cells of the chicken oviduct, which makes ovalbumin (for egg white), contain only a single copy of the ovalbumin gene per haploid set of chromosomes, but the cellular mRNA is long lived. Translational and transcriptional control are sometimes combined. For example, insulin (which regulates the synthesis of a large number of substances) andprolactin (another hormone) are required together for production of casein (milk protein) in mammary tissue. Both hormones are needed to initiate transcription but prolactin in addition, increases the lifetime of casein mRNA. The synthesis of some proteins is regulated by direct action of the protein on the mRNA. For instance, the concentration of one type of immunoglobulin is kept constant by self-inhibition of translation. This protein, like all immunoglobulins, consists of two H chains and two L chains. The tetramer binds specifically to H-chain mRNA and thereby inhibits initiation of translation.

Regulation of Protein Activity Many enzymes contain several subunits and are regulated by a process known as allostery. A common arrangement in enzymes is that the binding sites for the molecule that is acted on (the substrate) and the inhibitor (which may be the product) are located on different subunits. If binding of the inhibitor prevents binding of the substrate, the information from a site on one subunit must somehow be transmitted to the other subunit. This can be accomplished by the following subunit interactions. Binding of the inhibitor molecule alters the shape of the subunit to which it is bound, resulting in changes in the reactive sites on other subunits. If the subunits remain in contact, all subunits adjoining the first will undergo a conformational change at their respective subunit interaction sites, altering, in turn, the substrate binding site of other subunits. Proteins capable of undergoing such conformational interactions are called allosteric proteins (Chapter 7). In mammalian cells, cAMP is called a second messenger, because it regulates the activities of many proteins. Furthermore, certain hormones and cAMP work in concert to regulate enzymatic activities. Many hormones regulate metabolic processes, such as glucose metabolism and calcium utilization, through binding to specific receptors in the cell membranes of target cells. However, many

SECTION 26.6

Regulation of Cell Proliferation: Oncogenes

hormones are not capable of penetrating the target cells, and instead the binding of a hormone to a membrane receptor induces intracellular synthesis of cAMP and other second messengers; these second messengers then cause the desired metabolic effects (Chapter 30).

26.5 Regulation of Cell Death" Apoptosis Apoptosis (programmed cell death) is characterized by a complex series of biochemical changes that culminate in cell death without inflammation or swelling, which are signs of necrosis. Embryonic, fetal, and postnatal development involve cell death by apoptosis, which serves to eliminate excessive cell proliferation and migration. Apoptosis is initiated by a variety of external stimuli and molecular events such as oxidative stress, mitochondrial permeability transition, mitochondrial cytochrome c release, activation of caspase proteases, activation of endonucleases, transglutaminase activation, and poly(ADP-ribose) polymerase cleavage. Necrosis is a form of passive cell death that is very distinct from apoptosis and usually occurs in a regional group of cells after a particular event, such as stroke, infarction, hemorrhage, or infection. In contrast to necrosis, apoptosis is an active process requiring RNA synthesis, protein synthesis, and new enzyme activities. Apoptosis usually involves isolated single cells that may undergo programmed cell death in a particular organ at different times. Certain features of a cell undergoing death by apoptosis distinguish it from one dying by necrosis. First, the cell shrinks, the plasma membrane and vesiculates change shape, and phosphatidylserine is redistributed on the cell surface. Specific nuclear changes also are diagnostic of a cell undergoing death by apoptosis. Nuclear fragmentation occurs from chromatin condensation and internucleosomal DNA breakdown. Analysis of the DNA shows a laddering fragment pattern that is the result of abnormally activated endonucleases. Although apoptosis is essential to normal development, it also appears to be the cause of pathogenic changes in several chronic neurological diseases including amyotrophic lateral sclerosis (ALS), Parkinson 's disease, Huntington's disease, and spinocerebellar ataxias. Apoptosis also may play a key role in the development of Alzheimer's disease. Regardless of the activating signal for apoptosis, dying cells follow the same series of events. Caspase proteinases play a central role in apoptosis; they are cysteine proteinases that are activated by apoptotic signals and degrade cellular proteins by cleavage after aspartate residues. The first caspase discovered (caspase-1) in mammalian cells was recognized as a "cell death" enzyme by its similarity

609 to the cell death gene, ced-3, in the nematode Caenorhabditis elegans. Subsequent studies have identified almost a dozen mammalian caspases that function as apoptotic initiators (caspases 2, 8, 9, 10), apoptotic executioners (caspases 3, 6, 7), and cytokine processors (caspases 1,4, 5). Activation of caspases 1 and 3 appears to play a key role in the pathogenesis of chronic neurological diseases resulting from progressive cell death. On the other hand, caspase inactivation appears to promote oncogenesis by allowing cell accumulation. Thus, caspases are prime targets for intervention in a variety of diseases that progress to death. ALS was first described by the French neurologist Chariot in 1869. The disease involves rapid loss of motor neurons in the cortex, brain stem, and spinal cord; death usually follows 3-5 years after diagnosis. ALS is more commonly referred to as Lou Gehrig's disease because it struck down the New York Yankees immortal at the peak of his baseball career. ALS usually occurs in a sporadic form but in rare cases there is a familial factor; this provided the first clues to the molecular basis for ALS. One form of ALS results from mutations in the SOM gene that codes for the cystolic enzyme copper/zinc superoxide dismutase (Cu,Zn-SOD). Mice lacking this enzyme do not develop ALS but mice that overexpress the mutant gene do develop symptoms characteristic of ALS and motor neuron death. It is now known that a mutant SOD1 gene causes apoptotic motor neuron death by activating initiator caspases. Using drugs that inhibit caspase activity in ALS transgenic mice, the life span of the mice has been increased by about 70%. As more molecular details of caspase functions and apoptosis in cancer and neurological diseases are revealed, it eventually may be possible to develop drugs that ameliorate or suppress symptoms of these deadly diseases.

26.6 Regulat!on of Cell Proliferation" Oncogenes In the course of differentiation, most adult cell types of higher eukaryotes lose the ability to divide. Only a small number of cell types, (e.g., cells of the intestinal mucosa, erythropoietic cells, and male germline cells) continue to do so. Many genes and regulatory elements determine whether a cell can or cannot divide. However, a class of genes has been discovered that, in certain circumstances, restores the ability of the cells to divide and, more significantly, to do so with little or no control. These oncogenes have been detected in many tumors and in RNA and DNA oncogenic viruses that cause cancers in infected animals. The first oncogene discovered was src, a gene carried by Rous sarcoma virus that causes sarcomas in infected fowl.

610 LTR

CHAPTER26 Regulation of Gene Expression .

gag

F I G U R E 26-11 Genome of the Rous sarcoma virus (RSV). The single-stranded RNA is about 10 kb. Four proteins are encoded in this retrovirus: gag, a core protein; pol, reverse transcriptase; env, glycoprotein of the envelope; and src, a nonessential viral gene whose product is responsible for cellular transformation. LTR, Long terminal repeat sequences present at the ends of each viral RNA molecule.

This single-stranded oncogenic RNA virus infects chicken cells in which the RNA is copied by reverse transcriptase and converted to DNA (Figure 26-11). The DNA copy integrates into a chromosome, where it becomes a provirus. Integration of viral DNA is an essential step in the production of more virus particles. Expression of the src gene, which is nonessential to multiplication of the virus, causes tumor formation in infected fowl. Infected chicken fibroblast cells cultured in vitro become transformed and grow in an unregulated fashion that is visibly distinct from the growth of uninfected cells. Study of DNA from avian tissue has indicated that the src gene is present in cells from normal tissue. However, the cellular src gene, denoted c-src, contains introns, whereas the viral gene, v-src, does not. This has led to the conclusion that at some time in the distant past, processed v-src mRNA was converted to DNA by reverse transcriptase, a virus-encoded enzyme that makes DNA from a single-stranded RNA template, and the virus picked up this DNA copy by genetic recombination. Similar oncogenes have been found in other viruses, and in each case a related cellular copy of the gene is present. The cellular counterpart is called a p r o t o - o n c o g e n e . The oncogenes and proto-oncogenes are almost never identical because they have evolved separately. The product of the src gene is a tyrosine-specific protein kinase, an unusual enzyme because most protein kinases are active only against serine. This enzyme is capable of phosphorylating a large number of cellular proteins, causing either activation or inactivation. The first human oncogene was discovered by testing DNA fragments isolated from human cancer cells and testing for their ability to transform a culture of mouse cells. This experiment utilized DNA from a line of human bladder cancer cells that was applied to a culture of National Institutes of Health 3T3 cells, an established mouse cell line. Some of the mouse cells became transformed, which suggested that an oncogene was being expressed in the bladder cancer cells. Only a single DNA fragment could induce the transformation; therefore, this fragment contained the human bladder cancer oncogene, which was later named ras. A search for a matching sequence in non-

cancerous cells (in analogy to the src study) uncovered a proto-oncogene that differed from ras by a single base pair in the fragment that was sequenced. That is, ras is a mutant form of a normal gene. ras has also been found in many other viruses and eukaryotes, including yeast. A list of most known oncogenes is given in Table 26-6.Their names are abbreviations or acronyms for the virus or tissue of origin. Among the numerous oncogenes that have been identified in cancer cells, the ras genes are the most common. Expression of the oncogenic or mutant ras gene is found in 50% of human cancers. The protein produced by the ras genes is called p21 (M.W. 21,000) and contains 189 amino acid residues. The oncogenic ras gene proteins differ from the normal protein by a single amino acid substitution in most instances, and these changes may be responsible in some cases for converting a normal cellular gene product to a cancer-promoting one. The normal ras p21 binds GTP and GDP; it also hydrolyzes GTP to GDE The mutant ras p21 proteins show reduced or complete absence of GTPase activity. In this regard, normal p21 appears to be similar to the cellular G (GTP-binding) protein that activates adenylate cyclase and thereby mediates cellular activities. Chromosome changes are the hallmark of many cancer cells. The most common type of change is transloeation, a chromosome alteration in which two chromosomes have exchanged segments. For example, in Burkitt's lymp h o m a , fragments in chromosomes 8 and 14 are exchanged in 90% of affected individuals, between chromosomes 8 and 2 in 5%; and between chromosomes 8 and 22 in another 5%. In each case, a segment of chromosome 8 has been moved. The p r o t o - o n e o g e n e e - m y e is located on chromosome 8, and in each of the translocations, cmyc is relocated adjacent to a gene encoding an antibody. This relocation apparently places the proto-oncogene under control of the genes that regulate antibody synthesis, so that c-myc is made in large quantities. Involvement of a different oncogenic protein has been demonstrated in c h r o n i c m y e l o g e n o u s l e u k e m i a (CML). This form of leukemia is associated with the so-called Philadelphia (Ph) chromosome and is characterized by a translocation from chromosome 9 to a new location on the short arm of chromosome 22. The translocation results in the production of a chimeric abl oncogenic protein. The chimeric abl oncogene has enhanced tyrosine protein kinase activity both in vivo and in vitro that is correlated with the development of leukemia. The translocated segment of DNA is inserted into chromosome 22 in a region consisting of 200 bp called the b r e a k p o i n t cluster region (bcr). Consequently, in different CML patients the bcr/abl p21 proteins differ slightly in amino acid sequence, but all

SECTION 26.6

Regulation of Cell Proliferation: Oncogenes

611

TABLE 26-6 Some Oncogenes Involved in Human Cancers*

Oncogene K-ras N-ras H-ras erb-B erb-B2 C-myc N-myc L-myc bcl-2 cycd- 1 bcr-abl cdk-4 /3-cat hst mdm-2 gll ttg

Protein**

Cancer**

p21 GTPase p21 GTPase p21 GTPase EGF receptor growth factor receptor transcription factor transcription factor transcription factor antiapoptosis protein cyclin-D tryosine kinase cyclin-dependent kinase transcription factor growth factor p53 binding protein transcription factor transcription factor

pancreas, lung, colorectal endometrial, other carcinomas bladder gliomas, carcinomas breast, ovarian, gastric Burkitt's lymphoma, SCLC neuroblastoma, SCLC SCLC B-cell lymphoma B-cell lymphoma, carcinomas ALL (T cell), CML sarcoma melanoma, colorectal gastric sarcoma glioma, sarcoma ALL (T cell)

*Each oncogene must be activated by a mutation. Most are activated by one or more somatic mutations. Germ line mutations that activate oncogenes lead to familial cancers. **Abbreviations: EGF, epidermal growth factor; SCLC, small cell lung carcinoma; ALL, acute lymphocytic leukemia; CML, chronic myelogenous leukemia.

chimeric proteins are characterized by elevated protein kinase activity. If oncogenes are altered forms (or aberrantly expressed forms) of normal genes that usually participate in growth regulation, the number of different oncogenes should be fairly small. Thus, we should expect that if a large number of tumor viruses and tumors are screened for oncogenes, the same ones should appear repeatedly. This is indeed the case. For example, the oncogene in human bladder carcinoma (ras) has been found in human lung and colon carcinomas and in rat mammary carcinoma. A similar sequence but with a different base change was found in a human neuroblastoma and a fibrosarcoma. This variant was called N-ras. How oncogenes cause cellular transformation is unknown. However, when a tumor virus brings into a cell an oncogene whose sequence differs from the cellular protooncogene, the mutant gene product probably causes transformation because it is able to carry out some process that the proto-oncogene fails to do. That is, the protooncogene itself carries out a normal function or is silent; only a mutation or chromosomal rearrangement produces the cancerous cell. Certain biological functions are not determined simply by the presence of a particular gene product but by its

concentration. Thus, the conversion of a proto-oncogene to an active oncogene may occur simply by changes in concentration. The activity of a gene can be changed by altering the adjacent promoter or regulatory sequences (or both). Some oncogenes differ from the normal counterparts in that base changes exist only in the promoter and these changes presumably alter the rate of RNA synthesis. Another important mechanism by which the expression of a proto-oncogene can be altered is by moving the gene to a new location, e.g., by relocating it next to a different promoter, separating it from an adjacent regulatory element, or placing it adjacent to an enhancer. Thus, when the viral DNA is inserted into the chromosome of an infected cell, the viral oncogene may be expressed at a much greater rate than the proto-oncogene, which remains at its normal location. Whereas oncogenes are characterized by gain of function, another class of genes are characterized by loss of function; these are the tumor suppressor genes, w h i c h are often involved in familial (inherited) cancers (Table 26-7). The rare human cancer retinoblastoma provided the first significant insight into tumor suppressor genes. Retinoblastoma affects about 1 in 20,000 children who usually develop tumors in both eyes. Affected individuals have a small deletion in chromosome 13 that

612

CHAPTER26 Regulation of Gene Expression

TABLE 26-7

Some Tumor Suppressor Genes Involved in Human Cancers* Tumor Suppressor Gene Rb- 1 p53 p 16 APC MSH1, MLH2, PMS 1, PMS2 WT- 1 NF-1 VHL BRCA- 1 BRCA-2 PTEN DPC4 E-CAD DCC MEN- 1

Protein E2F1 binding transcription factor cyclin-dependent, kinase inhibitor regulates/3-catenin DNA mismatch repair transcription factor p2 lras-GTPase protein stability regulator ?DNA repair DNA repair tyrosine phosphastase TGF-/3 pathway transmembrane cell-cell adhesion molecule transmembrane receptor undetermined

Cancer retinoblastoma, osteosarcoma Li-Fraumeni syndrome, --50% of all cancers breast, lung, bladder, pancreas, --30% of all cancers familial adenomatous polyposis hereditary non-polyposis colorectal cancer Wilms' tumor melanoma, Neurofibromatosis type 1, neuroblastoma renal (clear cell), von Hippel Lindau syndrome ovarian, familial breast cancer familial breast cancer, pancreas breast, prostate, thyroid, glioma pancreas gastric, breast colorectal, other carcinomas parathyroid, adrenal, pituitary

*Mutations in tumor suppressor genes (many of which code for transcription factors) alter an essential cellular activity that regulates growth. Mutations occur both in somatic cells and in germ line cell and are often the cause of hereditary cancers.

predisposes mutations to occur in the homologous chromosome by mitotic recombination. Thus, in the majority of cases of retinoblastoma, one mutation is transmitted in the germline and the other arises as the result of a somatic mutation. It is estimated that 5-10% of breast cancers are due to germline mutations in the cancer susceptibility genes BRCA-1 and BRCA-2, which are also tumor suppressor genes. Carriers of the BRCA-I mutation have an 80-90% lifetime risk of developing breast cancer; carriers of the BRCA-2 mutation have a lesser risk. Ashkenazi Jews and the population of Iceland carry founder mutations since only one mutation, or a very few mutations, occurs repeatedly in the BRCA-1 and BRCA-2 genes among breast cancer patients in these populations. As a result, it is possible to genetically screen many individuals at high risk for harboring particular alleles of the B R C A - I and BRCA-2 genes. However, even with extensive counseling, learning that one is at high risk for developing breast cancer carries a heavy emotional burden. The only medical intervention offered at present for young, high-risk carriers of BRCA-1 or BRCA-2 genes is prophylactic mastectomy to avoid future development of breast cancer. Tumorigenesis is a complex, multistep process involving acquisition of a number of genetic lesions. Whereas

the mutations in oncogenes can cause unregulated growth, a deletion or a mutation in tumor suppressor genes (also called antioncogenes) can also predispose normal cells to become cancer cells. Thus, induction of a malignant phenotype involves a mutation in oncogenes, tumor suppressor genes, and perhaps other genes. In human colon cancer, defects in both oncogene and tumor suppressor genes are known. The latter have been assigned to chromosomes 5, 17, and 18.

26.7 Retroviruses and AIDS Since 1980 three types of infectious human T-cell lymphotrophic viruses (HTLVs) have been identified. HTLV-I is frequently associated with adult forms of T-cell leukemia-lymphoma, HTLV-II with hairy T-cell leukemia, and HTLV-III (now called human immunodeficiency virus; HIV) with acquired immunodeficiency syndrome (AIDS). Other related retroviruses have been isolated from human and primate populations. Molecular biologists have developed a wealth of information about the biology and genetics of retroviruses, including the complete sequencing of their genomes. Despite this enormous research effort it still

SupplementalReadingsandReferences

613

l

T Nucleocapsid-coreproteins l 5"-LTR

gag

Strong transcriptional activator

Promotes infectivity of cell-free virus

Binding sites for host transcription factors; tat response element

Regulator ofstructural . ~ gene expression

I

Reverse transcriptase, protease, integrase, and ribonuclease

LTR-3"

f env

Weak transcriptional activator

nef

[

repl'Fcati'oof n

Inhibits virus (negative factor) Viral coat proteins mediating

CD4binding and membrane fusion

FIGURE26-12 Genome of the human immunodeficiency virus (HIV, or the AIDS virus).

is not possible to prevent or cure infections by retroviruses. The well-characterized retroviruses, HTLV-I, HTLV-II, and HIV, all exhibit a strong affinity for T cells; HIV specifically recognizes a surface antigen (CD4) on T cells of the immune system. The organization of the HIV genome resembles that of the oncogenic RNA viruses described above but it is considerably more complex. In addition to the gag, pol, and env genes and LTR sequences characteristic of all retroviruses, the HIV genome encodes some additional functions (Figure 26-12). At least four regulatory proteins have been provisionally identified encoded by the vpr, tat, rev, and n e f genes. The envelope protein is synthesized as a single large polypeptide (gp 160), which is subsequently cleaved into two structural components. Most attempts at constructing an effective vaccine utilize part or all of the envelope proteins as antigens. Since these proteins do not elicit a strong antibody response by themselves, efforts are directed at coupling envelope antigens with more potent antigens to stimulate antibody response. The human retroviruses represent a unique class of infectious agents, and the mechanisms whereby they ultimately result in a variety of diseases are unknown. In comparison to infections caused by most other viruses, retroviruses are weak antigens. This failure to evoke a strong immune system response presumably enables them to become established and remain active in cells that they infect. They rarely produce a strong viremia and consequently may remain quiescent in infected individuals for years before clinical symptoms develop. Other yet-to-be-identified extrinsic or intrinsic factors also may be necessary to activate the expression and proliferation of human retroviruses. At present, the only protection against retroviral

infections is avoidance of exposure to these agents that are transmitted primarily through intercourse and exposure to contaminated blood.

Supplemental Readings and References A. A. Antson, E. J. Dodson, G. Dodson, et al.: Structure of the trp RNAbinding attenuation protein, TRAP, bound to RNA. Nature 401, 235 (1999). C. Brenner and G. Kroemer: Mitochondria--the death signal integrators. Science 289, 1150 (2000). L. C. Brody and B. B. Biesecker: Breast cancer susceptibility genes: BRCA1 and BRCA2. Medicine 77, 208 (1998). S. B. Cassidy and S. Schwartz: Prader-Willi and Angelman syndromes: Disorders of genomic imprinting. Medicine 77, 140 (1998). G. M. T. Cheetham, D. Jeruzalini, and T. A. Steitz: Structural basis for initiation of transcription from an RNA polymerase-promoter complex. Nature 399, 80 (1999). D. Chen, H. Ma, N. Hong, et al.: Regulation of transcription by a protein methyltransferase. Science 284, 2174 (1999). P. R. Cook: The organization of replication and transcription. Science 284, 1790 (1999). Z. Farfel, H. R. Bourne, and T. Iiri: The expanding spectrum of G protein diseases. New England Journal of Medicine 340, 1012 (1999). X. Y. Li, A. Virbasivs, and M. R. Green: Enhancement of TBP binding by activators and general transcription factors. Nature 399, 605 (1999). D. A. Haber and E. R. Fearon: The promise of cancer genetics. The Lancet 351, SII 1 (1998). E. Heard, P. Clerc, and P. Avner: X-chromosome inactivation in mammals. Annual Review of Genetics 31, 571 (1997). S. John and J. L. Workman: The facts of chromatin transcription. Science 282, 1836 (1998). J. O. Kahn and B. D. Walker: Acute human immunodeficiency virus type I infection. New England Journal of Medicine 339, 33 (1998). L. S. Honig and R. N. Rosenberg: Apoptosis and neurologic disease. American Journal of Medicine 108, 317 (2000). P. W. Laird and R. Jaenisch: The role of DNA methylation in cancer genetics and epigenetics. Annual Review of Genetics 30, 441 (1996).

61~ G. Larola, R. Cuesta, G. Brewer, et al.: Control of mRNA decay by heat shock-ubiquitin-proteasome pathway. Science 284, 499 (1999). D. S. Latchman: Transcription-factor mutations and disease. New England Journal of Medicine 334, 28 (1996). M. Li, V. D. Ona, C. Guegan, et al.: Functional role of caspase- 1 and caspase3 in an ALS transgenic mouse model. Science 288, 335 (2000). A. J. Lopez: Alternative splicing of pre-mRNA: Developmental consequences and mechanisms. Annual Review of Genetics 32, 279 (1998). R. G. Roeder: The role of general initiation factors in transcription by RNA polymerase II. Trends in Biochemical Sciences 21, 327 (1996). C. H. Schein: From housekeeper to microsurgeon: The diagnostic and therapeutic potential of ribonucleases. Nature Biotechnology 15, 529 (1997). R. Singal and G. D. Ginder: DNA methylation. Blood 93, 4059 (1999). V. Skulachev: Mitochondria in the programmed death phenomena;

CHAPTER26 Regulation of 6ene Expression a principle of biology: "It is better to die than to be wrong." IUBMB LIFE 49, 365 (2000). D. G. Tenen, R. Hromas, J. D. Licht, et al.: Transcription factors, normal myeloid development, and leukemia. Blood 90, 489 (1997). M-J. Tsai and B. W. O'Malley: Molecular mechanisms and action of steroid/thyroid receptor superfamily members. Annual Review of Biochemistry 63, 451 (1994). B. B. Wolf and D. R. Green: Suicidal tendencies: apoptotic cell death by caspase family proteinases. Journal of Biological Chemistry 274, 20049 (1999). J. L. Workman and R. E. Kingston: Alternation of nucleosome structure as a mechanism of transcriptional regulation. Annual Review of Biochemistry 67, 275 (1998). W. S. Yarnell and J. W. Roberts: Mechanism of intrinsic transcription termination and antitermination. Science 284, 611 (1999).

CHAPTER

27

Nucleotide Metabolism

A nucleotide consists of a purine or pyrimidine base, a pentose (or deoxypentose), and a phosphate. The synthesis and degradation of nucleotides are discussed in this chapter. Structural features of nucleotides were discussed in Chapter 23 and how nucleotides are used in the synthesis of DNA and RNA in Chapters 24 and 25, respectively. Table 27-1 gives the nomenclature of purine and pyrimidine nucleosides and nucleotides. Names of purine nucleosides end in -osine, whereas those of pyrimidine nucleosides end in -idine; guanine nucleoside is guanosine and should not be confused with guanidine, which is not a nucleic acid base; thymidine is a deoxyriboside. Nucleotides are synthesized by two types of metabolic pathways: de novo synthesis and salvage pathways. The former refers to synthesis of purines and pyrimidines from precursor molecules; the latter refers to the conversion of preformed purines and pyrimidines~derived from dietary sources, the surrounding medium, or nucleotide catabolism~to nucleotides, usually by addition of ribose5-phosphate to the base. De novo synthesis of purines is based on the metabolism of one-carbon compounds.

27.1 One-Carbon Metabolism One-carbon moieties of different redox states are utilized in the biosynthesis of the purine nucleotides and thymidine monophosphate (also known as thymidylate), the

metabolism of several amino acids (particularly serine and homocysteine), the initiation of protein biosynthesis in bacteria and mitochondria by formylation of methionine, and methylation of a variety of metabolites. These onecarbon reactions utilize coenzymes derived from folic acid, or folate. Folate is a vitamin for humans (and other animals because of their inability to synthesize it) (Chapter 38). Folic acid is the common name for pteroylglutamic acid, a compound consisting of a heterobicylic pteridine, p-aminobenzoic acid (PABA), and glutamic acid (Figure 27-1). The combination of the first two produces pteroic acid. Humans lack the enzymes capable of synthesizing PABA or of linking pteroic acid to glutamate. The antimicrobial activity of sulfonamides is due to their competitive inhibition of the bacterial enzyme that incorporates PABA into dihydropteroic acid (Chapter 6). Folates have a wide biological distribution; a rich dietary source is green leaves. They occur in nature largely as oligoglutamyl conjugates (e.g., in plants, predominantly pteroylheptaglutamate) in which the peptide linkages occur between the v-carboxyl group of one glutamate and the oe-amino group of the next (Figure 27-1). The mechanism of intestinal absorption of folate is not completely understood. The ingested folylpolyglutamates must be converted to folylmonoglutamate prior to absorption. The folylpolyglutamates are rapidly hydrolyzed in the intestines at neutral pH by the brush-border enzyme

615

616

CHAPTER 27

Nucleotide

Metabolism

TABLE 27-1

Nomenclature of Nucleosides and Nucleotides Base

Nucleoside (Base-Sugar)*

Nucleotide* (Base-Sugar Phosphate)

Purines Adenine (6-aminopurine) Guanine (2-amino-6-oxypurine) Hypoxanthine (6-oxypurine) Xanthine (2,6-dioxypurine)

Adenosine Deoxyadenosine Guanosine Deoxyguanosine Inosine Deoxyinosine Xanthosine

Adenosine monophosphate (AMP) or adenylic acid Deoxyadenosine monophosphate (dAMP) Guanosine monophosphate (GMP) Deoxyguanosine monophosphate (dGMP) Inosine monophosphate (IMP) Deoxyinosine monophosphate (dIMP) Xanthosine monophosphate (XMP)

Cytidine Deoxycytidine Thymidine (thymine deoxyriboside) Uridine

Cytidine monophosphate (CMP) Deoxycytidine monophosphate (dCMP) Thymidine monophosphate (TMP) (thymine deoxyribotide) Uridine monophosphate (UMP)

Pyrimidines Cytosine (2-oxy-4-aminopyrimidine) Thymine (2,4-dioxy-5-methylpyrimidine) Uracil (2,4-dioxypyrimidine)

*The sugar residue can be ribose or deoxyribose. If it is deoxyribose, it is identified as such; otherwise it is assumed to be ribose, with the exception of thymidine, which is deoxyriboside. tA nucleotide is a nucleoside monophosphate; the monophosphates are sometimes named as acids. pteroylpolyglutamate hydrolase (also called conjugase) to pteroylmonoglutamate (folic acid). If the folylpolyglutamates enter the intestinal epithelial cells intact, they may be converted to folylmonoglutamate within lysosomes by lysosomal hydrolase; however, the significance of this pathway appears to be minor. Folate transport in the intestine and the choroid plexus is mediated by a specific carrier, and the disorder hereditary folate malabsorption is associated with a defective folate carrier.

Individuals affected with hereditary folate malabsorption exhibit early onset of failure to thrive, megaloblastic anemia, and severe mental retardation. Therapy requires the administration of large doses of oral and systemic folates. Folate is reduced and converted to NS-methyltetrahydrofolate in intestines and secreted into the circulation. In the plasma, about two-thirds of the folate is proteinbound. Tissue needs for folate are met by uptake from

o N

!:

N

o

li

,,

~ N~

'

IOH

.....

~CH 2

."'-I

Pteridine'"""

7C--O-

'

I

residue

,

- - NH--CH

I

CH2

CH2

H 1~H2

CH,I

I

HNI~

II

c-o-

~ ~ - - N - - C H

I

CO--NH--

I

L

O II COO-I 9 PABA residue Glutamate AdditionalGlu residues residue

I .

.

.

.

Pteroyl residue

I

Pteroylmonoglutamate(folate) FIGURE 27-1 Structure of folic acid showingits components.The numberedpart participates in one-carbontransferreactions. In nature, folate occurs largelyas polyglutamylderivativesin which the glutamateresidues are attached by isopeptide linkages via the 7-carboxylgroup. The pteridine ring structureis also present in tetrahydrobiopterin,a coenzymein the hydroxylationof phenylalanine, tyrosine, and tryptophan(Chapter 17).

SECTION27.1 One-Carbon Metabolism

H N. N.. NyH

617 N H2N"',,,~ ~

[

II

NH2 OH

N ~

CH3 ~

I/

1

II

...coo I H

H

HN.

Methotrexate

Folate Dihydrofolate

0

\

N~ N NH2

L,,--NADPH + H+

reductase 1,,,~[,NADP+

H3CH2C ~'N

H

2

NH2

//, " ~ N H 2 H3CO~~! -OCH3 OCH3

OH

HN.

Pyrimethamine

Dihydrofolate

I f NADPH + H+

Dihydrofolate [ (

reductaseh

NADP+

H

~N..,.~ N~ / N ~ / H r~~N~CH ~ H OH

= I HN.

Tetrahydrofolate

FIGURE 27-2 Reduction of folate to tetrahydrofolate.

plasma. Within tissue cells, NS-methyltetrahydrofolate transfers its methyl group to homocysteine with the formation of methionine (Chapter 17). This reaction is catalyzed by homocysteine methyltransferase, a vitamin B 12 coenzyme-dependent enzyme, and appears to be the major site of interdependence of these two vitamins. In the tissues, tetrahydrofolate is converted to polyglutamyl forms by an ATP-dependent synthetase. In the liver, the major form is pteroyl pentaglutamate. Reduced polyglutamyl forms, each substituted with one of several one-carbon moieties, are the preferred coenzymes of folate-dependent enzymes. Reduction of folate (F) to tetrahydrofolate (FH4) occurs in two steps: F is reduced to 7,8-dihydrofolate (FH2), and FH2 is reduced to 5,6,7,8tetrahydrofolate (FH4). Both of these reactions are catalyzed by a single NADPH-linked enzyme, dihydrofolate reductase (Figure 27-2).

Inhibitors of Dihydrofolate Reductase Methotrexate (Figure 27-3), a structural analogue of FH2, is a potent inhibitor of dihydrofolate reductase and is used

Trimethoprim

F I G U R E 27-3 Inhibitors of dihydrofolate reductase. Methotrexate, a structural analogue of dihydrofolate, is effective against intact mammalian cells but ineffective against protozoa and some bacteria owing to permeability barriers. Trimethoprim and pyrimethamine (2,4-diaminopyrimidines) are effective against microorganisms. The former is antibacterial and antimalarial; the latter is primarily antimalarial.

in chemotherapy of neoplastic disease. Methotrexate is not effective against infections from bacteria and protozoa, since these organisms are impermeable to folate and its analogues. However, methotrexate inhibits dihydrofolate reductase of both bacterial and protozoal origin in cell-free preparations. Pyrimethamine is extremely effective against protozoan (e.g., malarial parasite) infections, is ineffective against bacterial infections., and is a mild inhibitor of the mammalian enzyme. Trimethoprim is an effective inhibitor of both bacterial and protozoal enzymes but has minimal inhibitory action against the mammalian enzyme. These selective enzyme inhibitors have been used in the treatment of bacterial and malarial infections.

Formation of One-Carbon Derivatives of Folate There are several one-carbon derivatives of folate (of different redox states) that function as one-carbon carriers in different metabolic processes. In all of these reactions, the one-carbon moiety is carried in a covalent linkage to one or both of the nitrogen atoms at the 5- and 10positions of the pteroic acid portion of tetrahydrofolate. Six known forms of carrier are shown in Figure 27-4. Folinic acid (NS-formyl FH4), also called leucovorin or citrovorum factor, is chemically stable and is used clinically to prevent or reverse the toxic effect of folate antimetabolites, such as methotrexate and pyrimethamine. The formation and interconversion of some metabolites of

618

CHAPTER27 Nucleotide Metabolism H

H2N

N

H

I

N

3. Hydrolysis of NS,Nl~ FH4 by a cyclohydrolase yields NS-formyl FH4 (folinic acid) and Nl~ FH4. The oxidation state of the one-carbon moiety is the same in all three species.

I

H2N~N~

N~

I

I I

1,0

Nl~

N~

H2C

FH4

"R

s lo

NS-MethylFH,

N ,N -MethyleneFH,

H20

H

H

I

H ~ ~,N ~

H2N~ ~ N ~ N ~

I

--H~o

/ /

N

N 5, N l~

FH4

" ~ ~ OH

I

jN~

O-"C

I

ff~

OH

I,o

H

R

H

I

I10

C--O/N~ [ H R H

H20"

N5-Formyl FH4

(citrovorumfactor)

4. Nl~ FH4 can be formed directly from formate in an ATP-requiring reaction.

?

H

I

H ~ ~"N ~ .,.,.,~N",~

H2N~N ~

N~ ,

FH4 -+- HCOO- -+- ATP OH

NS,N'~

II

HC~ N , ,

~X

NS-FormylFH,

FH,

H20

I,o

o.

~R

H

FH4

synthetase

>

NS-formyl FH4 + ADP + Pi

c=..

|

FH,

Nl~

Hj

~R

NS-FormiminoFH,

F I G U R E 27-4 The six one-carbon substituents of tetrahydrofolate. The oxidation state is the same in the one-carbon moiety of NS-formyl, Nl~ and NS,Nl~ FH4. Nl~ FH4 is required for de n o v o synthesis of purine nucleotides, whereas NS,Nl~ FH4 is needed for formation of thymidilic acid.

5. The formimino group (-CH=NH) of formiminoglutamate (Figlu), a catabolite of histidine (Chapter 17), forms formimino FH4. O O II II - O--C--CH2--CH2--CH--C--O-

+ FH4

)

Transferase

I NH

I HC--NH

FH4 are shown below.

Formiminoglutamate (Figlu) NS-Formimino FH4

1. Serine is the principal source of one-carbon units. FH4 + serine

+

serine hydroxymethyltransferase

o II

pyridoxal phosphate

NS,Nl~

FH4 -F glycine + H20

2. NS,Nl~ FH4 plays a central role, since it can be reduced to NS-methyl FH4 or oxidized to NS,Nl~ FH4. Red.

NS,Nl~

Ox

FH4 -k- NADH + H + reductase, NS-methyl FH4 + NAD + dehydrogenase

NS,Nl~ FH4 -Jr-NADP+ 5 10 N ,N -methenyl FH4 + NADP'H + H +

where "Ox." is oxidation and "Red." is reduction.

H

O II

- O--C--CH2--CH2--C--C--O-

I NH2 Glutamate

NS-Formiminoglutamate undergoes deamination to N 5, Nl~ FH4. If a loading dose of histidine is given to a patient who is deficient in folic acid, urinary excretion of Figlu is increased. This Figlu excretion test is useful in the diagnosis of folate deficiency, which clinically is manifested as a megaloblastic anemia (Chapter 38). The various catalytic roles of folate-mediated onecarbon transfer reactions in anabolic and catabolic

SECTION 27.2

Formation of 5-Phosphoribosyl-l-Pyrophosphate

619

Folic acid (F) NADPH + NADP +

FH, NADPH + NADP + FH,

ATP--~.--'~ HCOOH )

-~N5 -Formyl FH, ~ ATP ~

FH, + HCOO-

.

qO ~

rine biosynthesis

.....-- . . . .

glutamate Glutamate

FH,

Form used in .

l

N'~ ADP t +P, J H,O'~-'l

~ . , - , , - , - - . ~ . ~ + ' i'" r, NADPH T ' ~N"N' +' H+ "~

FH,-""

/

/-

Formiminoglycine / "--,.Glycine

NH3 FH' " ~

FH,

= N"F~176

FH~(~.Glutamat e

NADP+~*I dUMP TMP ~ Formiminoglutamate FH PyridO~xal p~sphate~-- N',N'~ FH, _t,._ 3= F.H, ~'FH, (Figlu),, { "x 7 t,,--'*NADH + H+ I,~--~ NADPH + H+ ' Serine Glycine/ [ + [ + Histidine CO,, N H 3 " ~ NADH + H+ [~"--*NAD ["---~NADP 9 ~,~,^n+

clne" / \ /

FH,

,~r,,.,

NS-MethylFH. I

FH. '

.. Lf~-Homocy steine I Vitamin u , , [ I ["---Methionine [ FH,

.]ReutilizedJ

FIGURE 27-5 One-carbontransferreactionsinvolvingfolate-derivedcarriers.Dashedarrowsindicatemultiple-stepreactionpathways; solid arrowsrepresentdirect, single-stepreactions.

reactions are shown in Figure 27-5. Serine, a nonessential amino acid, is the major source of one-carbon fragments. Some key processes that utilize folate-mediated one-carbon transfer reactions are as follows: 1. Synthesis of purine nucleotides (Nl~ FH4); 2. Methylation of deoxyuridylate to thymidylate (NS,Nl~ FH4) (an essential step in the biosynthesis of DNA; impairment of this reaction is responsible for the main clinical manifestations of folate deficiency); 3. Synthesis of formylmethionyl-tRNA (Nl~ FH4) required for initiation of protein synthesis in prokaryotes (Chapter 25) and in mitochondria; and 4. Conversion of homocysteine to methionine (NS-methyl FH4) (Chapter 17). Inherited disorders of folate transport and metabolism include defects in folate carrier (hereditary folate malabsorption, previously discussed), deficiency of NS,N l~ methylene FH4 reductase (Chapter 17), or functional deficiency of NS-methyl FH4 methyltransferase due to defects

of vitamin B12 metabolism (Chapter 38) or formiminotransferase (previously discussed). Several observational studies have shown that dietary supplementation of folate in women of childbearing age who might become pregnant can reduce the risk of fetal neural tube defects (e.g., spina bifida).

27.2 Formation of 5- Ph os p h o ri b osyl- 1 - Pyro p h osp h ate 5-Phosphoribosyl-l-pyrophosphate (PRPP) is a key intermediate in nucleotide biosynthesis. It is required for de n o v o synthesis of purine and pyrimidine nucleotides and the salvage pathways, in which purines are converted to their respective nucleotides via transfer of ribose 1-phosphate group from PRPP to the base; that is, Purine 4- P-ribose-P-P --+ purine-ribose-P 4- PPi (PRPP) PRPP is synthesized from ribose 5-phosphate by the following reaction:

620

CHAPTER27 Nucleotide Metabolism

_

2-O3POCH2

H

~

H

ATP

H

5-phosphate produced in this pathway is converted to ribose 5-phosphate as follows"

AMP

epimerase

PRPP synthetase

D-Xylulose 5-phosphate

H HO

OH

D-Ribulose 5-phosphate

2-O3POCH2

. .

H o

o

II II " ~~~J' O--P--O--P---OOH OH I I O-

O-

PRPP

In this reaction, the pyrophosphate group of ATP is transferred to ribose 5-phosphate; the product PRPP is a high-energy compound. PRPP synthetase has an absolute requirement for inorganic phosphate (Pi), which functions as an allosteric activator. The enzyme is inhibited by many nucleotides, the end products of the pathway for which PRPP is an essential substrate. The gene for PRPP synthetase is located on the X chromosome. Mutations in this gene have given rise to PRPP synthetase variants with increased catalytic activity, which leads to overproduction of uric acid (discussed below, under "Gout"). The ribose 5-phosphate needed comes from three sources:

1. The pentose phosphate pathway. This pathway is probably the major source of ribose 5-phosphate in liver and in bone marrow. 2. The uronic acid pathway (Chapter 15). Xylulose

>

The contribution of this pathway to the supply of ribose 5-phosphate may be a minor one. The epimerase and isomerase are the same enzymes that participate in the pentose phosphate pathway. 3. Glycolysis. Ribose 5-phosphate can also be produced from intermediates of glycolysis (Figure 27-6). The enzymes involved are those of the nonoxidative phase of the pentose phosphate pathway, that occur in many tissues.

27.3 Biosynthesis of Purine Nucleotides Purine nucleotides can be produced by two different pathways. The salvage pathway utilizes free purine bases and converts them to their respective ribonucleotides by appropriate phosphoribosyltransferases. The de novo pathway utilizes glutamine, glycine, aspartate, Nl~ FH4, bicarbonate, and PRPP in the synthesis of inosinic acid (IMP), which is then converted to AMP and GME

De Novo Synthesis The atoms of the purine ring originate from a wide variety of sources (Figure 27-7). C2 and C8 are derived from

Glucose

(Glycolysis)

/

P

,,

Fructose 6-phosphate + Glyceraldehyde3-phosphate J Transketolase 4-phosphate + Xylulose5-phosphate -~ ransaldolase

Sedoheptulose 7-phosphate + Glyceraldehyde3-phosphate I Transketolase Ribose 5-phosphate + Xylulose 5-phosphate Isomerase 2(Ribose 5-phosphate) -Sum:

isomerase

D-ribose 5-phosphate

Ribose 5-phosphate

~~[;se

>

Epimerase

92(Ribulose 5-phosphate)

2(Fructose6-phosphate) + Glyceraldehyde3-phosphate

= 3(Ribose5-phosphate)

FIGURE 27-6 Synthesis of ribose 5-phosphatefromintermediatesof glycolysisby enzymesof the nonoxidativephase of the pentose phosphate pathway.(See Chapter 15.)

SECTION27.3 Biosynthesis of Purine Nucleotides

621

co~

stepwise synthesis of IMP is depicted in Figure 27-8. The following features of purine biosynthesis should be noted.

Aspartate ~

r'~ ......... is ~-~-~Glycine "~'W--.N/g"~:, 'C ~ ~.~ .=. 11 is I i'"' 8c ~ N~~

1. The glycosidic bond is formed when the first atom of the purine ring is incorporated (reaction 1,

FH,

Figure 27-7). Free purines or purine nucleosides are not intermediates of this pathway. In pyrimidine biosynthesis, the pyrimidine ring is formed completely before addition of ribose 5-phosphate. 2. The biosynthesis is accomplished by several ATP-driven reactions. Ring formation (cyclization) is achieved by incorporating nucleophilic amino groups and electrophilic carbonyl groups at the appropriate positions. 3. The first purine nucleotide synthesized is inosinic acid (IMP), which is the precursor for the synthesis of

l, 1|

Amidonitrogenof91utamino F I G U R E 27-7 Sources of the atoms in d e n o v o synthesis of the purine ring. The numbered reactions correspond to the reaction steps in Figure 27-8.

Nl~ FH4. Glycine is incorporated entirely and provides C4, C5, and N7. The or-amino group of aspartic acid provides N1. The amide group of glutamine contributes N3 and N9. Carbon dioxide (or HCO~-) provides C6. The

/NH2

uc~. Glutamine

H20_.P

O

H2

|

O~P~OH OH

I

OH

No~Poc~O

Glycine ATP

OH

OH

ec-5-Phospho-D-ribosylI-pyrophosphate

OH

ON

13-5-PhosphoribosylI-amine

0

II

|176

H

II

~c..

c~

II

.c.....

OH

5"-Phosphoribosylglycinamide

~

.o.~'=a+~'oo C~NH

I P~R

PmR 5"-Phosphoribosyl5-amino-4-imidazole-

i P~R

5"-Phosphoribosyl5-aminoimidazole

r

5"-Phosphate-ribose

5"-Phosphoribosyl-o~-Nformylglycinamide OH

5"-Phosphodbosyl-a-Nformylglycinamidine

H,

II /C..,N II

O

II

/ (+~O) xCH

.c-..coo. c~\ I I

.

9

I

.~C-.c~ \

O

H2N/C~c/N~ H ~ / C ", ~ N /

N'~

~c. ~

P--R

O

I

... I Aspartic add

coo. ~[o ,;~ I

FH,

H

II

H=N/C~c/% L 2 H

.~...

C

.

(-H,O)

@

=

P--R

pmR

5"-Phosphoribosyl-5amino-4-imidazolesuccinocarboxamide

5"-Phosphoribosyl-5amino-4-imidazolecarboxamide

5"-Phosphoribosyl-5formamido-4-imidazolecarboxamide

F I G U R E 27-8 Biosynthesis of inosinic acid. The reactions are numbered. Enzymes: (1) amidophosphoribosyltransferase; (2) phosphoribosylglycinamide synthetase; (3) phosphoribosylglycinamide formyltransferase; (4) phosphoribosylformylglycinamidine synthetase; (5) phosphoribosylaminoimidazole synthetase; (6) phosphoribosylaminoimidazole carboxylase; (7) phosphoribosylaminoimidazolesuccinocarboxamide synthetase; (8) adenylosuccinate lyase; (9)phosphoribosylaminoimidazolecarboxamide formyltransferase; (10) IMP cyclohydrolase.

OH

OH

Inosine 5"-phosphate (inosinic acid)

622

CHAPTER27 Nucleotide Metabolism L -Aspartate

H O O C - - C m C - - COO H2 i--I......,:p

O

H

N %

NH3! "'"~~

/,

H -ooc--c-cmcoo H2 ...[........ NH ............

0

m

Fumarate

j

N

N/

H

H

i NH2i

-OOC~C--C~CO0-

"............

N

)

N

Adenylosuccinase

GTP, Mg2+ Adenylosuccinate [ synthetase Ribose 5-phosphate

N

I

Ribose 5-phosphate Adenylsuccinate (AMPS)

Inosine monophosphate

(IMP)

I

Ribose 5-phosphate Adenosine monophosphate (AMP)

NAD*K+H20 NADH~*"] IMP dehydr~

O " N

Glutamine

HN ::r.......J

O ~

Glutamate

,

H

I

iii~:~;

.N

y "~N"

Ribose 5-phosphate

Xanthosine monophosphate (XMP)

\ "N--

I

Ribose 5-phosphate Guanosine monophosphate (GMP)

FIGURE 27-9 Biosynthesis of AMP and GMP from IMP.

adenylic acid (AMP) and guanylic acid (GMP) in two different pathways (Figure 27-9). 4. All the required enzymes occur in the cytosol. This is also true for enzymes of salvage pathways, nucleotide interconversion, and degradation. 5. D e n o v o synthesis is particularly active in the liver and placenta. Nonhepatic tissues (e.g., bone marrow) depend on preformed purines that are synthesized in the liver and transported by red blood cells. They are very effective in salvaging the purines and exhibit little or no activity of xanthine oxidase, which oxidizes free purines. Salvage P a t h w a y s Reutilization of purine bases after conversion to their respective nucleotides constitutes salvage pathways. These pathways are particularly important in extrahepatic tissues. Purines arise from several sources: intermediary metabolism of nucleotides, degradation of polynucleotides, and dietary intake. Quantitatively, the first two sources are the more important. Salvage occurs mainly by the phosphoribosyltransferase reaction: Base + PRPP ~-- base-ribose-phosphate + PPi

Human tissue contains two phosphoribosyltransferases (Figure 27-10). Adenine phosphoribosyltransferase (APRT) catalyzes the formation of AMP from adenine. Hypoxanthine guanine phosphoribosyltransferase (HPRT) catalyzes the formation of IMP and GMP from hypoxanthine and guanine, respectively. HPRT also catalyzes the conversion of other purines (6-thiopurine, 8-azaguanine, allopurinol) to their respective ribonucleotides. The Keq of both phosphotransferases favors formation of ribonucleotides. During salvage, only two high-energy bonds are used, whereas in de n o v o synthesis of AMP or GMP, at least six high-energy bonds are expended. Deficiencies of these enzymes are discussed later. A quantitatively less significant salvage pathway uses purine nucleoside phosphorylase and nucleoside kinase: nucleoside phosphoryiase

Base + ribose l-phosphate < Base-ribose + ATP

> base-ribose 4- Pi

nucleoside kinase

> base-ribose-phosphate + ADP

The phosphorylase can catalyze the formation of inosine or deoxyinosine, and of guanosine or deoxyguanosine, but not adenosine or deoxyadenosine. However, the last twoi nucleosides can be converted to inosine and deoxyinosine by adenosine deaminase. The normal function of the phosphorylase appears to be the formation

SECTION27.3 Biosynthesisof PurineNucleotides

623 O

O

HN

PRPP

HN,~ I

PR

N

I H Hypoxanthine

0

_/I .~

"CH

~

I

cI

-O

cI

OH

HO IMP

Hypoxanthine-guanine phosphoribosyltransferase

O

O H N ~ % H2N~ N

I~N/~ H Guanine

H~N

PRPP

.,CH2 / 0

PR

HO__lpl~U

HC\H

i

H)CH

c

-O

cI

OH

HO

GMP NH2 NH2

PRPP

PP~ .

Adenine phosphoribosyltransferase H Adenine

0

I

-O

0

C

" C H

c

cI

OH

HO AMP

F I G U R E 27-10 Salvage pathways of purine nucleotide synthesis. The preformed purines can be converted to mononucleotides in a single step, using PRPP.

of free hypoxanthine and guanine for conversion to uric acid. Deficiency of adenosine deaminase or purine nucleoside phosphorylase results in immunodeficiency diseas (Chapter 35). In muscle, a unique nucleotide reutilization pathway, known as the purine nucleotide cycle, uses three enzymes: myoadenylate deaminase, adenylosuccinate synthetase, and adenylosuccinate lyase. In this cycle, AMP is converted to IMP with formation of NH3, and IMP is then reconverted to AME Myoadenylate deaminase deficiency produces a relatively benign disorder of muscle

(Chapter 21), which is characterized by muscle fatigue following exercise (see Myoadenylate Deaminase Deficiency). Nucleoside kinases specific for inosine or guanosine have been described. Adenosine kinase is widely distributed in mammalian tissues. Dietary Purines

Purines derived from food do not participate significantly in the salvage pathways described above; they are mostly

624

CHAPTER 27 Nucleotide Metabolism

NuCleO:)rotelns Proteolytic enzymes "'--* Amino acids Nucleic acids Nucleases and phosphodiesterases

polymerases, respectively. They are formed from the monophosphates in two stages. Conversion to diphosphates is catalyzed by kinases. These enzymes are basespecific but not sugar-specific. ATP is the usual source of phosphate; in some cases, other triphosphates or dATP may be used. Typical reactions are as follows" guanylate kinase

Nucleotides

GMP -t- ATP

~UFt Ph~ cleoside phosphorylase ibose 1-phosphate Free bases

Xanthine oxidase Uric acid (Excreted in urine, or catabolized by bacteria in the large intestine)

FIGURE 27-11 Fate of dietary nucleoproteins. Only the predominantreactions are shown. Dietary purines are mostlyconvertedto uric acid. converted to uric acid (Figure 27-11). Dietary nucleic acids, present predominantly as nucleoproteins, are converted in the small intestine to nucleic acids and protein digestive products by the action of proteolytic enzymes (Chapter 12). The liberated nucleic acids are depolymerized to nucleotides by pancreatic nucleases and phosphodiesterases. The nucleotides are hydrolyzed to nucleosides by nucleotidases and phosphatases. The nucleosides are either absorbed or further cleaved to free bases by nucleoside phosphorylase. The free bases may be oxidized to uric acid by xanthine oxidase. Nucleoside phosphorylase and xanthine oxidase are very active in human small intestinal mucosa. Uric acid produced in the small intestine may be absorbed and excreted in urine, or it may be further catabolized by bacteria in the large intestine. In experiments in which normal or gouty subjects ingested 15N-labeled nucleic acids, the labeled purines were converted to uric acid mainly by direct oxidative pathways without prior incorporation in tissue nucleic acids. Dietary purines (and pyrimidines; see below) do not serve significantly as precursors of cell nucleic acids in the body.

(dGDP)

adenylate kinase

~'--~Phosphate Nucleosides

> GDP -t- ADP

(dGMP)

Nucleotidases, phosphatases

AMP + A T P ,

, ADP + ADP

The diphosphates are converted to the triphosphates by the ubiquitous enzyme nucleoside diphosphate kinase. Remarkably, the lack of base or sugar specificity applies to the phosphate acceptor and the phosphate donor. Hence, the general reaction is nucleoside diphosphate kinase

dXDP 4- dYTP

(acceptor)

> 4- dXTP 4- dYDP

(donor)

Recall that conversion of ADP to ATP occurs mostly by mitochondrial oxidative phosphorylation coupled to electron transport (Chapter 14).

27.5 Formation of Purine Deoxyribonucleotides Conversion of ribonucleotides to the deoxy forms occurs exclusively at the diphosphate level. Ribonucleoside diphosphate reductase (ribonucleotide reductase) catalyzes the reaction. This enzyme is found in all species and tissues. The immediate source of reducing equivalents is the enzyme (E) itself in which two sulfhydryl groups are oxidized to a disulfide. The general reaction is ribonucleoside diphosphate

Ribonucleoside diphosphate + E(SH)2 reductase > deoxyribonucleoside diphosphate + E(S-S)+ H20 In this reaction, the T-hydroxyl group of ribose is replaced by a hydrogen, with retention of configuration at the 3'-carbon atom: 3-O6P2OCH2 .-.

Base

3-O6P2OCH2 _

2, OH

HI

OH

OH

~'~--~

Base

W1 H

Ribonucleotide~ ' ' j / reductase

27.4 Conversion of Nucleoside Monophosphates to Diphosphates and Triphosphates

f

,,,,

Thioredoxin-(SH)2

Thioredoxin-(S-S)

(red uced)

(oxidized)

Thioredoxin reductase

The triphosphates of nucleosides and deoxynucleosides are substrates for RNA polymerases and DNA

NADP+

\ NADPH + H+

+ H20

SECTION 27.6 Regulation of Purine Biosynthesis

Regeneration of the ribonucleotide reductase is accomplished in Escherichia coli and in mammals by thioredoxin, a dithiol polypeptide (M.W. 12,000) coenzyme, which also plays a role in other protein disulfide reductase reactions. In thioredoxin, two cysteine residues in the s e q u e n c e - C y s - G l y - P r o - C y s are converted to cystine. Reduced thioredoxin is regenerated by thioredoxin reductase, a flavoprotein enzyme that uses NADPH + H +. E. coli mutants unable to synthesize thioredoxin are still able to form deoxyribonucleotides. In these bacteria, a related substance, glutaredoxin, and two molecules of glutathione carry out the reduction. In Lactobacillus, the triphosphate is reduced and vitamin B12 is an essential coenzyme. Another example of this use of vitamin B12 is in Euglena, where the diphosphates are reduced. The mammalian system is nearly identical to that of E. coli.

Ribonucleotide reductase from E. coli consists of two subunits, B1 and B2, neither of which possesses catalytic function. B] is a dimer in which each monomer contains a substrate binding site and two types of allosteric effector binding sites. One type of effector site confers substrate specificity and the other is regulatory. B ] contains a pair of sulfhydryl groups that are required for catalytic activity. B2 also is a dimer, contains one nonheme Fe(III), and has an organic free- radical delocalized over the aromatic ring of a tyrosine residue in each of its polypeptide chains. The catalytic site is formed from the interaction of B1 and B2; a free-radical mechanism involves the tyrosyl residues, iron atom of B2, and sulfhydryl groups of B]. An antineoplastic agent, hydroxyurea, inhibits ribonucleotide reductase by inactivating the free radical. Ribonucleotide reductase is regulated so as to ensure a balanced supply of deoxynucleotides for DNA synthesis. For example, if excess dATP is present, decreased synthesis of all the deoxyribonucleotides ensues, whereas ATP stimulates formation of dCDP and dUDE Binding of TTP (also designated as dTTP) stimulates formation of dGDP and hence of dGTP; binding of dGTP stimulates the formation of dADP and hence of dATE In this way, these nucleotide effectors, by binding to various regulatory sites, tend to equalize the concentrations of the four deoxyribonucleotides required for DNA synthesis.

625

dophosphoribosyltransferase reaction, and the steps involved in the formation of AMP and GMP from IME

PRPP Synthetase Reaction PRPP synthetase requires inorganic phosphate as an allosteric activator. Its activity depends on intracellular concentrations of several end products of pathways in which PRPP is substrate. These end products are purine and pyrimidine nucleotides (Figure 27-12). Increased levels of intracellular PRPP enhance de novo purine biosynthesis. For example, in patients with HPRT deficiency, the fibroblasts show accelerated rates of purine formation. Several mutations of PRPP synthetase, which exhibit increased catalytic activity with increased production of PRPP, have been described in gouty subjects.

Amidophosphoribosyltransferase Reaction This reaction is the first and uniquely committed reaction of the de novo pathway and the rate-determining step. Amidophosphoribosyltransferase is an allosteric enzyme and has an absolute requirement for a divalent cation. The enzyme is inhibited by AMP and GMP, which bind at different sites. The enzyme also is inhibited by pyrimidine nucleotides at relatively high concentrations.

aibose 5-phosphate + ATP

e_A_.l-- e ............. ...... P R P P + Glutamine

"| @

5-Phosphodbosylamine

I

.....

,'. . . . . . . . I ! |

' I

of P u r i n e B i o s y n t h e s i s

Regulation of de novo purine biosynthesis is essential because it consumes a large amount of energy as well as of glycine, glutamine, N]~ FH4, and aspartate. Regulation occurs at the PRPP synthetase reaction, the ami-

l

S

......

AMP-S |

i

t

:::

~

--#,

, I

ADP

[

,,"

."

99 9

"

ATP

XMP

",.....t '" -'GMP [

AMP

!

Regulation

(Multiple steps)

IMP

-O

,- . . . . . .

27.6

|

'

9

GDP ........

I

9

I ! .

[

, 9 9

GTP

FIGURE 27-12 Feedback regulation of the d e n o v o pathway of purine biosynthesis. Solid lines represent metabolic pathways, and broken lines represent sites of feedback regulation. @, Stimulatory effect; Q, inhibitory effect. Regulatory enzymes: A, PRPP synthetase; B, amidophosphoribosyltransferase; C, adenylosuccinate synthetase; D, IMP dehydrogenase.

626

Inhibition by AMP and GMR is competitive with respect to PRPR The human placental enzyme exists in a small form (M.W. 133,000) and a large form (M.W. 270,000). The small form is catalytically active. Ribonucleotides convert the active form to the large form, whereas PRPP does the opposite. The regulatory actions of PRPP synthetase and amidophosphoribosyltransferase are coordinated. When there is a decrease in the intracellular concentration of adenine ribonucleotides, PRPP synthetase is activated; this results in increased synthesis of PRPP, which in turn converts the inactive form of amidophosphoribosyltransferase to the active form and increases production of purine nucleotides.

Regulation of Formation of AMP and G M P from IMP In the formation of AMP and GMP from IMR ATP is required for GTP synthesis, and GTP is needed to form ATP (Figure 27-9). In addition, adenylosuccinate synthetase is inhibited by AMP, and IMP dehydrogenase is inhibited by GMP (Figure 27-12).

27.7 Inhibitors of Purine Biosynthesis A variety of inhibitors of purine biosynthesis function at different stages and are used as antimicrobial, anticancer, and immunosuppressive agents.

CHAPTER27 Nucleotide Metabolism

acid (NS-formyl FH4), which decreases damage to bone marrow and prevents the development of leukopenia and thrombocytopenia. Methotrexate also produces mucositis, gastrointestinal symptoms, and liver damage by a direct toxic effect on hepatic cells. Chronic oral administration of methotrexate in psoriasis has been associated with an increased incidence of postnecrotic cirrhosis. The "rescue" of normal, but not tumor, cells from methotrexate toxicity by folinic acid is partly explained by differences in membrane transport. For example, osteogenic sarcoma cells (which do not respond to conventional doses of methotrexate treatment) are not rescued by folinic acid administered after methotrexate, presumably owing to the absence of transport sites for folinic acid in the neoplastic cells. The therapeutic effects of administration of methotrexate and "rescue" with folinic acid are superior to those of methotrexate alone. Resistance to methotrexate can develop from increased activity of dihydrofolate reductase, synthesis of an enzyme having a lower affinity for the inhibitor, decreased transport of the drug into tumor cells, decreased degradation of the reductase, and genetic amplification of the gene for dihydrofolate reductase. Azaserine (L-Serine diazoacetate), isolated from a species of Streptomyces, is a structural analogue of glutamine O § II - N= N~---CH~C~O~CH2~CH~COOH f I Azaserine NH2 O

lnhibitors of Folate Biosynthesis The sulfonamide drugs were the first effective antibacterial agents to be employed systemically in humans. These drugs resemble p-aminobenzoic acid in structure and inhibit utilization of that compound for the synthesis of folate in bacteria. Sulfonamides do not interfere with human metabolism.

Inhibitors of Formation of IMP

Folate analogues, such as methotrexate (Figure 27-3), are folate antagonists. They block production of FH2 and FH4 by dihydrofolate reductase and lead to diminished purine biosynthesis (inhibition of reactions 3 and 9 in Figure 27-8). Methotrexate also affects metabolism of amino acids and pyrimidine (inhibition of thymidylate synthesis) and inhibits DNA, RNA, and protein synthesis. It is effective in the treatment of breast cancer, cancer of the head and neck, choriocarcinoma, osteogenic sarcoma, and acute forms of leukemia. High doses of methotrexate can be tolerated provided that the patient also receives folinic

II

H2N-- C mCH 2mOll 2--CH ~COOH Glutamine

I

NH 2

that inhibits steps 1 and 4 of inosinic acid synthesis (Figure 27-8) and the conversion of XMP to GMP (Figure 27-9). A related substance, 6-diazo-5-oxo-Lnorleucine (DON), is also a glutamine analogue. Both compounds function as alkylating agents and become attached to an essential sulfhydryl group (a nucleophilic group) of the enzyme to form an inactive thioether derivative. The linkage occurs via the electrophilic carbon atom (designated by the arrow) of azaserine and release of N2.

Inhibitors of Formation of AMP and G M P

Hadacidin (N-formyl-N-hydroxyglycine) is an H

OH

I

.~--N--CH2uCOOH

SECTION27.7 Inhibitors of Purine Biosynthesis

627

analogue of aspartic acid isolated from fungi. By competing with aspartate, it inhibits adenylosuccinate synthetase and hence the synthesis of AMP, it is an experimental antineoplastic agent. Mycophenolic acid and ribavarin monophosphate inhibit IMP dehydrogenase and hence GMP synthesis.

I n h i b i t o r s of M u l t i p l e

S t e p s ~ P u r i n e Analogues

6-Mercaptopurine is a structural analogue of hypoxanthine and is converted to thio-IMP in the salvage pathway.

SH SH

6-Mercaptopurine is partially metabolized to 6-thiouric acid (which lacks antitumor activity) by xanthine oxidase. Allopurinol, a xanthine oxidase inhibitor used in the treatment of gout, potentiates the action of 6-mercaptopurine by preventing its conversion to 6-thiouric acid. This effect is taken into consideration in treatment. Mercaptopurines are also inactivated by S-methylation carried out by thiopurine S-methyltransferase (TPMT), particularly in hematopoietic tissues which lack xanthine oxidase. Deficiency of TPMT due to polymorphisms causes profound toxicity with the regular therapeutic regime. This is another example of the use of pharmacogenomics. Azathiopurine, a derivative of 6-mercaptopurine, functions as an antiproliferative agent. Its principal use is as an immunosuppressive agent. It presumably releases 6-mercaptopurine in the body by reacting with sulfhydryl compounds such as GSH. S

i'l

"N~CH3

N

~~

HPRT PRPP

pp,

i

2-O3POCH%o~

Thio-IMP N~~

I

H

i:-I~ ! OH

6-Mercaptopurine

O2N

I

H I OH

H Azathiopurine SH

N

Thio-IMP prevents production of AMP and GMP by inhibiting the following reactions" H2N

N ~~ t

Glutamine + PRPP

H 6-Thioguanine

| I I I

5-Phosphodbosylamine

,I

...|

,.

I Th,o-,MP I

/ IMP

XMP

Adenylosuccinate

I

i Ii

i Ii

- GMP

{

-'- AMP

6-Mercaptopurine is used in the treatment of acute leukemias. However, resistant tumor cells develop rapidly, probably because of altered specificity or lack of phosphoribosyltransferases, so that thio-IMP (the active inhibitor) is not formed. In support of this mechanism, resistant cells respond to 6-methylmercaptopurine ribonucleoside, which is phosphorylated to the corresponding nucleotide. Other mechanisms may include altered cell permeability and increased rate of destruction of 6-mercaptopurine.

6-Thioguanine is similar to 6-mercaptopurine in its action. The most active form is 6-thio-GMP, which inhibits guanylate kinase and, at higher concentrations, IMP dehydrogenase. Thio-IMP and thio-GMP also inhibit PRPP amidotransferase. Vidarabine (adenine arabinoside), an analogue of adenosine, does not interfere with purine biosynthesis but affects DNA synthesis. NH2

HOCH2 O/

ON N It is phosphorylated to adenine arabinoside triphosphate, which inhibits viral DNA polymerases, but not the

CHAPTER 27 NucleotideMetabolism

628

Acyclovir O HN~N~,

Valacyclovir O HN%N~

Ganciclovir

O H N ' ~I I IN

NZ

NH2 H2N"~N,"'~ N// H3C~ O _ _ _ . _ ] ~ O ~

HO~...A,..

H3C

HO- ~ 0 ~

0

OH

Penciclovir

Famciclovir

O

HN~N~, O H2N/'~N,,," ~ N HaN'~O

CHa

OH O F I G U R E 27-13 Structure of some of the purine nucleoside analogues used as antiviral agents.

mammalian enzymes by competition with dATE Originally developed as an antileukemia drug, it has proved useful in the treatment of herpes virus infections. Several other purine nucleoside analogues are used as antiviral agents (Figure 27-13); these include acyclovir, valacyclovir, ganciclovir, penciclovir, and famiciclovir. After conversion to their respective triphosphate derivatives, these drugs inhibit viral DNA polymerase. Viruses inhibited by these drugs are herpes simplex, varicella zoster, cytamegalovirus, and hepatitis B. Antiviral agents that are not purine nucleoside analogues are amantadine analogues and or-interferon.

Inhibition of Conversion of Ribonucleoside Diphosphate to Deoxyribonucleoside Diphosphate Hydroxyurea, an antineoplastic agent, acts by destroying an essential free radical in the active center of ribonucleotide reductase. 0 II

H2N~C~NHOH An unexpected finding of hydroxyurea treatment is induction of hemoglobin F (HbF) production in red blood cells. Hydroxyurea therapy results in an increase in both content and the number of red blood cells that contain HbF (known as F cells). This property of hydroxyurea has been

used in the treatment of sickle cell disease because of the antisickling effect of HbF (Chapter 28).

27.8 Catabolism of Purine Nucleotides Degradation of purines and their nucleotides occurs during turnover of endogenous nucleic acids and degradation of ingested nucleic acids (Figure 27-11), during which most of the purines are converted to uric acid. Degradation of purine nucleoside phosphates (AMP, IMP, GMP, and XMP) begins by hydrolysis by 5'-nucleotidase.(Figure 27-14) to produce adenosine, inosine, guanosine, and xanthosine, respectively, and phosphate. Adenosine is converted to inosine by adenosine deaminase (adenosine aminohydrolase). For inosine, guanosine, and xanthosine, the next step is catalyzed by purine nucleoside phosphorylase and involves a phosphorylation and a cleavage to produce ribose 1-phosphate and hypoxanthine, guanine, and xanthine, respectively. (The enzyme also acts on deoxyribonucleosides to release deoxyribose 1-phosphate.) The pentose sugars are metabolized further or excreted. Purine nucleoside phosphorylase functions in purine salvage, and deficiency of this enzyme results in decreased cell-mediated immunity. Hypoxanthine and guanine are converted to xanthine by xanthine oxidase and guanine aminohydrolase, respectively. Thus, all purine nucleosides produce xanthine. Xanthine oxidase is very active in the intestinal mucosa and the

629

SECTION27.8 Catabolismof Purine Nucleotides H20 NH3 AMP j

~/

H20 " ~ P,

GMP

H20 5'-Nucleotidase

~'--

)iMP

Adenylate aminohydrolase

I

Aden!.ln.

H20 ~/

NH3

H20

5'-Nucleotidase

Pi

H20 5'-Nucleottdase

Pi

, Ino.in.

I

A U .,IL

/ p ,

/

/

/

,,,~ Rlbose1-phosphate "XX~/ ,~, ~ / Purine -~ N -'-7ooc'eos~e HN/ " ~ ~ /phosphorylase

~

Ha" "~"" ~\

Hypoxanthlne

/

Guanine

Xanthine

o, +.,o

Xanthosine

p.~J ' "~Purine nucleoside . . . . . . ~'1 phosphorylase Hioose 1-pnospnate,P--"- I

i "~JPurine nucleoside JJl phosphorylase

5'-Nucleotidase

Pi

Guanoslne

Adenosine aminohydrolase p ~ J Ribose 1-phosphate , ~ t

XMP

/

/

~

~

H20

~

I

NH3 < " I

~

.,o,

J

/

i H

H

lanthine /

02 + H20 " ~ H202 ~

Xanthine oxidase

0

H

H

H

Uric acid

FIGURE 2"7-]4 Pathways for the formation of uric acid from purine nucleotides. [Reproduced with permission from G. Zubay, Biochemistry, 2nd ed. New York: (Macmillan 1988). @ 1988 by Macmillan Publishing Company.]

liver and is present in most tissues. The products of the xanthine oxidase reaction are uric acid and hydrogen peroxide. In primates (humans included) and birds, uric acid is excreted in the urine. In mammals other than primates, a liver enzyme, urate oxidase, converts uric acid to allantoin (Figure 27-15). In lower animals, allantoin is metabolized to allantoic acid (some fish), urea (most fish and amphibia), and ammonia (some marine invertebrates and crustaceans). In humans, uric acid, urea, and creatinine are the end products of nitrogen metabolism. All three are excreted in urine. The primary end product is urea (Chapter 17). Animals in which uric acid is the major end product excrete the rather insoluble uric acid as a semisolid mass, which allows for conservation of water. In humans, uric acid production and excretion is a balanced process. On a daily basis, about two-thirds of the uric acid produced is excreted by the kidneys and the remainder via intestinal

bacterial degradation. In chronic renal failure and hyperuricemia, extrarenal uric acid disposal is enhanced. Renal excretion of uric acid is a multicomponent bidirectional process involving glomerular filtration (100%) and proximal tubular reabsorption (98-100%), followed by proximal tubular secretion (50%) and proximal tubular reabsorption (40-44%). Ultimately, the renal clearance of uric acid is only about 7-10% of creatinine clearance (Chapter 17).

Xanthine Oxidase Reaction Xanthine oxidase is a flavoprotein that contains molybdenum, nonheme iron, and labile sulfur. The enzyme is present in two forms, one with dehydrogenase activity (xanthine dehydrogenase) and the other with oxidase activity. The former is converted to the latter by oxidation of

630

CHAPTER27 Nucleotide Metabolism O

Uric acid (keto form)

&N

Primates, birds uricotelic reptiles, insects (other than diptera)

"N _ _ O

t H

H

Orate oxidasel~H20' 02 O

H

H2N ~--~..~- N \

Allantoin

H

Mammals (otherthan primates), insects (diptera only)

H

Disorders of Purine Nucleotide Metabolism

Allantoinase ~ H20

COOH Allantoic acid

Some teleost fish

H

Gout

~'~Glyoxylic acid O

II

Most fishes, amphibia, some mollusks

2H2N--C-- NH2 U r e a s e ~ H20

Ammonia

Several disorders affect purine metabolism. They are gout and the syndromes associated with deficiency of HPRT, APRT, adenosine deaminase, nucleoside phosphorylase, myoadenylate deaminase, and xanthine oxidase.

H

fH,O

Allantoicase

Urea

which can cause tissue destruction. Thus, the xanthine oxidase reaction can cause tissue injury during reperfusion of organs that have been deprived of oxygen (e.g., ischemic myocardium, transplanted organs). The xanthine oxidase reaction is further promoted in hypoxic organs by providing increased substrates owing to the enhanced adenine nucleotide breakdown that occurs during oxygen deprivation. Hydrogen peroxide is inactivated by catalase or by peroxidase. Xanthine oxidase is inhibited by allopurinol (a purine analogue), which is used in the treatment of gout.

Some marine invertebrates, crustaceans

2NH3

F I G U R E 27-15 Metabolic degradation of uric acid in some animals.

thiol groups of the enzyme owing to the presence of high concentrations of oxygen. It is active on purines, aldehydes, and pteridines. The reactions catalyzed on purines are

Hypoxanthine + H20 +

0 2 --+

xanthine + H202

Gout is a heterogeneous group of genetic and acquired diseases characterized by elevated levels of urates in blood (hyperuricemia) and of uric acid in urine (urieuria). Hyperuricemia in men is defined as serum urate concentration greater than 7 mg/dL (420 #mol/L) and in women 6 mg/dL (357 #mol/L,). In gout, hyperuricemia is a common biochemical occurrence; however, many hyperuricemic subjects may not develop clinical gout (asymptomatic hyperuricemia). All clinical symptoms of gout arise from the low solubility of urate in biological fluids. The maximum solubility of urate in plasma at 37~ is about 7 mg/dL. However, in peripheral structures and in the extremities, where the temperature is well below 37~ the solubility of urate is decreased. When urate is present in supersaturated solutions, crystals of monosodium urate monohydrate form easily (Figure 27-16). Deposits of aggregated crystals, known as tophi, in and around joints of

and Xanthine + H20 +

0 2 ---> uric

OH

acid + H202

The reaction mechanism is incompletely understood. Molybdenum, an essential cofactor, is the initial acceptor of electrons during purine oxidation and undergoes reduction from Mo 6+ to Mo 4+. Deficiency of molybdenum can result in xanthinuria. The electrons from molybdenum are passed successively to the iron-sulfur center, to FAD, and finally to oxygen. The oxygen incorporated into xanthine and uric acid originates in water. Xanthine oxidase also yields the superoxide radical, O 2, which is then converted to hydrogen peroxide by superoxide dismutase (Chapter 14). This may yield free radicals,

H202-+-Fe z+ ---> Fe 3+ + OH- + OH"

O

'J...7 -o H Lactim form

H pK" = 5.75

H

Lactam form

pK'= 10.3 OH N

N

H Monosodium urate

F I G U R E 27-16 Structures of tautomeric forms of uric acid. In the lactim form, uric acid has two acidic hydrogens, with pK' values of 5.75 and 10.3. At physiological pH (7.4), the predominant species of the lactim form is urate monoanion.

SECTION 27.8

631

Catabolism of Purine Nucleotides

the extremities initiate an inflammatory foreign body reaction (acute arthritis) that involves leukocytes, complement, and other mediators (Chapter 35). This reaction causes severe pain, swelling, redness, and heat in the affected areas. Initial attacks are usually acute and frequently affect the metatarsophalangeal joint of one big toe. Tophi may also be present in subcutaneous tissues, cartilage, bone, and kidneys. Formation of urinary calculi (urolithiasis) of urate is common. Gout is potentially chronic and disfiguring. Primary gout is a disorder of purine metabolism seen predominantly in men. The condition is multifactorial and involves genetic and nongenetic factors. Occurrence in women is uncommon; when it does occur, it is usually found in postmenopausal women. The blood urate concentration of normal men is ~1 mg/dL higher than that in women, but this difference disappears after the menopause. Thus, in women, the postmenopausal rise in serum urate levels may increase the risk of developing gout. Gout is very rare in children and adolescents. Primary gout may be due to overproduction or underexcretion of uric acid or to a combination of both. Frequently, siblings and other close relatives of afflicted individuals have high levels of uric acid in blood but do not develop gout, indicating that hyperuricemia is not the only factor involved. Primary renal gout is due to underexcretion caused by a renal tubular defect in uric acid transport. Primary metabolic gout is due to overproduction of purines and uric acid. The prevalence of gout is high in some populations (e.g., 10% of adult male Maori of New Zealand). In Europe and the United States, the prevalence rate is 0.13-0.37%. Secondary gout develops as a complication of hyperuricemia caused by another disorder (e.g., leukemia, chronic nephritis, polycythemia). This type of hyperuricemia usually is associated with abnormally rapid turnover of nucleic acids. The rare cases of gout in adolescents and children are usually of this type.

Overproduction of PRPP The mechanism of the hyperuricemia in most individuals who have gout is unknown. Following is a discussion of biochemical lesions that lead to hyperuricemia and may eventually lead to gout. Enhanced PRPP synthesis results from X-chromosome-linked mutants of PRPP synthetase. Several variants show increased Vmax, resistance to feedback inhibition, or a low Km for ribose 5-phosphate. PRPP levels can also be increased as a result of underutilization in purine salvage pathways. Thus, HPRT deficiency (partial or complete) causes hyperuricemia as an X-linked recessive trait. In situations in which ATP is consumed more rapidly than it is synthesized or in which

ATP synthesis is impaired, ADP and AMP accumulate and eventually are converted to uric acid. Hyperuricemia can occur in hypoxic conditions (e.g., adult respiratory distress syndrome), glucose-6-phosphatase deficiency, and acute illness (e.g., severe hemorrhagic shock). Furthermore, hyperuricemia may serve as a marker for cellular energy crises. Ethanol ingestion causes hyperuricemia owing to increased degradation of ATP to AME The latter occurs in ethanol metabolism during the conversion of acetate to acetyl-CoA (Chapter 18). In all these instances of production of uric acid by xanthine oxidase, the formation of cytotoxic byproducts of the reaction, namely, hydrogen peroxide and superoxide radical, also is increased. Patients with glucose-6-phosphatase deficiency (glycogen storage disease type 1; see Chapter 15) exhibit hyperuricemia from infancy and some develop gout later in life. The hyperuricemia is due to decreased excretion and increased production of uric acid. Because of their hypoglycemia, these patients develop marked hyperlactic acidemia, and lactate reduces the renal clearance of uric acid by suppressing its tubular secretion. Renal excretion of urate is complex, comprising glomerular filtration, tubular reabsorption, and tubular secretion. The urate in the urine is thought to arise almost entirely by tubular secretion. The increased production of uric acid in glucose-6-phosphatase deficiency has been attributed to enhanced ATP turnover and consequent adenine nucleotide depletion, with release of feedback inhibition of amidophosphoribosyltransferase, and to acceleration of de novo purine nucleotide synthesis. Diminished levels of ATP and Pi result from the presence of increased levels of phosphorylated glycolytic intermediates (Chapter 15). A similar mechanism has been proposed for the hyperuricemia that results when fructose is administered intravenously to humans. Excessive production of organic acids leads to elevated levels of uric acid. Lactate, acetoacetate, and /3-hydroxybutyrate (the latter two are known as ketone bodies) compete with uric acid for secretion by the kidney tubules. Lactic acidemia can occur in glucose6-phosphatase deficiency and in alcohol ingestion. Ketonemia and ketonuria occur in untreated diabetes mellitus, starvation, glucose-6-phosphatase deficiency, etc. (Figure 27-17). Treatment

A variety of drugs are used in the management of gout in three clinical situations: 1. To treat acute gout arthritis, 2. To prevent acute attacks, and 3. To lower serum urate concentrations.

632

CHAPTER27 Nucleotide Metabolism Glycogen

From-] [ gluconeo- t--- Glucose 6-phosphate Pentose phosphate" Ribose 5-phosphate genesisj _[_(~ pathway [fATP Glucose

(~) - lkPRPP sythetase [ ' ' AMP PRPP + Gutamine Hypoxanthine-. Guanine

..'" . IMP

(~),-':"~"G

/ Hypoxanthine

L Urinary uric -acid

(~ ',

'[ 1AIIop urin~ Xanthine !-" inhibits [ ] these steps Uric acid

Enzyme (~ Glucose-6-phosphatase deficiency

Comments Leads to excess PRPP production enhanced A'FP turnover

(~) PRPP synthetase overactivity

Overproduction of PRPP

(~) Hypoxanthine-guanine phosphoribosyltransferase decrease or absence

Decreased purine salvage; conversion of bases to uric acid; enzyme is completely absent in Lesch-Nyhan syndrome

(~ Overproduction of organic acids

Decreased secretion of uric acid owing to competition from other organic acids in the kidneys

FIGURE 27-17 Compositediagram of the biochemical lesionsthat may lead to hyperuricemia. Acute gouty attacks commonly affect the first metatarsal joint of the foot. In aspirated joint fluids, birefringent urate crystals can be seen in the polarized light microscope, which is used in the definitive diagnosis. The treatment of acute gout includes the administration of colchicine, nonsteroidal anti-inflammatory drugs (NSAIDs), corticosteroids, adrenocorticotropic hormone (ACTH), and analgesics. Colchicine and NSAIDs also can be used prophylactically to prevent acute attacks in patients with gout. Drugs used to lower serum urate concentrations include probenecid, sulfinpyrazone, and allopurinol. Colchicine depolymerizes microtubules and structures (such as the mitotic spindle) consisting of microtubules. It is effective in decreasing pain and the frequency of attacks but its mechanism of action is obscure. Allopurinol, an analogue of hypoxanthine, inhibits xanthine oxidase and reduces formation of xanthine and uric acid. It is converted by xanthine oxidase to alloxanthine, which binds tightly to the active site by chelation with

Mo 4+. This greatly reduces reoxidation of Mo 4+ to Mo 6+ and affects catalytic activity (Figure 27-18). This type of inhibitor, in which a substrate analogue is converted to an inhibitor and not released from the active site, is known as a suicide enzyme-inactivator or a mechanismbased inhibitor (Chapter 6). Allopurinol also is converted to allopurinol ribonucleotide by HPRT. Uric acid production is decreased through depletion of PRPP. Allopurinol ribonucleotide also inhibits PRPP-amidotransferase allosterically. These effects are illustrated in Figure 27-19. Since xanthine is both a product and a substrate for xanthine oxidase, allopurinol therapy could be expected to cause accumulation of xanthine in the body. Since xanthine is only sparingly soluble in urine (but more soluble than uric acid), this could lead to urinary xanthine crystalluria, or stone formation. This complication has not been observed in patients who receive the drug for treatment of gout or uric acid stones, but it has occurred in a few patients with Lesch-Nyhan syndromeand lymphosarcoma.

SECTION27.8 Catabolism of Purine Nucleotides

633

OH

OH 8

N

~N ~N/

N

J N'~8

H

H

AIIopu ri nol

Hypo xant hi ne

Lesch-Nyhan Syndrome

Xanthine oxidase .-OH

oooO~176

Mo",+'''" N "'H'

H AIIoxanthin~

(chelating Mo at the active site of the enzyme)

FIGURE 27-18 Inhibition of xanthine oxidaseby allopurinol, an analogue of hypoxanthine. N7 and C8 of hypoxanthine are reversedin allopurinol. Allopurinol, a suicide enzymeinactivator,is convertedto alloxanthine, which binds tightly to the active site of the enzymevia M o 4+ and interrupts the reoxidation of M o 4+ to M o 6+ needed to initiate the next catalytic cycle.

Drugs that increase uric acid excretion in humans inwhich is effective in the regulation clude probener of hyperuricemia and the resolution and prevention of tophi, and sulfinpyrazone, which has similar effects. Both agents are weak organic acids and probably act as competitive inhibitors of tubular reabsorption of uric acid.

. r AIIoxanthine (suicide enzymeinactivator of xanthine oxidase)

AIIopurinol

PRPP Depletes the supply of PRPP needed for the first step of de novo purine nucleotide biosynthesis

Serum urate levels can be lowered by dietary and lifestyle changes. These include correction of obesity, avoidance of ethanol consumption, and avoidance of highpurine foods (e.g., organ meats). Psuedogout is a disorder caused by deposition of calcium pyrophosphate dihydrate usually in larger joints such as knee, wrist, and ankle.

AIIopurinol ribonucleotide Inhibits the first step of de novo purine nucleotide biosynthesis

FIGURE 27-19 Multiple actions of allopurinol inhibit formation of uric acid.

Lesch-Nyhan syndrome is characterized by virtual absence of HPRT, excessive production of uric acid, and abnormalities of the central nervous system. These abnormalities include mental retardation, spasticity (increased muscle tension resulting in continuous increase of resistance to stretching), choreoathetosis (characterized by irregular, jerky, or explosive involuntary movements, and writhing or squirming, which may involve any extremity or the trunk), and a compulsive form of self-mutilation. The disorder associated with partial deficiency of HPRT also leads to hyperuricemia but lacks the devastating neurological and behavioral features characteristic of the LeschNyhan syndrome. Both disorders are X-linked. The hyperuricemia in Lesch-Nyhan patients is explained, at least in part, on the basis of intracellular accumulation of PRPP leading to increased purine nucleotide biosynthesis de novo and increased production of uric acid. Such patients do not usually develop gouty arthritis early in life but do exhibit uric acid crystalluria and stone formation. In Lesch-Nyhan patients, all tissues are devoid of HPRT. The disorder thus can be detected by an assay for HPRT in erythrocytes and by cultured fibroblasts. The former test has been used in detection of the heterozygous state. HPRT is a 217-amino-acid cytosolic enzyme coded for by a single gene on the X chromosome. Several mutations of the HPRT gene are known (Table 27-2). Mutations include major gene alterations and missense mutations causing either gouty arthritis or LeschNyhan syndrome (Table 27-2). An example of a missense mutation causing Lesch-Nyhan syndrome is HPRTKinston, which has a substitution of asparagine for aspartic acid at position 194 (D194N). The mutant enzyme, although present in a concentration similar to that of the normal enzyme, is catalytically incompetent because of its very high Km values for hypoxanthine and PRPE The mechanism by which deficiency of HPRT causes central nervous system disorders remains unknown. Lesch-Nyhan patients do not show anatomical abnormalities in the brain. In normal subjects, HPRT activity is high in the brain and, in particular, in the basal ganglia where de novo purine biosynthesis is low. This suggests the importance of the purine salvage pathway in this tissue. However, the relationship between HPRT deficiency and

634

CHAPTER27 Nucleotide Metabolism

TABLE 27-2

Examples of Missense Mutations That Alter Human Hypoxanthine-Guanine Phosphoribsoyl-Transferase (HPRT) and Cause Lesch-Nyhan Syndrome (LN) or Gout

Mutant HPRT Kinston Urangan Mashad Montreal Toowong London Ann Arbor Tokyo Brisbane

Nucleotide change G--->A G--->A A--->T T--->C G--->A C--->T T--->G G--->A C--->T

neurological manifestations is not understood. The most significant abnormality identified in neurotransmitter systems is in the dopaminergic pathway (Chapter 32). There is no known treatment for the central nervous system dysfunction. Allopurinol has been used in the management of the hyperuricemia.

D194N G16S D20V M57T G58R S 110L I 132M G140D T168I

LN Gout Gout LN Gout Gout Gout LN Gout

also involved in conversion of guanosine and deoxyguanosine to guanine. In ADA or PNP deficiency, the appropriate substrates accumulate along with other alternative

NH2

~N APRT

N

Adenine P h o s p h o r i b o s y l t r a n s f e r a s e (APRT) Deficiency APRT isolated from erythrocytes is a dimer with each subunit having a molecular weight of 19,481: the gene is located on chromosome 16. This autosomal recessive trait results in inability to salvage adenine, which accumulates and is oxidized to 2,8-dihydroxyadenine by xanthine oxidase (Figure 27-20). The main clinical abnormality is the excretion of 2,8-dihydroxyadenine as insoluble material (gravel) in the urine. These stones can be confused with those of urate on routine analysis. Thus, appropriate chemical analyses are required, particularly in the pediatric age group, to identify APRT-deficient individuals. The neurological disorders characteristic of HPRT deficiency are not found in APRT deficiency, indicating that APRT may not play a significant role in the overall regulation of purine metabolism in humans. APRT deficiency is treated with a low-purine diet and allopurinol.

ADA deficiency and PNP deficiency are two autosomal recessive traits that cause immune system dysfunction. The reactions catalyzed by ADA and PNP are shown in Figure 27-21. Both enzymes function in conversion of adenosine and deoxyadenosine to hypoxanthine. PNP is

PRPP

N

N~

N

PP~

N

H

Adenine In the absence Xanthine of APRT oxidase

OH ~N

OH

Adenylic acid (AMP)

N

OH

N H 8-Hydroxyadenine Xanthine oxidase

~N N

N

[ Adenosine Deaminase (ADA) Deficiency and Purine Nucleoside Phosphorylase (PNP) Deficiency

Clinical presentation

Amino acid change

HO

~

OH

N> H 2,8-Dihydroxyadenine

FIGURE 27-20 Metabolic pathwaysfor the conversionof adenine. Adenineis normally salvaged by conversionto AMP by adeninephosphoribosyltransferase (APRT). In the absenceof APRT,adenineis oxidizedto the highly insoluble product 2,8-dihydroxyadenineby xanthineoxidase.

SECTIO27.8 N Catabolism of Purine Nucleotides

635

OH

H2o

NH3

,nosino

Adenosine

t/

,g~%

deaminase

Hyp

~N ~ N

.

OH H

2'-Deo~aOenosine

.

thine

,,,-

OH

H

Deo~inosine

o. .//'t'~/N. H2N" ~ N~

?

Punne

nucleoside

phosphorylase

P,

o.o.

H NO~ N>~ M2N I~N~N

HOC~ ~V- ~

Ribose-1-P04

N1% H2N"~

N ~

Guanine

Deoxyribose-l-PO4

OH H

Deoxyguanosine

FIGURE 27-21 Reactions catalyzedby adenosine deaminase (ADA) and purine nucleoside phosphorylase(PNP). ADA and PNP participate in the purine catabolic pathway,and deficiency of either leads to immunodeficiencydisease. [Slightly modified and reproduced, with permission, from N. M. Kredich and M. S. Hershfield, Immunodeficiencydiseases caused by adenosine deaminase and purine nucleoside phosphorylasedeficiency.In: The Metabolic Basis of Inherited Disease, 6th ed., C. S. Scriver,A. L. Beaudet, W. S. Sly, and D. Valle, Eds. New York: McGraw-Hill (1989).]

products to cause toxic effects on the cells of the immune system. Patients with ADA deficiency lack both T- and B-lymphocyte-mediated functions, namely, cellular and humoral immunity, respectively, and exhibit a severe combined immunodeficiency (SCID) disorder. Other genetic defects can cause SCID (Chapter 35), but ADA deficiency is responsible for about one-third of patients who have SCID. PNP deficiency is associated only with T-lymphocyte dysfunction. The exact mechanisms responsible for immune system dysfunction are not known. In ADA-deficient T lymphocytes, there is a 50- to 100-fold increase in the

concentration of dATP; similarly, PNP-deficient T lymphocytes exhibit elevation in dGTP concentration. Both dATP and dGTP inhibit ribonucleotide reductase, cause a reduction of other deoxyribonucleotides (e.g., dCTP) required for DNA synthesis, and inhibit cell division. S-Adenosylhomocysteine (SAH) also accumulates and inhibits many methylases required for normal functioning of the cell (Chapter 17). Accumulation of SAH results from the suicide inactivation of SAH hydrolase by deoxyadenosine. Formation and degradation of SAH are shown in Figure 27-22. In the hydrolysis of SAH, the substrate undergoes a temporary oxidation by NAD +, which is tightly coupled to the enzyme. This reaction eventually

636

CHAPTER27 Nucleotide Metabolism ATP~ E~P, +P, Methionine FH4~'',

NS-Methyl

FHj4.1

B12

~ S-Adenosylmethionine Methyl acceptorq Methyltransferas~ Transmethylation Methylated product ,,F I reaction I, S-Adenosylhomocysteine (SAH)

I[

SAH hydrolase

(deoxyadenosine inhibits this enzyme)

HomocIsteine + Adenosine

Cysteine F I G U R E 27-22 Formation and degradation of S-adenosylhomocysteine (SAH). In ADA deficiency, accumulation of deoxyadenosine, a suicide substrate, inhibits SAH hydrolase. This inhibition is accompanied by a higher steady-state level of SAH, which in turn inhibits several methyltransferase reactions. Normally, homocysteine is reconverted to methionine or cysteine.

leads to the hydrolysis of the substrate and to the reoxidation of NADH, (Figure 27-23). Deoxyadenosine binds to the enzyme and undergoes initial oxidation to a ketosugar intermediate, which is unstable and decomposes with elimination of adenine, leaving the enzyme in the reduced (NADH) state (Figure 27-23). The catalysis is stopped, and exogenous NAD+/NADH + H + cannot affect the reaction. Thus, inactivation of SAH hydrolase by conversion of enzyme-bound NAD + to NADH is a suicide inactivation, in which no covalent modification of the enzyme is involved. Immune system dysfunction in ADA deficiency has also been ascribed to the inhibition of pyrimidine nucleotide synthesis by adenosine, known as pyrimidine starvation. This may arise from inhibition of conversion of orotic acid to orotidine 5'-monophosphate or from inhibition of PRPP synthesis by excessive synthesis of adenine nucleotides. ADA deficiency causes death from massive infection before the patient reaches the age of 2 years. Some children with ADA or PNP deficiency have benefited from periodic infusions of irradiated erythrocytes (which contain ADA and PNP). Irradiation of erythrocytes is necessary to inactivate any white blood cells that may be present and thereby to reduce the risk of graft-versus-host disease (Chapter 35). PNP deficiency usually causes hypouricemia and hypouricosuria and excretion of inosine, guanosine, deoxyinosine, and deoxyguanosine. Another mode of enzyme replacement therapy is periodic administration of a polyethylene glycol-modified form of bovine intestinal ADA by intramuscular injection. The modified enzyme, which is prepared by conjugating polyethylene glycol with purified ADA (PEG-ADA), possesses a longer half-life as well as reduced immunogenicity. As a consequence of this treatment, the correction

of two biochemical abnormalities, namely, accumulation of toxic phosphorylated metabolites and inhibition of SAH hydrolase, preceded clinical improvement (i.e., the absence of infection and resumption of weight gain). Gene replacement therapy should be possible in both ADA and PNP deficiency, and such a trial was attempted with two patients having ADA deficiency. The patient's T cells were removed and a normal ADA gene was inserted into the T cells by means of a retroviral vector. The modified T cells were reintroduced into the patient's bloodstream. Patients were followed for clinical improvement while they continued to receive PEG-ADA treatment. No permanent cure was achieved.

Myoadenylate Deaminase Deficiency Myoadenylate deaminase (or AMP deaminase) deficiency is a relatively benign muscle disorder characterized by fatigue and exercise-induced muscle aches. This disorder is presumably inherited as an autosomal recessive trait. The relationship between the exercise-induced skeletal muscle dysfunction and AMP deaminase deficiency is explained by an interruption of the purine nucleotide cycle. The purine nucleotide cycle of muscle consists of the conversion of AMP --+ IMP --~ AMP and requires AMP deaminase, adenylosuccinate synthetase, and adenylosuccinate lyase (Figure 27-24). Flux through this cycle increases during exercise. Several mechanisms have been proposed to explain how the increase in flux is responsible for the maintenance of appropriate energy levels during exercise (Chapter 21 ). 1. During muscle contraction AMP deaminase activity increases. Nucleoside triphosphates are negative modulators, whereas nucleoside di- and monophosphates are positive modulators of the enzyme. The increased AMP deaminase activity prevents accumulation of AMP so that the adenylate kinase reaction favors the formation of ATP: ADP + ADP ~- ATP + AME 2. The NH3 produced in the AMP deaminase reaction and the decreased levels of ATP as a result of exercise stimulate phosphofructokinase to enhance the rate of glycolysis (Chapter 13). 3. The increased concentration of IMP may activate glycogen phosphorylase and further enhance glycolysis. 4. The production of fumarate (Figure 27-24; see also Chapter 13) may enhance the TCA cycle when demand for ATP production increases. 5. The formation of IMP may provide a means by which the intracellular purine nucleotide pool is maintained. AMP deaminase deficiency disrupts the purine

SECTION 27.9

Biosynthesisof Pyrimidine Nucleotides

FB I H2G. y / / O ' ~ Ad H"~H

OH

~/'H

Enzyme

C~H

~

S-AdenosyI-L-homocysteine

FB I H2G o ~ O ' ~ Ad RSH2C .y/'/"O" ~ Ad H O~/OH~Ad (H Enzyme - ~ ~ ~ / / "H Enzyme H-\ C H H OH ~ OH ~ C~H -- RSH

(SAH)

o.CO>,

HOH2C~

H20

637

H

OH

Enzyme ~

H

H /~H OH

Enzyme ~

H~~ OH

Enzyme ~-"

HOH2C ~O.~ Ad /~H H O OH

Deoxyadenosine

Ad = Adenine;

OH

Adenosine

(a)

HOH~I3THI~O H H~Ad H H I OH H

H

HOH2C O H~I~ ~ H ~

H

-t- Ad

07

Ketosugar (Unstable intermediate)

R=-O2C-CH- CHE- CH2"-" I NH3+

(b) F I G U R E 27-23 Mechanism of action of S-adenosylhomocysteine hydrolase (enzyme) and its inhibition by deoxyadenosine. (a) The enzyme uses enzyme-bound NAD + to temporarily oxidize substrate and eventually hydrolyze it to adenosine and homocysteine. [Reproduced with permission from R. H. Abeles, Suicide enzyme inactivators. Chem. Eng. News 61(38), 55 (September 19, 1983). @1983 by the American Chemical Society.] (b) Deoxyadenosine, a suicide substrate, is also oxidized by the enzyme with the formation of a ketosugar, which undergoes decomposition, with the product dissociating from the enzyme and leaving the enzyme in the reduced state (NADH).

nucleotide cycle and leads to muscle dysfunction during exercise; however, the cycle operates minimally at rest so that AMP deaminase deficiency should not cause muscle dysfunction during rest. Type

IIa (fast-twitch oxidative) and type I (slow-twitch oxidative) muscle fibers have greater oxidative capacity and are less dependent on the cycle than type IIb (fast-twitch glycolytic) fibers. Thus, gradual exercise programs that lead to production of a greater proportion of type IIa and type I fibers might improve exercise tolerance in AMP deaminase deficiency.

Xanthine Oxidase Deficiency This autosomal recessive trait results in hypouricemia and in increased urinary excretion of hypoxanthine and xanthine. Patients frequently have xanthine stones.

27.9 Biosynthesis of Pyrimidine Nucleotides

F I G U R E 27-24 Purine nucleotide. The cycle plays an important role in energy production in skeletal muscle during exercise.

Pyrimidine nucleotides, in common with purine nucleotides, are required for the synthesis of DNA and RNA. They also participate in intermediary metabolism. For example, pyrimidine nucleotides are involved in the biosynthesis of glycogen (Chapter 15) and of phospholipids

CHAPTER 27 NucleotideMetabolism

638 (Chapter 19). Biosynthesis of pyrimidine nucleotides can occur by a d e n o v o pathway or by the reutilization of preformed pyrimidine bases or ribonucleosides (salvage pathway).

Salvage Pathways Pyrimidines derived from dietary or endogenous sources are salvaged efficiently in mammalian systems. They are converted to nucleosides by nucleoside phosphorylases and then to nucleotides by appropriate kinases. Pyrimidine

/ Ribose5-phosphate4

A~ Phosphatem"

Phosphorylase

Pyrimidine-ribose (nucleoside)

ATP~ Kinase ADPr" ~ Pyrimidine-ribose-phosphate(nucleotide) UMP also can by synthesized from uracil and PRPP by uracil phosphoribosyltransferase. De Novo

Synthesis

The biosynthesis of pyrimidine nucleotides may be conveniently considered in two stages: the formation of uridine monophosphate (UMP) and the conversion of UMP to other pyrimidine nucleotides.

Formation of UMP The synthesis of UMP starts from glutamine, bicarbonate, and ATE and requires six enzyme activities. The sources of the atoms of the pyrimidine ring are shown in Figure 27-25, and the pathway is shown in Figure 27-26. 1. The pyrimidine base is formed first and then the nucleotide by the addition of ribose 5-phosphate from

From amide nitrogen of

glutamine

!

9

J i~--Fr~

[ i From C02

" .......

FIGURE 27-25 Sources of the pyrimidinering atomsin de novo biosynthesis.

PRPE In contrast, in de n o v o purine nucleotide biosynthesis, ribose 5-phosphate is an integral part of the earliest precursor molecule. 2. In the biosynthesis of both pyrimidine and urea (or arginine) (Chapter 17), carbamoyl phosphate is the source of carbon and nitrogen atoms. In pyrimidine biosynthesis, carbamoyl phosphate serves as donor of the carbamoyl group to aspartate with the formation of carbamoyl aspartate. In urea synthesis, the carbamoyl moiety of carbamoyl phosphate is transferred to ornithine, giving rise to citrulline. In eukaryotic cells, two separate pools of carbamoyl phosphate are synthesized by different enzymes located at different sites. Carbamoyl phosphate synthetase I (CPS I) is located in the inner membrane of mitochondria in the liver and, to lesser extent, in the kidneys and small intestine. It supplies carbamoyl phosphate for the urea cycle. CPS I is specific for ammonia as nitrogen donor and requires N-acetylglutamate as activator. Carbamoyl phosphate synthetase II (CPS II) is present in the cytosol. It supplies carbamoyl phosphate for pyrimidine nucleotide biosynthesis and uses the amido group of glutamine as nitrogen donor. The presence of physically separated CPSs in eukaryotes probably reflects the need for independent regulation of pyrimidine biosynthesis and urea formation, despite the fact that both pathways require carbamoyl phosphate. In prokaryotes, one CPS serves both pathways. 3. In mammalian tissue, the six enzymes are encoded by three genes. One gene codes for a multifunctional polypeptide (Pyr 1-3) that is located in the cytosol and has carbamoyl phosphate synthetase II (Figure 27-27), aspartate transcarbamoylase, and dihydroorotase activity. Each subunit of Pyr 1-3 has a molecular weight of 200,000-220,000, and the native enzyme exists as multiples of three subunits. The second gene codes for dihydroorotate dehydrogenase which is located on the outer side of the inner mitochondrial membrane. Dihydroorotate, the product of Pyr 1-3, passes freely through the outer mitochondrial membrane and converted to orotate. Orotate readily diffuses to the cytosol for conversion to UME The third gene codes for another multifunctional polypeptide known as UMP synthase (Pyr 5,6). Pyr 5,6 (M.W. 55,000) contains orotate phosphoribosyltransferase and orotidylate (orotidine-5'-monophosphate) decarboxylase activity. Use of multifunctional polypeptides is very efficient, since the intermediates neither accumulate nor become consumed in side reactions. They are

SECTION27.9 Biosynthesisof Pyrimidine Nucleotides

639

-OOC O

II ~_ = NHE--C--OPO3 +

ATP

(Z)

CO2 Glutamine

+2

-ooc.

(~ #

H2N

HC--NH3

:

~COOCarbamoyl

phosphate

I

O CH2

(~

HN -- ~.....~

I/H

O~.-~'C~N-/C'~co OH Carbarnoyl aspartate

Aspartate

I

tO

"N / COOH Dihydroorotate (~ AD+

O

O

HN

~" NADH + H+

HN

O

CO.~

'-O3PO

O

(~

COO- ~ P P

2-O3PO

O

HN

(~

oo-

H

H~I OH UMP

F H HO

H~ I

OH

I/H

Orotate

I

OH

Orotictine 5"-monophosphate

FIGURE 27-26 pathway of uridine-5'-monophosphate (UMP) synthesis. Enzymes: (1) carbamoyl phosphate synthetase II; (2) aspartate transcarbamoylase; (3)dihydroorotase; (4)dihydroorotate dehydrogenase; (5)orotate phosphoribosyltransferase; (6) orotidine-5'-monophosphate decarboxylase (orotidylate decarboxylase).

De novo

rapidly channeled without dissociation from the polypeptide. Other pathways in eukaryotic cells, such as fatty acid synthesis, occur on multifunctional polypeptides.

4. Conversion of dihydroorotate to orotate is catalyzed by dihydroorotate dehydrogenase, a metalloflavoprotein that contains nonheme-iron atoms and flavin adenine nucleotides (FMN and FAD). In this reaction, the electrons are probably transported via iron atoms and flavin nucleotides that are reoxidized by NAD +. 5. Biosynthesis of purine and pyrimidine nucleotides requires carbon dioxide and the amide nitrogen of glutamine. Both use an amino acid "nucleus"--glycine in purine biosynthesis and aspartate in pyrimidine biosynthesis. Both use PRPP as the source of ribose 1-phosphate. 6. UMP is converted to UTP by uridylate kinase and nucleoside diphosphate kinase. uridylate kinase

UMP + ATP .

, UDP § ADP

nucleoside diphosphate kinase

UDP + ATP. F I G U R E 27-27 Schematic representation of the intracellular location of the six enzymes of UMP biosynthesis in animals. Pyr 1-3 = 1, Carbamoyl phosphate synthetase II; 2, aspartate transcarbamoylase; 3, dihydroorotase 4, dihydroorotate dehydrogenase; Pyr 5,6 = 5, orotate phosphoribosyltransferase; 6, orotidine-5t-monophosphate decarboxylase.

, UDP + ADP

Formation of Other Pyrimidine Nucleotides UMP is the parent compound in the synthesis of cytidine and deoxycytidine phosphates and thymidine nucleotides (which are deoxyribonucleotides).

640

CHAPTER 27

Nucleotide Metabolism

FIGURE 27-28 Synthesisof thymidylicacid (TMP). Fluorodeoxyuridylateinhibitsconversionof dUMPto TMP,and methotrexate inhibits regenerationof the tetrahydrofolatecoenzyme.

Synthesis of Cytidine Nucleotides CTP is synthesized from UTP by transfer of the amide nitrogen of glutamine to C-4 of the pyrimidine ring of UTP. This reaction requires ATP as an energy source.

O

4- Ogp3OCH2

O

H

I

NH2

CTP synlhetase

~

4- Ogp3OCH2

O

curs exclusively by methylation of the C-5 of dUMP (Figure 27-28) by thymidylate synthase. The methylene group of NS,Nl~ FH4 is the source of the methyl group, and FH4 is oxidized to FH2. For sustained synthesis, FH4 must be regenerated by dihydrofolate reductase. Recall that deoxynucleotides are formed at the diphosphate level by ribonucleotide reductase; thus, UDP is converted to dUDP, then to dUTP, dUMP is then generated mainly by dUTPase.

H

H I HO

i

Synthesis of Thymidine Nucleotides De novo synthesis of thymidilic acid (TMP) oc-

OH

OH

i H OH

The deoxycytidine phosphates result from reduction of CDP to dCDP by a mechanism analogous to that described for the purine nucleotides. Then dCDP is converted to dCTP by nucleoside diphosphate kinase.

dUTPase

dUTP

> dUMP + PPi

The dUTPase reaction is very important because the DNA polymerases cannot distinguish dUTP from TTP and catalyze significant incorporation of dUMP into DNA when dUTP is present. Incorporation of the base U into DNA is not deleterious because all cells contain a uracil

SECTION 27.9

Biosynthesisof Pyrimidine Nucleotides

CDP

=dCDP .

-dCTP.

9DNA o

CTP

...'

NH~)]dld!Teap ~in.a'se

641 pyrimidines uracil and thymine (see page 643). The deficiency of the initial rate-limiting enzyme dihydropyrimidine dehydrogenase can cause adverse drug reactions in patients receiving standard chemotherapy. This is another use of pharmacogenomics.

[ TTPI

UDP '

"dUDP/-

[

dUTPase

TDP

--dCMP

-dUMP '

: TMP [

Thymidylate synthase

Thymidine

Thymine FIGURE 27-29 Pathwayfor TTP synthesis.The key intermediateis dUTP,whichis convertedto dUMPby dUTPase.

N-glycosylase that removes U from DNA. Figure 27-29 summarizes the pathway for TTP synthesis. Because there is only one pathway for synthesis of TMP, it can be used to synthesize radioactively labeled DNA or to inhibit DNA synthesis selectively. In bacterial thymidylate synthase mutants (thy-), DNA synthesis is not possible without added thymine or thymidine, both of which can be utilized by salvage pathways. Neither thymine nor thymidine engages in synthetic reactions other than production of TMP, so radioactive thymine or thymidine can serve as unique precursors for synthesis of radioactive DNA. The radioactivity usually is in the methyl group; since this group is not metabolized, the appearance of radioactivity in any other compound is avoided. Thymidylate synthase is competitively inhibited by fluorodeoxyuridylate (FUDRP), with formation of a stable ternary complex with methylene FH4. FUDR:P is generated by salvage from exogenous 5-fluorouracil (FU) or fluorodeoxyuridine (FUDR). FUDR is a useful drug in cancer chemotherapy because it inhibits TMP formation in proliferating cells. Thymidine nucleotide deficiency can also be induced by competitive inhibitors of dihydrofolate reductase, e.g., aminopterin (4-aminofolate) and methotrexate (4-amino-10-methylfolate; see Figure 27-3). Fluoropyrimidines are catabolized by enzymes that normally participate in the breakdown of endogeneous

Pyrimidine Analogues This group of compounds (Figure 27-30) includes several drugs that are clinically useful in the treatment of neoplastic disease, psoriasis, and infections caused by fungi and DNA-containing viruses. The mechanism of action of 5-fluorouracil, a halogenated pyrimidine, was mentioned above. An antifungal agent, flucytosine (5-fluorocytosine), acts through conversion to 5-fluorouracil by cytosine deaminase in the fungal cells. Idoxuridine (iododeoxyuridine), another halogenated pyrimidine derivative, is used in viral infections. It is a competitive inhibitor (via phosphorylated derivatives) of the incorporation of thymidylic acid into DNA. Cytarabine (cytosine arabinoside) is an analogue of 2'-deoxycytidine. The T-hydroxyl group of the arabinose moiety is in a trans position with respect to the 3'-hydroxyl group (instead of in a cis position, as in the ribose) and causes steric hindrance to rotation of the base around the nucleoside bond (Chapter 23). Phosphorylated derivatives of cytarabine inhibit nucleic acid synthesis as well as being incorporated into nucleic acids. 6-Azauridine and its triacetyl derivative, azaribine, inhibit pyrimidine biosynthesis. Azaribine is better absorbed than 6-azauridine, but it is converted in the blood to 6-azauridine, which undergoes intracellular transformation to 6-azauridylic acid (6-AzUMP). 6-AzUMP, a competitive inhibitor of orotidylate decarboxylase, blocks formation of UMP (Figure 27-26), resulting in high rates of excretion of orotic acid and orotidine in the urine. 5-Azacytidine, after conversion to a phosphorylated derivative, arrests the DNA synthesis phase of the cell cycle. It also affects methylation of certain bases, which leads to hypomethylation (Chapter 26). Azathymidine, 3'-azido-3'-deoxythymidine (AZT), after conversion to the corresponding 5'-triphosphate, inhibits viral reverse transcriptase. Hence, AZT is used in the treatment of AIDS (Chapter 25).

Regulation of de Novo Pyrimidine Biosynthesis Regulation of the de novo pathway is complex. In prokaryotic cells, aspartate transcarbamoylase, an allosteric protein, is inhibited by the end products of pyrimidine

642

CHAPTER27 Nucleotide Metabolism

(a) Inhibitors of de novo pyrimidine biosynthesis (by inhibiting orotidylate decarboxylase) O

O

N 6

Azauridine R = m O H O

II

Azaribine R= m O - - C - - C H

I

3

I

R

R

(b) Inhibitors of nucleic acid synthesis; also incorporated into nucleic acids NH2

O HN

N

jCII3

Cytarabine (cytosine arabinoside; 1-13-D-arabi nofurano sylcytosine)

HOG~jO

HOCUO

H ~ ....

H\I + ]

13"

12'

_

OH

H

N=N~N

(c) Inhibitors of thymidylate synthase O

O..,,../~ N j N H

H

3'-Azido3'-deoxythymidine (AZT)

, H

NH2 Flucytosine (5-fluorocytosine; converts to 5-FU)

5-Fluorouracil (5-FU) H

FIGURE 27-30 Pyrimidineanalogues.All of these compoundsrequire conversionto appropriatenucleotidesbefore they become active.

biosynthesis, in particular by CTP (Chapter 7). However, mammalian aspartate transcarbamoylase is not regulated in this manner, and carbamoyl phosphate synthetase II (CPS II) appears to be a primary regulatory site. CPS II is inhibited by UTP and is activated by PRPP and ATE Activation by ATP may be important in achieving a balanced synthesis of purine and pyrimidine nucleotides. PRPP is also an essential (and probably a rate-limiting) substrate for the orotate phosphoribosyltransferase reaction (reaction 5 in Figure 2726) promoting d e n o v o pyrimidine nucleotide synthesis by induction of Pyr 1-3. Another potential site of regulation is orotidine-5-phosphate decarboxylase, which is inhibited by UMP, CMP, allopurinol nucleotide, and oxypurinol.

27.10 Coordination of Purine and Pyrimidine Nucleotide Biosynthesis A balanced synthesis of pyrimidine and purine nucleotides is essential for the biosynthesis of nucleic acids in growing cells. Several mechanisms exist for coordinating nucleotide synthesis. 1. PRPP affects purine and pyrimidine nucleotide biosynthesis. PRPP formation is activated by inorganic phosphate and inhibited by several end products of pathways that use PRPE In the purine d e n o v o pathway, PRPP activates amidophosphoribosyltransferase and is the rate-limiting substrate for the enzyme. In purine

SECTION27.11 Catabolismof PyrimidineNucleotides Cytosine NH2 HN O

H

Uracil O

H20 3.3= Cytosine deaminase

643 Thymine O HN

..

"[l"

L.)

H

H

H+'NADPHfi Dihydropyrimidinep NADPH'H+ NADP~.~ dehydr~ ~NADP + O O HN

HN~"~

Dihydrouracil

CH3

O' ' ~ N)

Dihydrothymine

H H H20"~ Dihydropyrimidinase~/~H20 OOC -OOC [3-Ureidopropionate NH2 ~(~H2 NH2 ~CH --CH3 O,,,,~ N'/CIH2 [3-Ureidoisobutyrate (N-carbamoyl-13-alanine) O"" ~ " N/CH2 H H H'O~ [3-Ureidopropionase1/'-H20 NH,+ + CO, +

CO0 I

~H2

13-AlanineCH2 +NIH3

CO0

I + CO= + NH,+ CHCH3

! CH2 +N]H3 13-Aminoisobutyrate

FIGURE 27-31 Pathways for pyrimidine catabolism. The major end product from cytosine and uracil is/%alanine, from thymine it is /3-aminoisobutyrate.

salvage pathways, PRPP is the substrate for HPRT and APRT. In the pyrimidine pathway, PRPP activates CPS II, may induce Pyr 1-3, and is a rate-limiting substrate for orotate phosphoribosyltransferase. Thus, changes in the levels of PRPP bring about concordant changes, while enhanced utilization by one pathway might result in reciprocal changes in the other. This interrelationship between the synthesis of purine and pyrimidine nucleotides was seen in a patient who had a deficiency of PRPP synthetase and exhibited orotic aciduria and hypouricemia, consistent with decreased synthesis of purine and pyrimidine nucleotides. 2. Coordination of synthesis of purine and pyrimidine nucleotides is affected by activation of CPS II by ATR 3. Cultured mammalian cells (e.g., human lymphocytes) fail to grow when exposed to adenosine and have increased pools of ADR ATR and GTR decreased pools of UDR UTR and CTR and decreased

incorporation of [14C] orotic acid. These effects are reversed by exogenous uridine. These results suggest that adenosine inhibits the conversion of orotic acid to orotidine-5'-monophosphate. Adenosine deaminase reduces the toxic effect of adenosine by converting it to inosine. Increased levels of purine nucleotides may also inhibit PRPP synthesis. This inhibition of formation of pyrimidine nucleotides in the presence of excess purine nucleosides and nucleotides has been termed "pyrimidine starvation."

27.11

C a t a b o l i s m of P y r i m i d i n e N u c l e o t i d e s

Pyrimidine catabolism occurs mainly in the liver. In contrast to purine catabolism, pyrimidine catabolism yields highly soluble end products. Pyrimidine nucleotides are converted to nucleosides by 5'-nucleotidase.

644 Cytidine so formed is converted to uridine by cytidine aminohydrolase, while uridine and thymidine are converted to free bases by pyrimidine nucleoside phosphorylase. In mammalian systems, catabolism of uracil and thymine proceeds in parallel steps, catalyzed by the same enzymes (Figure 27-31). The rate-determining step is reduction to a 5,6-dihydroderivative by dihydropyrimidine dehydrogenase. In the second step, dihydropyrimidinase hydrolyzes cleavage of the dihydropyrimidine rings to/3ureido compounds. In the third step,/3-ureidopropionase hydrolyzes the /3-ureido compounds to/3-alanine or/3aminoisobutyrate (BAIB), with release of ammonia and carbon dioxide. Thus, the major end product of the catabolism of cytosine and uracil is/3-alanine, whereas that of thymine is BAIB. High concentrations of BAIB in urine follow excessive cell turnover or destruction (e.g., owing to leukemias or radiation therapy). High levels of BAIB excretion has been observed in some Asian families. Its significance is not known and the high excretors are otherwise normal. Degradation of/3-alanine and BAIB or their reutilization in various biosynthetic pathways is possible.

27.12 Abnormalities of Pyrimidine Metabolism Hereditary orotic aciduria is a rare autosomal recessive trait. In this disorder, both orotate phosphoribosyltransferase and orotidine-5'-phosphate decarboxylase activities (reactions 5 and 6 in Figure 27-26) are markedly deficient. Recall that these activities occur on the polypeptide Pyr 5,6. Orotic aciduria is characterized by failure of normal growth and by the presence of hypochromic erythrocytes and megaloblastic bone marrow, none of which are improved by the usual hematinic agents (e.g., iron, pyridoxine, vitamin B~2, and folate). Leukopenia is also present. Treatment with uridine (2-4 g/d) results in marked improvement in the hematological abnormalities, in growth and development, and in decreased excretion of orotic acid. These patients are pyrimidine auxotrophs and require an exogenous source of

CHAPTER27 Nucleotide Metabolism

pyrimidine just as all humans need vitamins, essential amino acids, and essential fatty acids. Deficiency of folate or vitamin B12 can cause hematological changes similar to hereditary orotic aciduria. Folate is directly involved in thymidylic acid synthesis and indirectly involved in vitamin B12 synthesis. Orotic aciduria without the characteristic hematological abnormalities occurs in disorders of the urea cycle that lead to accumulation of carbamoyl phosphate in mitochondria (e.g., ornithine transcarbamoylase deficiency; see Chapter 17). The carbamoyl phosphate exits from the mitochondria and augments cytosolic pyrimidine biosynthesis. Treatment with allopurinol or 6-azauridine also produces orotic aciduria as a result of inhibition of orotidine-5'phosphate decarboxylase by their metabolic products. Dihydropyrimidine dehydrogenase deficiency can cause unexpected toxic effects to 5-fluorouracil administration (see page 641).

Supplemental Readings and References H. H. Balfour, Jr.: Antiviral drugs. New England Journal of Medicine 340, 1255 (1999). R. Curto, E. O. Voit, and M. Cascante: Analysis of abnormalities in purine metabolism leading to gout and to neurological dysfunctions in man. Biochemical Journal 329, 477 (1998). B. T. Emmerson: The management of gout. New England Journal of Medicine 334, 445 (1996). M. D. Harris, L. B. Siegel, and J. A. Alloway: Gout and hyperuricemia. American Family Physician 59, 925 (1999). S. Haste, E. De Clercq, and J. Balzarini: Role of antimetabolites of purine and pyrimidine nucleotide metabolism in tumor cell differentiation. Biochemical Pharmacology 58, 539 (1999). W. L. Nyhan and D. F. Wong: New approaches to understanding LeschNyhan disease. New England Journal of Medicine 334, 1602 (1996). J. R. Pittman and M. H. Bross: Diagnosis and management of gout. American Family Physician 59, 1799 (1999). J. G. Puig, A. D. Michan, M. L. Jimenez, et al.: Clinical spectrum and uric acid metabolism. Archives oflnternal Medicine 151,726 (1991). D. Rush: Periconceptional folate and neural tube defect. American Journal of Clinical Nutrition 59(Suppl), 511 S (1994). A. Saven and L. Piro: Newer purine analogues for the treatment of hairy-cell leukemia. New England Journal of Medicine 330, 691 (1994). V. Serre, G. Hedeel, and X. Liu: Allosteric regulation and substrate channeling in multifunctional pyrimidine biosynthetic complexes: analysis of isolated domains and yeast-mammalian chemeric proteins. Journal of Molecular Biology 281, 363 (1998).

CHAPTER

28

Hemoglobin

The easy availability of blood has resulted in many studies of its constituents. Probably the most extensively studied component is hemoglobin, the predominant protein in the red blood cell and the molecule responsible for transporting oxygen, carbon dioxide, and protons between the lungs and tissues. The study of hemoglobin has led to a detailed knowledge of how oxygen and carbon dioxide transport is accomplished and regulated and has provided insight into the functioning of other allosteric proteins (Chapter 7). Hemoglobin is the first allosteric protein for which molecular details of allosteric effector binding and the mechanism of allosteric action are known. Studies of the genes coding for the globin polypeptide chains have provided a better understanding of many anemias and of the regulation of expression of other eukaryotic genes. Correction of the genetic defects in sickle cell anemia and thalassemia by the introduction of new genetic information into bone marrow cells (gene therapy) is being actively explored.

However, the differences in tertiary structure among them are critical to the functional properties of each subunit. Each globin subunit has associated with it, by noncovalent interaction, an Fe2+-porphyrin complex known as a heme group. Oxygen binding occurs at the heme iron. The predominant hemoglobin in adult erythrocytes is oe2f12, known as hemoglobin A1 (HbA). The structural and functional characteristics of hemoglobin have been worked out almost entirely through studies of HbA and its naturally occurring variants. Each tetramer has a molecular weight of about 64,500, and each oe-like and fl-like chain has a molecular weight of about 15,750 and 16,500, respectively. The subunits are situated at the corners of a tetrahedron (Figure 28-1). The structure changes slightly during the binding and release of oxygen. In a tetramer, dissimilar chains are more strongly joined than similar chains. In dilute solution, oxyhemoglobin dissociates as follows:

28.1 Structure of Hemoglobins Globin Chains Mammalian hemoglobins are tetramers made up of two oe-like subunits (usually or) and two non-oe subunits (usually 13, V, or 8). These subunits differ in primary structure but have similar secondary and tertiary structures.

There is no evidence for the formation of |174 or |174 although in the absence of ol-chains ~4 (HbH) forms. Association involves salt bridges, hydrogen bonds, van

645

646

CHAPTER28 Hemoglobin

Heme Group

(") F I G U R E 28-1 Arrangement of hemoglobin subunits. The four polypeptide subunits are located at the corners of a tetrahedron to give a roughly spherical structure.

der Waals bonds, and hydrophobic forces. About 60% of the contact between subunits does not change during reversible oxygen binding (packing contacts), while about 35% does (sliding contacts), otot and tiff contacts form about 5 % of the total intersubunit contacts, the remainder being olfl contacts. In deoxyhemoglobin, there is no tiff contact. This information was derived principally from x-ray crystallographic studies of Perutz, Kendrew, and coworkers and clarifies how 2,3-bisphosphoglyeerate (BPG) or (2,3-diphosphoglyeerate, 2,3-DPG) regulates oxygen transport (Note: 2,3-bisphosphoglycerate and 2,3-diphosphoglycerate are identical.) If each subunit is labeled, the dissociation in dilute solution becomes clearer:

@

Q(

QQ

It also shows the asymmetry in the tetramer, since the oe~/3 ~ contact differs from the Otlfl 2 contact, and O/2fl I differs from OtZfl 2. However, c~l/31 and Of2fl 2 contacts are similar, as are Otlfl2 and lYZfl I contacts. During oxygenation and deoxygenation, the c~1/31 and ffZf12 contacts do not change, but the O/lf12 and lYZfll c o n t a c t s do. Thus, the dimers or 1/3 l and lYZfl 2 function as a unit and can slide relative to each other. Stable o~/3 dimers can be present as a result of a mutation that prevents the formation of the tetramer. An example in which hemoglobin dissociates into stable dimers in the liganded state is the variant hemoglobin Rothschild, which is due to a mutation affecting the/3 chain (/337 Trp --~ Arg). Figures 28-2 and 28-3 are schematic diagrams of the secondary structures of o~ and/3 chains, respectively. Each chain contains helical and nonhelical segments and surrounds a heme group. Almost 80% of the amino acids exist in an o~-helical conformation.

Heine consists of a porphyrin ring system with an Fe 2+ fixed in the center through complexation to the nitrogens of four pyrrole rings. The porphyrin system is a nearly planar aromatic ring formed from four pyrrole rings linked by = C H - (methene) bridges. The pyrrole rings are substituted so that different porphyrins are distinguished by variations in their side chains (see also Chapters 14 and 29). The heine porphyrin is protoporphyrin IX. Iron has a coordination number of six, i.e., each atom of iron can bond with six electron pairs. In heme, two coordination positions of iron are not occupied by porphyrin nitrogens. When heine is associated with a globin chain, a histidyl nitrogen (from histidine FS, important in the allosteric mechanism) bonds with the fifth coordination position of iron, while the sixth position is open for combination with oxygen, water, carbon monoxide, or other ligands. Portions of the seven or eight helices of a globin chain form a hydrophobic crevice near the surface of the subunit. Heine lies in this crevice, between helices E and F. The low dielectric constant of the crevice prevents permanent oxidation of iron (from Fe 2+ to Fe 3+) by oxygen and is responsible for the reversible binding of oxygen. When Fe 2+ in solution combines with oxygen, it is oxidized to Fe 3+, which does not bind reversibly with oxygen. Evidence from electron paramagnetic resonance studies shows that, in oxyhemoglobin, the oxygen can be considered to oxidize the iron as long as the oxygen is bound. When the oxygen is released, Fe 3+ is again reduced to Fe 2+. Reversible 02 binding is a unique property of hemoglobin. If the Fe 2+ in hemoglobin is permanently oxidized to the ferric state (as in methemoglobin), the Fe 3+ binds tightly to a hydroxyl group, and oxygen will not bind. Heme groups can be removed from hemoglobin by dialysis, indicating that they are not covalently attached. Not counting the proximal histidine [His/392 (F8) or ot87 (F8)], about 60 amino acid residues come within 0.4 nm of one or more atoms of the heme group. This is approximately the maximal length of an effective hydrogen bond or hydrophobic interaction. Of these contacts, only one in the ot subunit and two in the/3 subunit are polar, which emphasizes the highly hydrophobic environment of the hemes. The three polar interactions involve the carboxyl groups of the propionic acid side chains on heme.

28.2 Functional Aspects of Hemoglobin Oxygen Transport The primary function of hemoglobin is to transport oxygen from the lungs to the tissues. Hemoglobin forms a

SECTION 28.2

FunctionalAspects of Hemoglobin

647

FIGURE 28-2 Secondary structure of the c~ chain of human hemoglobin. The helical regions (labeled A-H, after Kendrew), N and C termini, and the histidines located near the heine group are indicated. The axes of the B and C helices are indicated by dashed lines. Note that the o~ chain lacks helix D present in the fl chain. The amino acid residues are numbered by two different methods: from the N terminus of the polypeptide chain and from the N-terminal amino acid residue of each helix. Nonhelical regions are designated by the letters of helices at each end of a region.

dissociable complex with oxygen. Deoxyhemoglobin + 402 ~ oxyhemoglobin This reaction goes to the fight with an increase in oxygen pressure (as in the lungs) and to the left with a decrease in oxygen pressure (as in the tissues), in accordance with the law of mass action. Table 28-1 shows typical partial pressures of oxygen between the atmosphere and tissue mitochondria. Hemoglobin increases the solubility of oxygen in the blood 70-fold (from 0.3 to 20.3 mL of oxygen per milliliter of blood). In Figure 28-4, the oxygen saturation percent for hemoglobin is plotted against oxygen pressure in the gas above the surface of the hemoglobin solution. The curves, known as binding or dissociation curves, are often characterized by their Ps0 value, the oxygen pressure at which the hemoglobin is 50% saturated with oxygen. For

comparison, the dissociation curve for myoglobin is also shown. The hemoglobin curves are sigmoid (S-shaped), whereas the myoglobin curve is a rectangular hyperbola. This difference arises because hemoglobin is allosteric and shows cooperative oxygen binding kinetics (Chapter 7), whereas myoglobin is not allosteric. The binding of each molecule of oxygen to hemoglobin causes binding of additional oxygen molecules. This cooperativity occurs because hemoglobin is a tetramer. The cooperative binding of oxygen by hemoglobin is the basis for the regulation of oxygen and, indirectly, carbon dioxide levels in the body. The molecular understanding of this phenomenon is a major accomplishment of x-ray crystallographic studies of protein structure. Myoglobin is a heme protein found in high concentrations in red muscle, particularly cardiac muscle, where it functions as a storage site for oxygen. At the oxygen pressures found in

648

CHAPTER28 Hemoglobin

F I G U R E 28-3 Secondary structure of the/3 chain of human hemoglobin. The helical regions (labeled A-H, after Kendrew), N and C termini, and the histidines located near the heme group are indicated. The axes of the B, C, and D helices are indicated by dashed lines.

tissues, the dissociation curve for myoglobin lies to the left of the hemoglobin curves; consequently, hemoglobin (Hb) can readily oxygenate myoglobin (Mb). In the equation Hb(O2)4 -+- 4Mb ~- Hb + 4MbO2 the equilibrium lies far to the right. In the hemoglobin curves, the change from A to B to C is termed a rightward shift. The farther to the right a curve is shifted, the larger is the Ps0 value and the lower is the oxygen affinity. In this example, the shift is due to an increase in Pco2, since pH, temperature, and 2,3-DPG concentration were held constant. However, the shift could also have been caused by an increase in temperature (as in strenuous exercise or fever), a decrease in pH (as in acidosis or exercise), an increase in 2,3-DPG concentration (see below), or a combination of these variables. For

example, an increase in Pco2 is usually accompanied by a decrease in pH. Substances that cause a rightward shift are called negative allosteric effectorsor allosteric inhibitors

T A B I , E 28-1

Partial Pressures of Oxygen

POz(mm Hg) 158 100 95 40

GyAy HPFH

Hb KENYA GyAy(B/~)~ thai

---e J

INV~.RTED

Gy (8~)~ GY ( 8 ~ ) o thai

,(.--

l

(yS~) ~ thai (y8,8)~ thal X~Othal

F I G U R E 28-14 Chromosomal organization of the human/~-globin gene family and deletions that cause thalassemia (thal). In the/~-gene family, the exons are shown as dark boxes and the introns as open boxes. The various deletions are shown by horizontal bars under the/4-gene family. Cross-hatched regions indicate uncertainty in the termination points of the deletion. Arrows indicate unmapped end points of the deletion. [Reproduced with permission from R. A. Spritz and B. G. Forget, The thalassemias: molecular mechanisms of human genetic disease. Am. J. Hum. Genet. 35, 336 (1983). @ 1983 by the University of Chicago Press.]

Presumably, sequences somewhere in that region repress expression of the ygenes; removal of these sequences allows unrestrained expression of the y loci. If the deletion extends too close to the G• locus, however, expression of the G• gene is reduced, producing the more severe phenotype seen in G• Finally, in y-/3-thalassemia, the 6 locus and both g loci are deleted, while the/3 gene is normal; yet there is no synthesis of any of the/f-like globins. The reason for the nonexpression of this normal/3 locus is unknown. Thus, these disorders hint at the presence of regulatory information within the/3gene complex. It may eventually be possible to activate the ygenes to produce HPFH-like states and reduce the morbidity associated with/~-thalassemias and with mutations affecting the function or stability of the/3-globin chain. In normal adults, the small amount of HbF present is distributed nonuniformly among the erythrocytes. The distribution pattern was established by selective extraction of HbA from erythrocytes in a peripheral blood smear. The more poorly soluble HbF left behind stains pink with eosin. In most persons who have HPFH, all erythrocytes contain some HbE

Hemoglobinopathies The term hemoglobinopathies refers to hemoglobin disorders caused by normal synthesis of qualitatively abnormal globin chains. Transcription and translation of mutant genes usually proceed at a normal rate, but the products denature rapidly or function abnormally.

A selected list of mutant hemoglobins is given in Table 28-5. The most common mutations are the singleamino-acid substitutions caused by one nucleotide change in a globin gene. The table also lists several deletion mutations and one nonsense mutation. Globin chains that contain two mutations are extremely rare and appear to have resulted from a second mutation in a common mutant (usually HbS or HbC).

Molecular Pathology The effect of a mutation on the structure and function of hemoglobin is determined by the effects of the amino acid change. Many residues in hemoglobin have been conserved during evolution, probably because they are essential for normal hemoglobin function. The most critical regions for normal structure and function are the heme contacts and oelfll and Ofl~ 2 contacts. Substitutions near the heme group can cause weak or absent heme binding, resulting in loss of the heme group from the affected subunits (Hb Sydney, Hb Hammersmith). They can also stabilize the iron as Fe 3+ and prevent reversible oxygen binding (as in hemoglobin M). The heme group is also less tightly bound in methemoglobin than normal hemoglobin. Substitutions in amino acids involved in subunit contacts can have several effects. In Hb Philly, a Tyr ~ Phe change in the at ot 1/61 interface eliminates a hydrogen bond needed to stabilize deoxyhemoglobin (T-state) and leads to dissociation into monomers, which precipitate. This dissociation decreases cooperativity and increases oxygen

TABLE 28-5 Molecular and Clinical Characteristics of Some Abnormal Hemoglobins Position*

Amino Acid Change

t~-Globin Mutations Memphis 23(B4) Torino 43(CEI)

Glu---~Gln Phe---~Val

Hemoglobin

DNA Codon t Alterations

GAG---~CAG TTC---~GTC

Ann Arbor Chesapeake

80(F1) 92(FG4)

Leu---~Arg CTG---~CGG Arg---~Leu CGG-+CTG

G-Georgia

95(G2)

Pro-+Leu

CCG---~CTG

Leu---~Pro

CTG-+CCG

Glu-+Val Glu-+Ala Glu-+Lys

GAG---~GTG GAG-+GCG GAG-+AAG

Bibba 136(H19) /3-Globin Mutations S 6(A3) G-Makassar 6(A3) C 6(A3) Leiden

Glu---~---

GAG---~---

G-San Jose Saki Savannah E

6 or 7 (A3 or 4) 7(A4) 14(All) 24(B6) 26(B8)

Glu-+Gly Leu---~Pro Gly-+Val Glu-+Lys

GAG---~GCG CTG-+CCG GGT-+GTT GAG---~AAG

Genova S t. Louis

28(B 10) 28(B 10)

Leu-+Pro Leu---~Gln

CTG---~CCG CTG-+CAG

Tacoma Abraham Lincoln Philly

30(B 12) 32(B 14) 35(CI)

Arg-+Ser Leu---~Pro Tyr---~Phe

AGG-+AG(T,C) CTG-+CCG TAC---~TTC

Hammersmith

42(CD1)

Phe-+Ser

TTT-+TCT

Louisville

42(CD1)

Phe--~Leu

TTT-+TT(A,G) or TTC

Tochigi Bristol

56-59 deletion (D7-E3) 67(Ell) Val---~Asp

GTG-+GA(T,G)

Sydney

67(Ell)

Val-+Ala

GTG-+GGG

Seattle Shepherd's Bush

70(E 14) 74(E18)

Ala-+Asp Gly---~Asp

GCC-+GAC GGC-+GAC

Tours

87(F3)

Thr---~---

ACA-+---

Gun Hill

91-95

deletion

KOln Nottingham Kempsey

98(FG5) 98(FG5) 99(G 1)

Val---~Met Val---~Gly A sp---~Asn

GTG-+ATG GTG-+GGG G AT---~AAT

Remarks ~

Reduces sickling when present in HbS homozygotes. Decreases 0 2 affinity; unstable; mild anemia; decreases heme binding. Unstable. Increases 0 2 affinity; R state stabilized; a 1~ z -c~ mutant. Oxy form dissociates to dimers; decreases O 2 affinity; a 1/32-contact mutant. Dissociates. See discussion of sickle cell anemia in this chapter Benign; does not interact with HbS. Crystallizes in red blood cells; enhances sickling when heterozygous with HbS. Deletion decreases O 2 affinity; mild hemolysis; unstable. Compare to HBS; benign. Unstable; Pro disrupts a-helix. Unstable. Mild hemolytic anemia; increases subunit dissociation; normal O z affinity; a 1/31-contact mutant Increases O 2 affinity; unstable. Iron readily oxidized; polar group in heme pocket; increases 0 2 affinity; unstable. Normal O z affinity; decreases Bohr effect; unstable. Also called Perth; unstable. Loss of H bond needed to stabilize T state; high 0 2 affinity; unstable. Decreases O 2 affinity; unstable; poor heme binding severe anemia; inclusion bodies. Decreases 0 2 affinity; unstable; mild anemia; heme misoriented but not lost. Shortening of E-helix; unstable. Unstable; polar group in heme pocket; unusual codon needing 2 mutations. Unstable; mild hemolysis; inclusion bodies; poor heme binding. Unstable; mild hemolysis; decreases O 2 affinity. Unstable; increases 0 2 affinity; negative charge in DPG site; decreases DPG binding. Deletion shortens F-helix; unstable; increases 0 2 affinity. Deletion shortens F-helix; unstable; increases 0 2 affinity. Unstable; increases O z affinity; al/32-contact mutant. Unstable; increases 0 2 affinity; T-state destabilized. Increases 0 2 affinity; R-state stabilized.

(continued)

fi66 T A B L E 28-5

CHAPTER28 Hemoglobin

(Continued)

Hemoglobin

Position*

Amino Acid Change

Kansas Beth Israel Yoshizuka Presbyterian San Diego Peterborough Indianapolis D-Los Angeles

102(G4) 102(G4) 108(G10) 108(G10) 109(G11) 1 ll(G13) 112(G14) 121 (GH4)

O-Arab North Shore Syracuse

121 (GH4) 134(H12) 143(H21)

McKees Rocks

145(HC2)

Bethesda Cowtown

1145(HC2) 146(HC3)

DNA Codon r Alterations

Remarks ~

AAC-->ACC "[ Verylow 0 2 affinity; cyanosis. AAC---~AGC J AAC-->GAC '~ AAC--->AA(A,G) ~.Unstable; increases O 2 affinity; a 1/31-contact GTG---~ATG [ mutant. GTC---~TTC J TGT---~CGT Unstable. GAA---~CAA Also called D-Punjab; benign; enhances sickling in heterozygotes with HbS. As for HbD-LosAngeles. Glu---~Lys GAA---~AAA GTG---~GAG Unstable; mild hemolysis; normal O 2 affinity. Val---~Glu CAC---~CCC Increases O 2 affinity, decreases Bohr effect; loss of His--->Pro positive group at DPG binding site. Decreases DPG binding and Bohr effect; increases Tyr-+term. TAT---~TAA O2 affinity; deletes C-terminal residues; eliminates C-terminal bonding. TAT---~CAT "[ Increases O 2 affinity; decreases Bohr effect and Tyr--~His CAC---~CTC J DPG binding; disrupts C-terminal H bond, His---~Leu destabilizes T-state. Methemoglobinemia Asn---~Thr Asn-->Ser Asn---~Asp Asn-->Lys Val-->Met Val-->Phe Cys-->Arg Glu---~Gln

Mutations Causing Congenital o~-Chain: M-Boston 58(E7) Hi s---~Tyr CAC---~TAC M-Iwate 87(F8) Hi s----~Tyr CAC---~TAC /3-Chain: M-Freiburg 23(B5) Val---)--GTT---) .... M-Saskatoon 63 (E7) His---~Tyr C AT---~TAT M-Milwaukee 67(E11) Val---~Glu GTG---~GAG M-Hyde Park 92(F8) His---~Tyr CAC---~TAC

Only known in heterozygotes; all cause a benign cyanosis.

*The first number is the residue position, with 1 being the N-terminal amino acid; Kendrew's helical notation is given in parentheses. *Codon assignments are based on published sequences for human or- and [3-globin genes. Sequences of the alpha-1 and alpha-2 globin genes do not differ from each other at any of the codons used above. In a few instances, the normal codon could have mutated to either of two or three codons; these ambiguities are indicated by: term = termination codon; --- means codon (and amino acid) deleted. The codons shown here would occur on the nontranscribed strand in DNA. SBenign means no observable symptoms.

affinity. In hemoglobins Yoshizuka, Presbyterian, and Peterborough, the c~fl~ interface also is affected, but they exhibit decreased oxygen affinity because the R-state (oxyhemoglobin) is more destabilized than the T-state. Contacts at the O(lfl 2 interface stabilize the interaction of the two oefl dimers through the packing contacts and allow subunit motion during the T Z R transformation at the sliding contacts. Mutations in the /3 chains of Hb Kansas and Hb Beth Israel at this interface eliminate a hydrogen bond found only in the R-state, destabilizing the R-state and decreasing oxygen affinity and dissociation of the tetramer. The oxygen affinities of these variants are so low that they cause cyanosis, usually seen only in methemoglobinemia. Hb Chesapeake has an or-chain mutation

that affects the ot l f12 interface, stabilizing the R-state and increasing oxygen affinity. Other otlfl2 interface mutants, such as HbG Georgia, dissociate into dimers upon oxygenation and reassociate to tetramers when oxygen is removed. The deleterious effect of an amino acid substitution is due to replacement of a useful side chain by one that cannot perform the necessary function. The importance of a side chain may reside in its size, shape, or charge. A side chain that is too small (Hbs Torino, Hammersmith, and Sydney) or too large (Hbs Savannah and Peterborough) can be disruptive. Because 80% of the globin chain has a helical conformation, mutations that introduce proline residues are likely to disrupt this structure because proline

SECTION 28.3

Inherited Disorders of Hemoglobin Structure and Synthesis

cannot fit into an c~ helix (Chapter 4). The disruption can cause severe disease (Hbs B ibba, Abraham Lincoln, and Genova) or be benign (Hb Saki). If proline is introduced at one end of a helix, it does not disrupt it. However, in Hb Syracuse, new proline at the end of the H helix eliminates positive charge needed for 2,3-DPG binding. Loss of a charged or polar group (Hb Syracuse) or introduction of a charged or polar group into the hydrophobic interior of the molecule (Hbs Ann Arbor, St. Louis, North Shore, and Indianapolis) usually distorts the structure and causes altered function. If the mutation occurs at a site where the side chain is not very important or where only the type of side chain is important, the mutation may well be benign. A number of mutations of this type have been detected during routine screening in many populations.

Unstable Hemoglobins Many of the substitutions mentioned above adversely affect the stability of the tetrameric molecule and cause easy denaturation and precipitation within erythrocytes to form Heinz bodies. Deletion of one or more amino acids is also likely to cause instability, precipitation (e.g., Hbs Leiden, Tochigi, Tours, Gun Hill), and membrane damage. Consequently, these variants are associated with intravascular hemolysis, anemia, reticulocytosis, splenomegaly, and, in some patients, intermittent urobilinuria. If the hemoglobin precipitates soon after synthesis, before the cells leave the bone marrow, a thalassemialike syndrome may occur (Hbs Nottingham and Indianapolis). These mutations as well as Hbs Hammersmith, Abraham Lincoln, B ibba, and others produce a severe clinical disorder with massive hemolysis that is not improved by splenectomy. Other mutations (e.g., Hbs Torino, Ann Arbor, St. Louis, Koln, and Shepherd's Bush) in which precipitation occurs in the circulation cause severe hemolysis but are improved by splenectomy, which increases the erythrocyte lifetime. Although sometimes difficult, it is important to distinguish these two groups clinically when deciding whether splenectomy is indicated. Most unstable hemoglobins are associated with only mild hemolysis and occasional hemolytic crises, usually brought on by some stress such as a mild infection or treatment with sulfonamide or other oxidizing drugs (e.g., Hbs Philly, Sydney, North Shore, Leiden, Gun Hill, Seattle, and Louisville). Another group (e.g., Hbs Saki and Tacoma) is clinically benign, and the unstable hemoglobin is discovered incidentally by routine electrophoretic screening. The difference in disease severity among the unstable hemoglobins may reside in the number and time of

667

formation of Heinz bodies. Heinz body formation is believed to start when hemoglobin is oxidized to methemoglobin. The unstable methemoglobins are degraded to hemichromes, a process promoted by their tendency to dissociate (separate subunits form hemichromes more readily than tetramers). The tendency of the chains to form hemichromes decreases in the order c~ >/3 > y. Hemichromes precipitate and form Heinz bodies, which attach hydrophobically to the erythrocyte membrane, increasing membrane permeability and the rate of lysis. Loss of heme and oxidation of sulfhydryl groups do not appear to be necessary for precipitation. The degree of morbidity associated with an unstable hemoglobin is also determined by the effect of the mutation on oxygen affinity. If the oxygen affinity is decreased, delivery of oxygen to the tissues will be higher than normal and compensate, in part, for the lower hemoglobin concentration. This is the case with Hbs Leiden, Seattle, Louisville, and Peterborough. In contrast, Hbs K61n, St. Louis, and Shepherd's Bush, which cause severe hemolytic anemia and Heinz body formation, have increased oxygen affinity. The effect of the anemia is exacerbated by the inability of erythrocytes to unload the oxygen they carry. Both groups have similarly low hemoglobin levels and rapid hemolytic rates. Thus, the phenotype caused by a mutation is the net result of all its effects on hemoglobin structure and function.

Secondary Polycythemia Syndromes Mutations thatincrease oxygen affinity (reduce Ps0) can decrease tissue oxygenation more than expected on the basis of hemoglobin concentration. At low oxygen tension, production of erythropoietin is stimulated, causing a secondary polycythemia that increases oxygen delivery to the tissues and partially compensates for the hemoglobin abnormality. Any mutation that stabilizes the R (oxy) state (Hb Chesapeake) or destabilizes the T (deoxy) state (Hb Kempsey) can cause polycythemia. More than 28 such variants are known. Fourteen of these high-affinity hemoglobins have mutations at the 0/1,8 2 interface. Because the C terminus of the /3 chain supplies a hydrogen bond that is important for stabilizing the T-state, its replacement (Hb Bethesda), deletion (Hb McKees Rocks), or alteration in its environment (Hbs Syracuse and Cowtown) markedly decreases Ps0. Hb San Diego is unique in having high oxygen affinity due to a substitution in the 0/1/81 interface. Mutations that decrease the stability of the hemoglobin and cause precipitation or dissociation can also increase oxygen affinity.

668

Congenital Methemoglobinemias and Cyanosis Cyanosis is a bluishcoloration of the skin caused by the presence of more than 5 g of deoxyhemoglobin or methemoglobin per deciliter of blood in the subcutaneous capillary beds. It occurs in severe cardiopulmonary disease from poor oxygenation of normal hemoglobin or methemoglobin production. An extremely rare cause of cyanosis is the presence of a hemoglobin variant with very low oxygen affinity (e.g., Hb Kansas, Hb Beth Israel). Two groups of genetic defects can cause methemoglobinemia. 1. Defects in oxidoreductase in erythrocytes (methemoglobin reductase, glutathione reductase, glucose-6-phosphate dehydrogenase; Chapter 15). Patients who have these defects usually are not cyanotic unless subjected to oxidative stress (e.g., treatment with primaquine or sulfonamide). 2. Defects in which substitution of an amino acid in the region of the heme pocket increases the stability of Fe 3+ in the heme group and produces methemoglobin. Although heme oxidation occurs in several unstable hemoglobins, cyanosis is principally a manifestation of the Hbs M (Table 28-5). Since a single mutation can affect only one type of subunit (a or fi), half of each tetramer is normal. Most of these hemoglobins undergo reduced T-to-R transitions but with more difficulty. In addition, since only heterozygotes, for the Hbs M have been identified, half the tetramers are normal, and the carriers have no functional impairment. Cyanosis is visible because only 1.5-2 g of methemoglobin are needed to produce the same degree of discoloration as 5 g of deoxyhemoglobin. Diagnosis of patients with Hbs M is important because of the seriousness of the alternative causes of cyanosis.

Hemoglobin S and Sickling Disorders The most common, severe, and best studied hemoglobin mutation is HbS, which causes sickle cell anemia (or disease) in homozygous individuals and sickle cell trait in heterozygous individuals. The characteristic change in shape of erythrocytes from biconcave disk to curved and sicklelike occurs at low oxygen pressures. Carriers of sickle cell trait are asymptomatic, since their red cells do not sickle unless the Po2 drops below about 25 mm Hg; this happens only under unusual circumstances, such as unassisted breathing at high altitudes, some severe forms of pneumonia, and occasionally during anesthesia. The symptoms of sickle cell anemia are due to sequestration and destruction of the abnormal, sickled erythrocytes in the spleen and to the inability of many sickled

CHAPTER28 Hemoglobin

cells to pass through capillaries. The normal life span of 120 days for red blood cells is decreased to 10-12 days. Painful vaso-occlusive crises can occur anywhere in the body and are the major complication in sickle cell anemia. These crises are caused by blockage of capillaries in the affected tissue by the deformed red cells, which causes hemostasis, anoxia, further sickling, and eventually infarction. Increased adhesion of sickled erythrocytes to capillary endothelial cells may also contribute to capillary obstruction. Crises occur only when the circulation slows or hypoxia is present because the normal circulatory rate does not keep cells deoxygenated for the approximately 15 seconds required for sickling to begin. In contrast to the splenomegaly usually seen in chronic hemolytic anemias, a small, fibrous spleen is usually seen in adults with sickle cell anemia. This autosplenectomy is due to repeated infarction of the spleen. The clinical complications of sickle cell disease are highly variable. The severe forms of the disease occur in homozygous SS disease and S/fl~ and a milder disease occurs in double heterozygous SC disease and S/fl+-thalassemia. Patients who have a concurrent ot-thalassemia trait or high HbF levels have a mild course of the SS disease. The latter observation has been utilized in the pharmacological approaches that raise HbF levels (discussed later). Some of the clinical consequences in SS disease include megaloblastic erythropoiesis, aplastic crisis, stroke, bone pain crisis, proneness to infection particularly by Pneumococcus, Salmonella, and Haemophilus due to hyposplenism and acute chest syndrome. Prophylactic use of penicillin and antipneumococcal and Haemophilus vaccines has aided in the management of life-threatening infectious complications of SS disease. Neonatal screening has been used in the identification of infants with sickle cell disease so that risk of infection can be modulated by appropriate immunizations and penicillin prophylaxis. The acute chest syndrome characterized by chest pain is due to clogged pulmonary capillaries; in a small number of studies, patients have been treated with inhaled nitric oxide, which dilates blood vessels with clinical improvement. Sickle cell trait is present in about 8% of black Americans and to a much greater extent (as high as 45%) in some black African populations. The homozygous condition causes considerable morbidity and about 60,00080,000 deaths per year among African children. HbS also occurs in some parts of India, the Arabian region, and occasionally in the Mediterranean area. The HbS mutation that occurs in eastern Saudi Arabia and India, known as the Asian haplotype, has different flanking DNA sequences surrounding the fi-globin locus. Thus, the Asian haplotype may represent an independent occurrence of the HbS mutation that is distinct from its African counterpart. The deleterious gene likely has persisted in these

SECTION 28.3

669

Inherited Disorders of Hemoglobin Structure and Synthesis

populations because the HbS that increases resistance to malaria caused by Plasmodium falciparum, which was, until recently, endemic in those areas. A similar explanation has been advanced for the high frequencies of /~-thalassemia and single or-locus genotypes in the same regions. The biological basis of resistance to malaria has been established in laboratory experiments. As schizonts of P. falciparum grow in erythrocytes, they lower the intracellular pH and generate hydrogen peroxide. The lower pH promotes sickling and the hydrogen peroxide damages cell membranes of thalassemic erythrocytes. In both cases, the erythrocyte membranes become more permeable to potassium ions; the resulting decrease in intracellular potassium kills the parasites. The mutation in HbS replaces glutamic acid (a polar amino acid) by valine (a nonpolar residue) at position 6 of the/~ chains. The solubilities of oxy- and deoxy-HbA and oxy-HbS are similar, being about 50 times that of deoxy-HbS. Upon deoxygenation, HbS precipitates, forming long, rigid, polymeric strands that distort and stiffen the red cells. The very high concentration of hemoglobin in the erythrocyte (340 mg/mL), giving an average intermolecular distance of about 1 nm (10 A), minimizes the time necessary for precipitations to occur. Dilution of ribS, as in sickle cell trait (HbS/HbA heterozygotes), reduces the concentration below the point at which sickling readily occurs. Similarly, the mildness of homozygous HbS disease in populations such as those of eastern Saudi Arabia and Orissa, India is attributed to an accompanying elevation of HbF, which reduces the effective concentration of HbS tetramers. Other abnormal hemoglobins can interact with HbS and alter the course of the disease. The most common is HbC, which has a lysine in place of glutamic acid, also at/~6. The gene has a frequency second only to that of HbS in black Americans and in some black African populations. Persons heterozygotic for HbC are asymptomatic, but homozygotic individuals have a mild hemolytic anemia with splenomegaly. Because of its insolubility, crystals of HbC can sometimes be seen in peripheral blood smears from homozygous individuals. As a result of the coincidental distribution of the genes for HbS and HbC, heterozygotes for both hemoglobins are not uncommon. HbSC disease has a severity intermediate between that seen in persons homozygotic for HbS and those homozygotic for HbC. Unlike HbA, HbC copolymerizes with HbS. In contrast, replacement of the/~6 glutamic acid by alanine (HbG-Makassar) or deletion of/36 (Hb Leiden) results in hemoglobin that neither precipitates nor interacts with HbS. Hb San Jose (Glu --+ Gly at residue/37) is a harmless variant. Individuals heterozygous for HbS and HbO Arab or HbD Los Angeles have a hemolytic anemia of a severity

intermediate between that of HbSC disease and sickle cell anemia. Certain or-chain variants (e.g., Hb Memphis), when present in HbS homozygotes, can ameliorate the clinical course of the disease. The severity of HbS-/~thalassemia depends on whether the thalassemia is/3 ~ or /~+ and, if it is/3 +, on how much normal/~ chain is synthesized. Severity also depends on the HbF concentration. A patient with heterozygosity for both HbS and Hb Quebec-chori exhibited clinical symptoms suggestive of sickle cell disease. Hb Quebec-chori, an electrophoretically silent variant at acid and alkaline pH (see Appendix VII) (/~87 Thr --+ Ile), polymerizes with HbS with the stabilization of the polymer under hypoxic conditions leading to sickling of red blood cells. Thus, Hb Quebec-chori provides an example of a hemoglobin that has the potential to polymerize with HbS and causing sickle cell disease in a sickle cell trait condition which is otherwise benign by itself. Determination of the structure of crystalline HbS has shown that in the/3 subunits of oxy-HbA and oxy-HbS, a "hydrophobic pocket" between helices E and F is closed; this opens in the deoxy form. In HbA, the residues at the surfaces of the globin subunits are hydrophilic (polar) and do not interact with this pocket. In HbS, however, the/36 valine is hydrophobic and fits into the hydrophobic pocket (formed by leucine and phenylalanine at/~85 and/388) of an adjacent/3 chain to form a stable structure. Since each /~ subunit in deoxy-HbS has an "acceptor" hydrophobic pocket and a "donor" valine, linear aggregates form (Figure 28-15). Understanding of the sickling process and of the structure of the HbS polymer provides a rational basis for ways of correcting the molecular defect. Thus, dilution of the HbS in the red cells, blockage of the interaction of the /36 valine with the hydrophobic pocket, and decrease of

F I G U R E 28-15

Structure of hemoglobin S (HbS)polymer.The valine at the/~6 position of the deoxy-HbSfits into the hydrophobicpocket formedby leucine and phenylalanine at/~85 and/~88 of an adjacent/~ chain. Sinceeach/~ chain has an "acceptor"pocket and a "donor" valine, the HbS polymerhas a double-stranded, half-staggeredstructure. [Reproducedwith permission from S. Charache, Advancesin the understanding of sickle cell anemia. Hosp. Pract. 21(2), 173 (1986). J. E. Zupko, illustrator.]

670 the deoxy-HbS oxy-HbS ratio should reduce the likelihood of sickling and the severity of the disease. These approaches have been tried but so far have failed to ameliorate the disease. Among Bedouin Arabs and some populations of central and southern India, high HbF levels reduce the severity of sickle cell disease by inhibiting the formation HbS polymers. This observation has led to therapeutic approaches to induce higher levels of HbF in patients with sickle cell disease. The therapeutic agents are hydroxyurea and short-chain fatty acid derivatives. Hydroxyurea is an antineoplastic agent that inhibits ribonucleotide reductase (Chapter 27). It is thought that in bone marrow, hydroxyurea selectively kills many precursor cells but spares erythroblasts that produce HbE Hydroxyurea therapy also results in decreased circulating granulocytes, monocytes, and platelets. These changes, along with increased HbF, reduce vaso-occlusion due to a decreased propensity for sickling and adherence of red blood cells to endothelium. The long-term toxicity of hydroxyurea due to its myelosuppressive and teratogenic effects is not known. Induction of HbF by short-chain fatty acids such as butyrate was discovered in infants of diabetic mothers who had a delay in switching from HbF to HbA in association with elevated serum levels of amino-n-butyrate. This led to studies of HbF induction with several short-chain acids, including butyrate, all of which induce production of HbE The mechanism by which butyrate and other short-chain fatty acids affect gene expression involves transcriptional activation of a v-globin gene at the promoter site. One of the mechanisms of regulation of transcription, which occurs at the promoter site, is due to changes in histone acetylation and deacetylation (Chapter 26). Acetylation of specific histones by acetylases allows increased binding of transcription factors to target DNA that stimulates transcription. Deacetylation by histone deacetylases of histones prevents transcription. Butyrate is an inhibitor of deacetylase and it is thought that this action leads to induction of v-globin gene expression. Since hydroxyurea acts by a different mechanism in the elevation of HbF, butyrate's action can be additive or synergistic. Sustained induction of HbF by pulse butyrate therapy in patients with sickle cell anemia has resulted in up to a 20% increase in HbF along with marked clinical improvement. Hemoglobin E results from the substitution of glutamic acid with lysine at the 26th residue of the/~ chain; it has a high prevalence in Southeast Asia. Neither heterozygous nor homozygous states are associated with significant clinical abnormalities. The homozygous state exhibits a mild anemia, and both states show red blood cell indices resembling those of heterozygous thalassemic states, namely, hypochromic microcytosis. The latter has been attributed

CHAPTER 28

Hemoglobin

to activation of a cryptic splice site in exon 1 of/3-globin mRNA caused by/~E mutation. This leads to abnormal splicing of/~-globin mRNA, producing a less stable and ineffective mRNA. Coinheritance of the HbE gene with different forms of thalassemia genes is also known. 9'- and 6-Globin M u t a n t s The V- and 3-chain mutants are difficult to study because of the small fraction of HbF and HbA2 present in adult erythrocytes. Overt clinical symptoms associated with ?,- and g-chain variants are rare. By routine screening, 14 6-globin variants have been discovered but are of no clinical consequence. Thirty-five mutant V sequences (involving the G• or the A• chains) have been identified. They are all benign except for HbF Poole [G• Tyr ~ Gly], an unstable hemoglobin that causes hemolytic disease in the newborn.

Screening and Prenatal Diagnosis Screening programs are aimed at detection of the most common and most harmful hemoglobin genes and the identification of heterozygotes. Screening can provide a basis for genetic counseling and decrease the number of homozygous individuals conceived. Electrophoresis is used to differentiate among sickle cell trait, sickle cell anemia, and HbSC disease and to detect thalassemia, hereditary persistence of HbF, and HbC trait and HbC disease (see Appendix VII). Prenatal diagnosis of genetic disease often complements programs for detection of heterozygotes. If both parents are known to be carriers, a homozygous fetus can be detected early in pregnancy. The first prenatal tests measured the ability of fetal reticulocytes to direct the synthesis in vitro of normal or abnormal /3 chains. Fetal blood was obtained either by percutaneous aspiration from the placenta or transvaginally by direct aspiration from one of the fetal vessels on the surface of the placenta. The cells were incubated with radioactive amino acids, and the labeled globin chains were separated by chromatography and quantitated. This method detects normal and sickle cell anemia genotypes, absence of or- or /~-globin synthesis (homozygous c~~ or/~~ and some types of/~+-thalassemia. DNA has been isolated directly from cultured fetal cells (obtained by amniocentesis and from trophoblast biopsy) and subjected to restriction endonuclease digestion. (Culturing of fetal cells is necessary, since their number in the amniotic fluid sample is small.) The restriction fragments are separated according to size by electrophoresis and reacted with a labeled DNA probe that is complementary to the gene being sought. The hybridization pattern is compared with

671

SECTION28.4 Derivatives of Hemoglobin

F I G U R E 28-16 Diagram of the 5' region of the human fl-globin gene and its flanking region, showing the cleavage sites of the restriction endonuclease Mst II (vertical arrows). The fragments obtained by digestion of normal and sickle fl-globin genes are shown in the lower part of the diagram. Shaded regions of the gene are the exons, and IVS indicates the introns (intervening sequences). [Reproduced with permission from J. C. Chang and Y. W. Kan, A sensitive prenatal test for sickle cell anemia. N. Engl. J. Med. 307, 30 (1982).]

one from a normal individual (Chapter 23). Differences between the normal and some mutant globin genes at recognition sites for restriction endonucleases will cause differences in the cuts made and, hence, in the sizes of the fragments detected. In the ideal situation, a restriction endonuclease recognizes a site in the DNA that includes the mutated base. Restriction enzymes Mnl I, Dde I, and Mst II recognize part of the coding sequence for amino acids 5, 6, and 7 of the normal fi-globin gene. The mutation in the fi-globin gene in HbS abolishes these recognition sites, converting two small pieces of DNA found in the digest of normal /~-globin DNA into one larger fragment (Figure 28-16). Mst II is the most useful enzyme because it generates larger, more easily detected fragments. Loss of recognition sites due to deletions is seen in most o~-thalassemias and occasionally in fi-thalassemias, since most fi-thalassemias are not caused by deletion. In gene deletion, multiple sites are lost. However, point mutations are more common and therefore of greater importance than deletions. The endonuclease that is most useful in the detection of each disorder must be carefully selected. Because fi-thalassemias are caused by many different mutations with similar phenotypic expressions, it is not possible to find one or even a few restriction endonucleases that can detect all fi-thalassemia genes. What has been accomplished so far is the detection of affected individuals among members of the same kindred or racial group in which a single type of fi-thalassemia occurs. Alternatively, sites adjacent to an abnormal gene may differ from those on normal chromosomes. Such linked polymorphisms have been found for the sickle cell gene and for several fi-thalassemias. Because different individuals having a particular hemoglobin abnormality will not

always have the same linked polymorphism, this method suffers from lack of sensitivity. Under proper conditions, short, synthetic oligonucleotide probes, complementary to the normal DNA sequence in the region that contains the mutation, hybridize with completely homologous sequences but not with sequences that differ by as little as one nucleotide. This method is potentially more powerful than restriction endonuclease mapping because an oligonucleotide probe can be synthesized for any mutation once its sequence is known. Furthermore, multiple mutations can be detected simultaneously by hybridizing the DNA with a mixture of different probes, making this technique useful for screening. This technique has been used successfully to detect the sickle cell gene and several fl-thalassemias that could not be detected by restriction endonuclease mapping. All these methods depend on a satisfactory sample being collected from the fetus. Studies of globin chain synthesis require an adequate volume of fetal blood. Collection methods result in 4-6% fetal death under optimal conditions. Furthermore, fi-globin synthesis cannot be detected until about the middle of the second trimester. Use of amniotic fluid cells obtained by amniocentesis is less dangerous. However, an adequate volume of fluid cannot be obtained until the middle of the second trimester of pregnancy (14-20 weeks); if culturing of the cells is necessary, there is an additional delay of 1-2 weeks. Chorionic villus biopsy can be obtained at about 8-12 weeks of gestation. This allows more time for deliberation regarding termination or continuation of the pregnancy, thereby increasing the margin of safety for the mother, should termination be desired. This technique is no more dangerous than either of the other methods. Some other techniques for laboratory evaluation of hemoglobin disorders are presented in Appendix VII.

28.4 Derivatives of Hemoglobin Carbon Monoxide-Hemoglobin Carbon monoxide, an odorless gas, has an affinity for hemoglobin that is 210 times that of oxygen. Thus, in the equilibrium reaction HbO2 + CO ~ HbCO-4-02

Keq -- 2.1 • 102

the equilibrium lies far to the right. Like oxygen, CO binds to the sixth position of the heme iron. When CO and 02 are present together in appreciable quantities, CO is bound preferentially and oxygen is excluded, effectively causing an anemic hypoxia. Even if some O2 is bound to the hemoglobin, it cannot be released owing to the tight

CHAPTER28 Hemoglobin

672 binding of CO and to its action as an allosteric activator, with the result that hemoglobin is trapped in the R-state. Carbon monoxide poisoning may result from breathing automobile exhaust, poorly oxygenated coal fires in stoves and furnaces, or incomplete combustion of a carbon-containing compound. Breathing air containing 1% CO for 7 minutes can be fatal; automobile exhaust contains about 7% CO. Unconsciousness, a cherry-red discoloration of the nail beds and mucous membranes (due to large amounts of R-state carbon monoxide-hemoglobin), and spectrophotometric analysis of the blood are useful for clinical diagnosis. Treatment of carbon monoxide poisoning consists of breathing a mixture of 95% O2 and 5% CO2, which will usually eliminate carbon monoxide from the body in 30-90 minutes, or of breathing hyperbaric oxygen. The enzymatic oxidation of heme produces CO and biliverdin in equimolar amounts (Chapter 29). The CO is transported via the blood to the lungs, where it is released. Although no cases are known in which endogenous CO proved toxic, this carbon monoxide may contribute significantly to air pollution, particularly in crowded and enclosed areas.

Carbaminohemoglobin Some of the C02 in the bloodstream is carried as carbamino compounds (Chapter 1). These compounds form spontaneously in a readily reversible reaction between CO2 and the free c~-amino groups in the N-terminal residues of the Hb chains.

H2N~

NH2 4-4002 ~

H2N

~-

NH2 Hemoglobin

0%

H

I

H

I //o

Ho/C--N~N--C~oH O~c__N//xL'A-'~ N C~ 0 HO/ I H HI- ~OH Carbaminohemoglobin Although Hb can carry as many moles of C02 as of 02, the HCO~- system is a more important way of transporting CO2. When the N-terminal amino groups are blocked by carbamylation with CNO-, no marked degree of acidosis develops. The carbamino groups decrease the affinity thus can affect oxygen affinity of Hb for 02. The Pr independently of any effect it may have on pH and may

influence the position of the oxygen dissociation curve under various conditions.

Methemoglobin In the presence of oxygen, dissolved hemoglobin is slowly oxidized to methemoglobin, a derivative of hemoglobin in which the iron is present in the ferric (Fe 3+) state. In metHb, the ferric ion is bound tightly to a hydroxyl group or to some other anion. The heme porphyrin (protoporphyrin IX) that contains an Fe 3+ ion is known as hemin. A small but constant amount of methemoglobin is produced in the body. It is reduced by specific enzymes (methemoglobin reductases) and NADH, which is generated in glycolysis. Reductases isolated from human red cells also use NADPH but to a lesser extent. Inability to reduce metHb produces methemoglobinemiaandtissue anoxia. Hereditary methemoglobinemia may arise from the following: 1. Deficiency of one or more of the metHb reducing enzymes (usually a recessive trait). MetHb values may range from 10% to 40% of the total Hb (normal = 0.5%). Treatment involves administration of an agent that will reduce the metHb (e.g., ascorbic acid, methylene blue). 2. Defects in the hemoglobin molecule that make it resistant to reduction by metHb reductases and ascorbate or methylene blue. These hemoglobins are collectively called the M hemoglobins (Table 28-5). HbM is inherited as a dominant trait, since homozygosity for an M fl-chain hemoglobin would be lethal. In four types that involve an His ---> Tyr substitution, the phenolic hydroxyl group forms a stable complex with Fe 3+, making it resistant to reduction to Fe z+. In HbM Milwaukee, a glutamic acid residue substitutes for a valine at position 67 near the distal histidine and forms a stable complex with Fe 3+. Although a brownish cyanosis is characteristic of blood containing HbM, in contrast to the purplish cyanosis caused by excess deoxyhemoglobin, diagnosis should be confirmed by the absorption spectrum of either the Hb or its CN- derivative. The spectrum of the cyanide derivative is preferred, since it differs more from that of HbA. Some of the M hemoglobins cannot form CN- derivatives, presumably because a Tyr or Glu blocks the sixth iron position. In these cases, the absorption spectrum of the methemoglobin itself must be examined. Because of the change in the environment of the heme due to the change in the globin structure, the spectra of M

SECTION 28.4

Derivatives of Hemoglobin

673

hemoglobins differ among themselves and from that of normal metHb. Theoretically, hereditary methemoglobinemia could occur if the hemoglobin was altered in that region where methemoglobin reductase binds in performing its function. In such a mutant Hb, the reductase would not bind. Acquired acute methemoglobinemia is a relatively common condition caused by a variety of drugs such as phenacetin, aniline, nitrophenol, aminophenol, sulfanilamide, and inorganic and organic nitrites and nitrates. The condition is less commonly produced by chlorates, ferricyanide, pyrogallol, sulfonal, and hydrogen peroxide. These compounds appear to catalyze the oxidation of Hb by oxygen. Symptoms include brownish cyanosis, headache, vertigo, and somnolence. Diagnosis is based on occurrence of brownish cyanosis and presence of excessive amounts of metHb (measured spectrophotometrically). Treatment usually consists of removal of the offending substance and administration of ascorbic acid (in mild cases) or methylene blue (in severe cases). These reducing agents function according to the reactions below (MB -- methylene blue, oxidized; MBH2 -- methylene blue, reduced). MBH2 ~ MB + Hz(gas) 2Hz(gas) 4- Hb(Fe 3+)4 --+ Hb(Fe 2+) + 4H + (The release of H2 by MBH2 is spontaneous because methylene blue is autoxidizable.) " O---C

I

HO--C

II

HO--C

I

H--C

I

I

T

HO--CH

I CH2OH Ascorbic acid (vitamin C)

O--C

I

j/ Hb(Fe+3)4

4H+

O--C

I

O--C Hb(Fe+2)4

I

H--C

I

i

O

I

HO--CH

I

CH2OH Dehydroascorbic acid

Sulfhemoglobin Many drugs that cause methemoglobinemia also stimulate production of sulfhemoglobin, a greenish derivative. Methemoglobin and sulfhemoglobin may appear together in poisoning by phenacetin, acetanilid, or sulfanilamide. Dapsone (used to treat leprosy) and exposure to sulfurcontaining compounds either occupationally or from air pollution can also produce sulfhemoglobinemia. The sulfur appears to attach covalently to the porphyrin ring rather than to the iron atom. It is unclear whether the sulfur is derived from hydrogen sulfide generated by anaerobic

metabolism of intestinal bacteria, from glutathione, or from some other source. Because of its color, patients with circulating sulfhemoglobin appear cyanotic like those with methemoglobinemia. However, unlike methemoglobinemia, sulfhemoglobinemia cyanosis becomes clinically apparent at extremely low concentrations and is usually not accompanied by cardiopulmonary pathology. This fact, together with the overlap in the list of causative agents, may lead to underdiagnosis of sulfhemoglobinemia. Both sulfHb and metHb show an absorption peak at about 620 nm that is not present in deoxyHbA (the most common cause of cyanosis). They can be discriminated by changes in this peak on addition of cyanide, carbon monoxide, or dithionite (a reducing agent). Isoelectric focusing of samples treated with the same chemicals is also useful for differential diagnosis. Oxygen does not bind to a subunit having a sulfurated porphyrin ring. The presence of one or two such subunits, together with normal subunits in a hemoglobin tetramer, alters the cooperativity, favors the deoxy structure, and thereby decreases the oxygen affinity of the normal subunits. Thus, at low levels of sulfuration, the percentage of affected hemoglobin tetramers is greater than the percentage of sulfurated monomers. (This is also true of the methemoglobins). In contrast, binding of carbon monoxide to one or two subunits increases the oxygen affinity of the tetramer, since CO stabilizes the oxyhemoglobin conformation. In a person who has HbA as the predominant hemoglobin, the presence of sulfhemoglobin is probably innocuous. Tissue oxygenation is relatively normal because the decrease in the number of binding sites for oxygen is offset by the lower binding affinity. However, since sulfuration stabilizes the deoxy form, sulfhemoglobin, in individuals who have sickle cell anemia, should increase the likelihood of sickling and thereby exacerbate the disease. No way is known to reverse formation of sulfhemoglobin. If the causative agent is removed, sulfhemoglobin will disappear from the circulation at the same rate as that of the erythrocytes that contain it.

Cyanmethemoglobin Cyanide poisoning does not cause production of cyanohemoglobinemia or cyanosis. It does produce cytotoxic anoxia by poisoning cytochrome oxidase and other respiratory enzymes, thereby preventing utilization of O2 by tissues. Cyanide poisoning is detected by the characteristic odor of HCN gas (odor of bitter almonds) on the breath and by laboratory tests (absorption spectra, tests for CN-).

674 Treatment of cyanide poisoning consists of diverting the cyanide into the production of cyanmetHb. First, some of the normal hemoglobin is converted to methemoglobin by intravenous infusion of a solution of NaNO2 or inhalation of amyl nitrite. Once metHb is formed, CN- can replace OH- at position 6 of the iron, since it has a higher affinity of Fe 3+ than does OH-. CyanmetHb is no more toxic than metHb and cells containing it can be eliminated by normal body processes. The cyanide bound to metHb is always in equilibrium with free CN-, and this uncomplexed cyanide is converted to thiocyanate (SCN-; nontoxic) by administration of thiosulfate (Chapter 6).

Glycated Hemoglobins Several minor varieties of hemoglobin are produced by nonenzymatic posttranslational modification (e.g., glycation). Because these minor hemoglobins are present in such small amounts, none is pathological. Both or- and s-amino groups of hemoglobin form amino-l-deoxyfructose adducts upon reaction with glucose (Chapter 2). Other hexoses can give rise to similar adducts (e.g., galactose in galactosemia; Chapter 15). The major sites of in vivo glycation in order of prevalence are fl-Vall, fl-Lys66, c~-Lys61, fl-Lysl 7, and ot-Vall. The adduct formed with the amino terminus of the fl chains is known as HbAic, which makes up about 4-6% of the total hemoglobin in normal red blood cells. Its concentration is increased in uncontrolled diabetics who have hyperglycemia. Glycated hemoglobins are detected as a fast-moving hemoglobin in electrophoresis at alkaline pH. Because HbAlc accumulates within the erythrocyte throughout the cell's normal life span, it is used as an indicator of the success of long-term blood glucose control in diabetics (Chapter 22). Also known are adducts of hemoglobin with glucose-6-phosphate and fructose1,6-diphosphate, which probably compete with 2,3-DPG for binding to the fl chains of deoxyhemoglobin, and a complex between hemoglobin and glutathione, particularly in older erythrocytes.

Supplemental Readings and References M. O. Arcasoy and E G. Gallagher: Molecular diagnosis of hemoglobinopathies and other red blood cell disorders. Seminars in Hematology 36, 328 (1999).

CHAPTER28 Hemoglobin G. F. Atweh, M. Sutton, L. Nassif, et al.: Sustained induction of fetal hemoglobin by pulse butyrate therapy in sickle cell disease. Blood 93, 1790 (1999). H. E Bunn: Pathogenesis and treatment of sickle cell disease. New England Journal of Medicine 337, 762 (1997). H. E Bunn: Induction of fetal hemoglobin in sickle cell disease. Blood 93, 1787 (1999). A. Cao, R. Galanello, and M. C. Rosatelli: Genotype-phenotype correlations in fl-thalassemias. Blood Reviews 8, 1 (1994). M. Cazzola, E Mercuriali, and C. Brugnara: Use of recombinant human erythropoietin outside the setting of uremia. Blood 89, 4248 (1997). D. H. K. Chui, R. Hardison, and C. Riemer: An electronic database of human hemoglobin variants on the World Wide Web. Blood 91, 2643 (1998). S. C. Davies and L. Oni: Management of patients with sickle cell disease. British Medical Journal 315, 656 (1997). B. L. Ebert and H. E Bunn: Regulation of the erythropoietin gene. Blood 94, 1864 (1999). A. Ferster, C. Vermylen, G. Cornu, et al.: Hydroxyurea for treatment of severe sickle cell anemia: a pediatric clinical trial. Blood 88, 1960 (1996). J. R. Girman, Y-L. Chang, S. B. Hayward, et al.: Causes of unintentional deaths from carbon monoxide poisonings in California. Western Journal of Medicine 168, 158 (1998). E Grosveld, E. De Boer, N. Dillon, et al.: Dynamics ofglobin gene expression and gene therapy vectors. Annals of the New York Academy of Sciences 550, 18 (1998). C. C. W. Hsia: Respiratory function of hemoglobin. New England Journal of Medicine 338, 239 (1998). E. Liakopoulou, C. A. Blau, L. Qiliang, et al.: Stimulation of fetal hemoglobin production by short chain fatty acids. Blood 86, 3227 (1995). J. A. Little, N. J. Dempsey, M. Tuchman, et al.: Metabolic persistence of fetal hemoglobin. Blood 85, 1712 (1995). J. M. Manning, A. Dumoulin, X. Lee, et al.: Normal and abnormal protein subunit interactions in hemoglobins. Journal of Biological Chemistry 273, 19359 (1998). P. G. McCaffrey, D. A. Newsome, E. Fibach, et al.: Induction of v-globin by histone deacetylase inhibitors. Blood 90, 2075 (1997). R. T. Means Jr. and S. B. Krantz: Progress in understanding the pathogenesis of the anemia of chronic disease. Blood 80, 1639 (1992). M. Noor and E. Beutler: Acquired sulfhemoglobinemia. Western Journal of Medicine 169, 386 (1998). N. E Olivieri: The fl-Thalassemias. New England Journal of Medicine 341, 99 (1999). J. P. Scott, C. A. Hillery, E. R. Brown, et al.: Hydroxyurea therapy in children severely affected with sickle cell disease. Journal of Pediatrics 128, 820 (1996). G. Serjeant: Sickle-cell disease. Lancet 350, 725 (1997). M. H. Steinberg: Management of sickle cell disease. New England Journal of Medicine 340, 1021 (1999). I. A. Tabbara: Erythr~176 oflnternal Medicine 153, 298 (1993). P. M. Tibbles and J. S. Edelsberg: Hyperbaric-oxygen therapy. New England Journal of Medicine 334, 1642 (1996). D. J. Weatherall and J. B. Clegg: Genetic disorders of hemoglobin. Seminars in Hematology 36, 24 (1999). H. E. Witkowska, B. H. Lubin, Y. Beuzard, et al.: Sickle cell disease in a patient with sickle cell trait and compound heterozygosity for hemoglobin S and hemoglobin Quebec-Chori. New England Journal of Medicine 325, 1150(1991).

CHAPTER

29

Metabolism of Iron and Heme

Heme, an iron-porphyrin complex, is the prosthetic group of many important proteins. The central role of hemoglobin and myoglobin in oxygen transport and storage was discussed in Chapter 28. Heme proteins or enzymes are involved in redox reactions (e.g., cytochromes) and participate in many oxidation reactions needed for synthesis of metabolically important compounds as well as for degradation and detoxification of waste products and environmental toxins. Ionic forms of iron (referred to hereafter as iron) also participate in a variety of enzymatic reactions as nonheme irons, which are present as iron-sulfur clusters (e.g., mitochondrial electron transport). There are also both storage and transportable forms of iron that are bound to proteins. Under normal physiological conditions only trace amounts of free iron exist. In the body, if iron exceeds the sequestration capacity of the iron-binding proteins present in different physiological compartments, the free iron can cause tissue damage. Cellular injury is caused by reactive oxygen species that are produced from H202 in a reaction catalyzed by iron. Thus, iron homeostasis in the body is in a delicate balance. Either the deficiency or the excess results in abnormalities and presents as a common cause of human diseases.

29.1 Iron Metabolism Total-body iron of a 70-kg adult is about 4.2-4.4 g. The distribution of iron in various body compartments is given in Table 29-1. The key players of iron metabolism include iron-responsive elements of appropriate mRNAs, iron regulatory proteins divalent metal transporter 1, major histocompatibility complex (MHC) class I-like protein designated as HFE protein,/32-microglobulin, transferrin, transferrin receptor, and ferritin.

Absorption of Iron from the Diet The dietary requirement for iron depends on the amount and composition of the food, the amount of iron lost from the body, and variations in physiological state such as growth, onset of menses, and pregnancy. The average North American diet contains about 6 mg of iron per 1000 calories and supplies about 10-15 mg/d. Of that ingested, 8-10% (1-1.5 mg/d) is absorbed. Thus, dietary factors that affect absorption are more important than the iron content of the diet and may be more important for correction of iron deficiency than addition of iron to the diet.

675

CHAPTER29 Metabolism of Iron and Heme

676

TABLE 29-1 Distribution of Iron in a 70-kg Adult I Circulating erythrocytes Bone marrow (erythroid) Muscle myoglobin Heme and non-heme enzymes Liver parenchyma 3 Reticuloendothelial macrophages 4 Plasma transferrin 5

1800 300 300 180 1000 600 3

mg 2 mg mg mg mg mg mg

1 These are approximate values. Premenopausal women have lower iron stores due to periodic blood loss through menstruation. Iron balance in the body is maintained by intestinal absorption of 1-2 mg/day and by loss of 1-2 mg/day. 21 mg = 17.9/xmol 3 Primarily storage forms of iron. 4 Senescent red blood cells are catabolized by the macrophages, the salvaged iron is temporarily stored, and made available via transferrin for erythron and for hemoglobin synthesis. 5 Transportable form of iron. Iron in food exists mainly in the ferric (Fe 3+) state, complexed to proteins, amino acids, organic acids, or heme. It is absorbed in the ferrous state, reduction being accomplished in the gastrointestinal tract by ascorbate, succinate, and amino acids. Gastric acid potentiates iron absorption by aiding in formation of soluble and absorbable ferrous chelates. In achlorhydria or after partial gastrectomy, absorption is subnormal but is increased by administration of hydrochloric acid. Carbonates, tannates, phosphates, phytates, and oxalates may decrease iron absorption by formation of insoluble complexes, but their effect can be prevented by adequate dietary calcium, which complexes with them and makes them unavailable for reaction with iron. Absorption of heme iron is not affected by these agents. Heme is absorbed intact from food and more effectively than inorganic iron. Antacids, such as aluminum hydroxide and magnesium hydroxide, also decrease iron absorption. In general, foods of animal origin provide more assimilable iron than foods of vegetable origin, since on a weight basis, vegetables contain less iron and more substances (e.g., phytates) that inhibit iron absorption. Foods that contain more than 5 mg of iron per 100 g include organ meats (liver, heart), wheat germ, brewer's yeast, oysters, and certain dried beans. Foods that contain 1-5 mg of iron per 100 g include muscle meats, fish, fowl, some fruits (prunes, raisins), most green vegetables, and most cereals. Foods that contain less than 1 mg of iron per 100 g include milk and milk products and most nongreen vegetables. Ferrous iron is absorbed principally from the mature enterocytes lining the absorptive villi of the duodenum. The amount of iron absorption by these enterocytes is

determined by the prior programming of the duodenal crypt cells based on iron requirements of the body as they undergo maturation. The regulation of intestinal iron absorption is critical because iron excretion from the body is a limiting physiological process (discussed later). The small intestine is also an excretory organ for iron, since that stored as ferritin in the epithelial cells is lost when they are shed and replaced every 3-5 days. Heme iron is transported intact into the mucosal cells and the iron removed for further processing. The mechanism of entry of ferrous iron from the intestinal lumen into the enterocytes and its eventual transport into the portal blood is beginning to be understood. The first step is the programming of the duodenal undifferentiated deep-crypt cells with regard to sensing the iron requirements of the body. The programming of the crypt cells for capacity to absorb iron is thought to occur as follows. A protein (HFE) that spans the cell membrane like an HLA molecule associates with flz-microglobulin like an HLA protein. The HFE protein is coded for by a gene (HFE) located on the short arm of chromosome 6 near the MHC gene loci. Mutations in the HFE gene are associated with an inherited disorder of excessive dietary iron absorption that is known as hereditary hemoehromatosis (discussed later). HFE protein spans the cell membrane of the crypt enterocytes with its N-terminal domain projecting outside. Near the cell membrane a segment of HFE protein, like an HLA protein, associates with /32-microglobulin (Figure 29-1) and stabilizes the HFE protein. Plasma transferrin transports iron in the ferric state and is an indicator of iron stores in the body. Each molecule of transferrin binds with two ferric ions (diferric transferrin) and undergoes receptor-mediated endocytosis when bound to transferrin receptors (discussed later) with the aid of HFE protein/32-microglobulin complex. In the cytosol, iron is released from the endosomes. The level of cytoplasmic iron regulates the translation of mRNA of a protein known as divalent metal transporter-I (DMT1), which participates in iron entry into the enterocytes located on the villus tip (Figure 29-2). Regulation of DMT1 synthesis is coupled to cytoplasmic iron levels and involves the presence of a stem-loop hairpin structure in the 3'-untranslated region that resembles an iron regulatory element (IRE) and its interaction with an iron regulatory protein (IRP1). In the iron-deficient state, IRP binds to IRE and stabilizes the mRNA of DMT1. This stabilization of mRNA leads to increased production of DMT1 and its eventual localization on the cell surface. The transport of ferrous iron across the apical membrane of the villus enterocyte that is mediated by DMT1 occurs through a proton-coupled process. DMT1 also transports a number

SECTION29.1 Iron Metabolism

677

F I G U R E 29-1 A diagrammatic representation of the transmembrane HFE protein. The extracellular portion of the HFE protein consists of three o~ domains. The/32-microglobulin is noncovalently associated with the oe3 domain of HFE protein, stabilizing its structure. The extracellular missense mutations H63D and C282Y are identified in patients with hereditary hemochromatosis. HFE protein is involved in sensing circulating iron concentration and participates in the regulation of gene expression of products involved in iron absorption, transport, or storage. [Reproduced with permission from: A novel MHC class l-like is mutated in patients with hereditary hemochromatosis. J. N.Feder, A. Gnirke, W. Thomas et al. Nature Genetics 13, 399 (1996).]

of divalent metal ions, including Mn 2+, Cu 2+, Zn 2+, Cd 2+, and Pb 2+. At normal levels of iron intake, absorption requires uptake from the intestinal lumen by the mucosa and transfer from the mucosa to the portal blood. Both events are inversely affected by the state of body iron stores. In iron deficiency states, nonferrous metals such as cobalt and manganese, which have an ionic radius similar to that of iron and form octahedral complexes with six-coordinate covalent bonds, also are absorbed at an increased rate. Oral administration of a large dose of iron reduces (or temporarily inhibits) the absorption of a second dose of iron by the absorptive enterocytes even in the presence of systemic iron deficiency. The mechanism of mucosal block, which resists acquiring additional iron by the enterocytes with high amounts of intracellular iron, is not yet understood. It probably involves set points established in the enterocytes for iron recently consumed in the diet (dietary regulator), o Iron absorption also is affected by erythropoiesis. When erythropoiesis is accelerated by bleeding, hemolysis, or hypoxia, iron absorption is increased. Conversely,

diminished erythropoiesis due to starvation, blood transfusion, or return to sea level from a high altitude decreases iron absorption. How the size of body iron stores and the rate of erythropoiesis transmit information to the duodenum is not known. Feedback control seems to be weak or absent, since in iron-deficient subjects enhanced iron absorption continues long after hemoglobin is restored to normal levels. Furthermore, in chronic hemolytic anemia, iron absorption is increased, persists for prolonged periods, and leads to iron overload. The need for dietary iron is ultimately determined by the rate of iron loss from the body and the amount required for maintenance and growth. Iron is tightly conserved once it is absorbed. Its excretion is minimal and unregulated, and facilitated by normal exfoliation from the surfaces of the body (dermal, intestinal, pulmonary, urinary), loss of blood by gastrointestinal bleeding, and loss in bile and sweat. Insignificant amounts are lost in urine, since iron in plasma is complexed with proteins that are too large to pass through the kidney glomerular membrane. Iron in feces is primarily unabsorbed dietary iron. Obligatory iron

6~

CHAPTER29 Metabolism of Iron and Heme

F I G U R E 29-2 Intestinal absorption of dietary iron. Ferrous iron is absorbed by the duodenal villus tip enterocytes mediated by divalent metal transporter-1 (DMTI). Iron transport mediated by DMTI of the apical surface and the basolateral transporter at the basolateral surface are coupled to ferric reductase and ferroxidase that change the iron oxidation state, respectively. The degree of iron entry is determined by the level of DMT 1 and its level of expression is programmed in the crypt cells. The programming of the crypt cells is coupled to the body iron stores via transferrin-mediated and HFE protein-modulated iron transport. [Modified and reprinted with permission from B. R. Bacon, L. W. Powell, R C. Adams, et al. Molecular medicine and hemochromatosis: at the cross roads. Gastroenterology 116, 193 (1999).]

loss for a 70-kg man is 0.5-1 mg/d, an amount equal to that normally absorbed from the diet. During growth, menstruation, and pregnancy, the requirement reaches about 2-2.5 mg/d. Recommended daily iron intake for various groups is shown in Appendix IV.

The principal loss of iron in nonpregnant women during the reproducing years is through menstruation. In one study, the mean menstrual blood loss was 43.4 4- 2.3 mL. Since each milliliter of blood from a normal woman contains about 0.5 mg of iron, the amount lost every 27 days is

SECTION29.1 Iron Metabolism

about 20-23 mg. Increased menstrual flow (menorrhagia) significantly augments iron loss and leads to iron deficiency anemia (see below). In pregnancy use of supplemental iron is recommended. A newborn has about a 3- to 6-month supply of iron in its liver and may require ironrich foods from the sixth month onward, since milk is poor in iron.

Plasma Iron Transport Over 95% of plasma iron is in the Fe 3+ state bound to the glycoprotein transferrin, a monomeric/~ 1-globulin (M.W. 80,000). Electrophoretic studies have revealed the existence of 21 genetic variants. In some, single-aminoacid substitutions account for variation in electrophoretic mobility. Transferrin is synthesized primarily in the liver and appears at the end of the first month of fetal development. Its half-life in humans is about 8 days. Desialylation may be a requirement for its removal from plasma by the liver, as it is for other plasma proteins (Chapter 10). In fact, asialotransferrin is more rapidly cleared from plasma than transferrin. It is not required for intestinal absorption of iron. Each molecule of transferrin can bind two Fe 3+ ions. The binding is extremely strong under physiological conditions, and the binding constants of the two sites are not significantly different. For each Fe 3+ bound, one HCOy ion is also bound and three H + ions are released from the protein. Thus, diferric transferrin gains two net negative charges. 2Fe 3+ + apotransferrin + 2HCO~-

679

and HFE protein. This process of iron release from the complex is inhibited by HFE protein. Thus, dysfunctional HFE protein can cause excessive release of iron from the transferrin-transferrin receptor-HFE protein complex. In the acidified endosomes, DMT1 facilitates iron transport into the cytosol. Both apotransferrin (and a fraction of ironbound transferrin) and transferrin receptor are returned to cell surfaces for reuse. In this type of receptor-mediated endocytosis of transferrin-transferrin receptor complex, the endosomes do not come into contact with lysosomes. The process is therefore unlike that of low-density lipoprotein receptor-mediated internalization (Chapter 20). In the erythroid cells, most of the iron released from the endosomes is transported into mitochondria for heme synthesis (discussed later); in nonerythroid cells, the iron is stored predominantly as ferritin and to some extent as hemosiderin.

Storage of Iron There are two storage forms of iron, ferritin and hemosiderin. Ferritin is the predominant storage form and contains diffusable, soluble, and mobile fractions of iron. Hemosiderin is aggregated deposits resulting from the breakdown of ferritin in secondary lysosomes and its level increases progressively with increasing levels of iron overload. Apoferritin is a protein shell consisting of 24 subunits of two types; a light (L) subunit (M.W. 19,000) and a heavy (H) subunit (M.W. 21,000). The H subunit has ferroxidase activity and the L subunit facilitates nucleation and mineralization of the core made up of hydrated ferric oxide phosphate complex.

[Fe2-transferrin-(HCO3)2] 2- + 6H + The metal binding sites are located in N- and C-terminal domains. The protons released upon binding of each Fe 3+ ion are probably derived from ionization of two tyrosyl residues and of a water molecule bound to Fe 3+ ion. The bulk of transferrin iron is delivered to immature erythroid cells for utilization in heme synthesis. Iron in excess of this requirement is stored as ferritin and hemosiderin. Unloading of iron to immature erythroid cells is by receptor-mediated endocytosis. The process begins in the clathrin-coated pits with the binding of diferric transferrin to specific plasma membrane transferrin receptors that are associated with the HFE protein complex. The next step is the internalization of the transferrintransferrin receptor-HFE protein complex with formation of endosomes. The iron transporter DMT1 present in the cell membrane is also internalized into the endosomes. In the endosomes, a proton pump acidifies the complex to pH 5.4, and by altering conformation of proteins, iron is released from transferrin bound to transferrin receptor

Coordinate Regulation of Iron Uptake and Storage in Non-Erythroid Cells Iron uptake is regulated by transferrin receptor and storage of iron as ferritin which occurs post-transcriptionally for these two proteins. The regulation maintains an optimal intracellular-transit-chelatable iron pool for normal functioning in the body. The regulatory process consists of an interaction between IREs and IRPs 1 and 2. One copy of each IRE has been identified in the 5'-untranslated region of H and L ferritin mRNAs and five copies in the 3'-untranslated region (UTR) of transferrin receptor mRNA. IRE sequences are highly conserved and have a stem-loop structure with a CAGUGN sequence at the tip of the loop. IRPs are RNA-binding proteins that bind to IREs and regulate the translation of the respective mRNAs. During low levels of intracellular chelatable iron, iron storage declines due to inhibition of ferritin synthesis; cellular entry of iron increases due to enhanced transferrin receptor synthesis. An opposing set of events occurs

680

CHAPTER29 Metabolism of Iron and Heme

At low cytosolic mobile iron pool 9 IRP levels increase and bind to mRNA-IREs of ferritin and transferrin receptors.

5,~c~f~f~fXj ~ RE Coding Region UTR > Inactive ferritin mRNA

~

An 3'

IRP IRE

5

'

~ UTR

I

Coding Region

~

"',~""'--.~

An 3'

~- '

Active Transferrin receptor mRNA At high cytosolic mobile iron pool : O ~-'" IRE IRP levels decrease causing opposite II effects on the mRNAs. , ~ ~ ~ . J ~ 5 UTR > i Coding Region g ~ F ~ F x ~ ' ~ A n 3' P~ UTR Active ferritin mRNA IRE

5

'

~

Coding Region

UTR

An 3'

Inactive transferrin receptor mRNA F I G U R E 29-3 Coordinate translational regulation of ferritin mRNA and transferrin receptor mRNA in nonerythroid cells. Iron regulatory proteins (IRP) are RNA-binding proteins that bind to iron regulatory elements (IREs). IREs are hairpin structures with loops consisting of CAGUGN sequences and are located at the 5'-untranslated region (UTR) and 3'-UTR for ferritin mRNA and transferrin mRNA, respectively.

during intracellular chelatable iron excess or iron-replete states. A coordinated control occurs when IRP binds to IRE at the 5'-UTR of ferritin mRNAs inhibiting ferritin synthesis; simultaneously, the binding of IRP to IRE at the 3'-UTR of transferrin receptor mRNA stimulates transferrin receptor synthesis (Figure 29-3). Intracellular iron regulates the level of IRPs. During the expansion of the iron pool, IRPs are inactivated, leading to efficient translation of ferritin mRNA and rapid degradation of transferrin receptor mRNA. In iron-replete cells, IRP1 acquires iron by the formation of iron-sulfur clusters (4Fe-4S) that bind to IREs with low affinity. During iron deficiency states, IRP 1 lacks a 4Fe-4S cluster and binds to IREs with high affinity. IRP1, when it possesses an iron-sulfur cluster, has aconitase activity, normally TCA cycle enzyme (Chapter 14). Mutations that change IREs can lead to constitutive ferritin biosynthesis. An autosomal dominant disorder of IRE leads to hyperferritinemia without any iron overload. Patients with this inherited disorder also exhibit early onset of cataract that may also be cotransmitted as an autosomal

dominant trait. Other factors also regulate ferritin synthesis. For example, nitric oxide enhances binding of IRP to IRE and inhibits ferritin synthesis. One of the causes of anemia of chronic inflammatory diseases may be due to increased ferritin synthesis in the reticuloendothelial macrophages by inflammatory cytokines interleukins 1 and 6 (IL-I and IL-6), which act by preventing efficient release of iron. Measurement of serum ferritin levels has diagnostic utility. In iron deficiency anemia (discussed later), serum ferritin levels are low; in iron storage disease, the levels are high. However, serum ferritin levels can also be elevated under many other circumstances, including liver diseases and chronic inflammatory diseases. Alterations of Plasma Transferrin Concentration Plasma transferrin levels are commonly measured in the evaluation of disorders of iron metabolism (see below). It is customary to measure transferrin concentration

SECTION29.1 Iron Metabolism

indirectly from the maximum (or total) iron binding capacity (TIBC) of plasma (reference range for adults, 250-400 #g/dL). It can also be measured directly by immunological methods (reference range for adults, 220-400 mg/dL). Hypertransferrinemia (or increased TIBC) can occur with diminished body iron stores as in iron deficiency anemia or during pregnancy (because of enhanced mobilization of storage iron to supply maternal and fetal demands). Hypertransferrinemia of iron deficiency is corrected by oral iron supplementation, whereas that due to pregnancy is not. Exogenous administration of estrogens (e.g., oral contraceptives) also causes hypertransferrinemia. Hypotransferrinemiacan result from protein malnutrition and accompanies hypoalbuminemia. Since transferrin has a much shorter half-life (8 days) than albumin (19 days), measurement of the transferrin level may be a more sensitive indicator of protein malnutrition than albumin measurement (see also chapter 17). Hypotransferrinemia also results from excessive renal loss of plasma proteins (e.g., in nephrotic syndrome).

Disorders of Iron Metabolism

Iron Deficiency Anemia Iron deficiency anemia is the most prevalent nutritional disorder. Its cause may comprise many overlapping factors: dietary iron deficiency; absence of substances that favor iron absorption (ascorbate, amino acids, succinate); presence of compounds that limit iron absorption (phytates, oxalates, excess phosphates, tannates); lack of iron absorption due to gastrointestinal disorders (malabsorption syndrome, gastrectomy); loss of iron due to menstruation, pregnancy, parturition, lactation, chronic bleeding from the gastrointestinal tract peptic ulceration, hemorrhoids, cancer, colonic ulceration, or hookworm infestation or the genitourinary tract (uterine fibroids); enhanced demand for growth or new blood formation; deficiency of iron transport from mother to fetus; abnormalities in iron storage; deficiencies in release of iron from the reticuloendothelial system (infection, cancer); inhibition of incorporation of iron into hemoglobin (lead toxicity); and rare genetic conditions (transferrin deficiency, impaired cellular uptake of iron by erythroid precursors). In the initial phase of depletion of the iron content of the body, the iron stores maintain normal levels of hemoglobin and of other iron proteins. With exhaustion of storage iron, hypochromic and microcytic anemia becomes manifest. The clinical characteristics of iron deficiency anemia are nonspecific and include pallor, rapid exhaustion, muscular weakness, anorexia, lassitude, difficulty in concentrating,

681 headache, palpitations, dyspnea on exertion, angina on effort, peculiar craving for unnatural foods (pica), ankle edema, and abnormalities involving all proliferating tissues, especially mucous membranes and the nails. The onset is insidious and may progress slowly over many months or years. Physiological adjustments take place during the gradual progression of the disorder, so that even a severe hemoglobin deficiency may produce few symptoms. Iron deficiency may affect the proper development of the central nervous system. Early childhood iron deficiency anemia may lead to cognitive abnormalities. Individuals who have congenital atransferrinemia lack apotransferrin and suffer from severe hypochromic anemia in the presence of excess iron stores in many body sites, susceptibility to infection (transferrin inhibits bacterial, viral, and fungal growth, probably by binding the iron required for growth of these organisms), and retardation of growth. This condition does not respond to administration of iron. Intravenous administration of transferrin normalizes the iron kinetics. A rare congenital defect in uptake of iron by red cell precursors has been reported that leads to severe hypochromic anemia with normal plasma iron and transferrin levels. Microcytic anemia occurs frequently in thalassemia syndromes (Chapter 28), but these patients do not require iron supplementation unless they have concurrent iron deficiency as assessed by measurement of serum iron levels and TIBC. Serum iron concentration exhibits a morning peak and an evening nadir; this pattern is reversed in nightshift workers. The circadian variation is primarily due to differences in rate of release of iron by the reticuloendothelial system. Transferrin levels do not show circadian fluctuation. Iron deficiency anemia can also be assessed from the plasma ferritin concentration (which when decreased reflects depleted iron stores), red cell protoporphyrin concentration (increased because of lack of conversion to heme), and the number of sideroblasts in the bone marrow (which parallels iron stores). Sideroblasts are erythrocyte precursors (normoblasts) containing free ferritin-iron granules in the cytoplasm that stain blue with the Prussian blue reagent. There is a close correlation between plasma iron levels, TIBC, and the proportion of sideroblasts in bone marrow. In hemolytic anemias, pernicious anemia, and hemochromatosis, the serum iron level increases and sideroblast number reaches 70% (normal range, 30-50% of total cells). In iron deficiency, the sideroblasts are decreased in number or absent. Before treatment is initiated, the cause of the negative iron balance must be established. Treatment should correct the underlying cause of anemia and improve the iron balance. In general, oral therapy with ferrous salts is

682

CHAPTER 29

satisfactory; however, sometimes parenteral therapy is preferred, e.g., in proven malabsorption problems, gastrointestinal disease and excessive blood loss, and for patients who cannot be relied on to take oral medication.

Iron-Storage Disorders A type of iron storage disorder characterized by general increase in tissue iron levels without damage to parenchymal cells is known as hemosiderosis. Hemosiderin is a storage form of iron in which ferric hydroxide is present as micelles. It appears as insoluble granules that contain denatured aggregated ferritin, nonferritin proteins, lipids, heme, and other pigments. Hemosiderosis results when iron is present in excessive quantities in a diet that permits maximum iron absorption. For example, the African Bantu eat a diet high in corn (low in phosphate) cooked in iron pots, drink an indigenous beer brewed in iron pots, and suffer from Bantu siderosis. Hemosiderosis can progress to hemochromatosis with hepatic cirrhosis and diabetes mellitus.

Metabolism of Iron and Heine

progressive deterioration in pancreatic, hepatic, gonadal, and cardiac function. Clinical manifestations include cirrhosis, diabetes mellitus, life-threatening arrhythmias, and intractable heart failure. Removal of excess iron produces clinical improvement, particularly of diabetes and congestive heart failure. In iron storage diseases accompanied by normal erythropoiesis (e.g., hereditary hemochromatosis), removal of excessive iron is accomplished by repeated bloodletting (phlebotomy). Therapeutic phlebotomy of a unit of blood (which contains about 250 mg of iron) may be performed up to three times per week. When the iron stores become depleted, reaccumulation of iron is prevented by four to six phlebotomies per year. In asymptomatic patients, periodic determination of serum ferritin provides a measure of storage of iron. In hemochromatosis secondary to refractory anemias (e.g., Cooley's anemia, sickle cell anemia), patients require repeated blood transfusions to survive childhood and adulthood. Therapy consists of administration of ironchelating agents. Deferoxamine, an iron chelator isolated from Streptomyces pilosis, has the structure:

H2N--(CH2)5--N--C--(CH2)2--C--N--(CH2)5--N--C--(CH2)2--C--N--(CH2)s--N--C--CH3 I II II I I II II I I il HO O O H HO O O H HO O

Excessive accumulation of iron (chronic iron overload) can result from the following. I. Defective erythropoiesis (dyserythropoiesis); impaired hemoglobin synthesis leading to lack of utilization and consequent accumulation of iron in mitochondria, e.g., from inhibition of ALA synthase activity by dietary vitamin B6 deficiency; inhibition of heme synthesis by lead; impairment of pyridoxine metabolism in alcoholic patients; familial sideroblastic anemias; and Cooley's anemia. 2. Repeated blood transfusions, e.g., in Cooley's anemia or sickle cell disease. 3. Hereditary hemochromatosis, an autosomal recessive defect in which there is increased rate of absorption of iron in the presence of normal or enlarged iron stores and normal hematopoiesis (discussed later). 4. High dietary iron and substances that enhance its absorption (e.g., Bantu siderosis). 5. Hereditary atransferrinemia. In all of these disorders, the gastrointestinal tract cannot limit absorption of iron to significant extent. Thus, the "mucosal block" responsible for keeping out unneeded iron on a daily basis is susceptible to disruption, perhaps at more than one point. Iron overload leads to

It contains six nitrogen atoms separated by fairly long, flexible stretches of methylene groups. Since each iron atom can bind six ligands, one molecule of deferoxamine is probably capable of occupying all six coordination sites and producing a 1:1 iron-deferoxamine complex. For ferric iron, the Kas~ocof deferoxamine is about 103~ while the Ka~oc for Ca 2+ is about 102. Iron in hemoproteins is not affected by this agent, while the ferric iron of ferritin and hemosiderin is chelated in preference to that found in transferrin. Such selectivity makes the compound useful in treatment of iron storage problems and acute iron poisoning. The deferoxamine-iron complex is excreted in urine. (Iron is not normally excreted by this route.) Deferoxamine given orally complexes with dietary iron, making the drug and the iron unavailable for absorption. The preferred route of administration is by intramuscular injection. Irritation and pain at the site of administration and the need for daily injections make the treatment unpopular. In addition, even with coadministration of large amounts of ascorbic acid, the iron loss produced is far below that necessary to remove all of the iron accumulated during chronic transfusion therapy. Slow, continuous intravenous infusion or continuous subcutaneous administration may be more effective in

SECTION29.1 Iron Metabolism

establishing negative iron balance and eliminating stored iron. A small, labile (chelatable) iron pool may be in slow equilibrium with a much larger (storage) pool. When deferoxamine is administered, the labile pool is rapidly emptied. Any deferoxamine that remains in the body or is administered thereafter finds no iron to bind. Thus, most of a single intramuscular dose is simply excreted unchanged. If the chelator is given as a continuous infusion, the labile pool is initially depleted and kept empty. As iron is released from storage sites, it is immediately chelated and removed. Removal of up to 180 mg of iron per day has been accomplished by this method, making it as effective as phlebotomy. Massive intravenous injections of deferoxamine have also been reported to produce excretion of large amounts of iron in iron-overloaded patients.

Hereditary Hemochromatosis Hereditary hemochromatosis is a common inherited autosomal recessive disorder of excessive iron accumulation in parenchymal cells of liver, heart, pancreas, endocrine organs, skin, and joints. The term hemochromatosis is used when organ damage has occurred in the presence of impaired function. It occurs predominately in Caucasian populations; about 1 in 200-400 caucasians are at risk for developing clinical symptoms. Individuals with hereditary hemochromatosis absorb about 3-4 mg/d of iron as compared with a normal rate of about 1-2 mg/d. This excess iron, absorbed over several years, causes accumulation of as much as 20-40 g as compared to normal amounts of about 4 g. In untreated patients, progressive iron accumulation can cause organ damage resulting in hepatic dysfunction, diabetes, cardiomyopathy, hypogonadism with infertility, arthritis, and skin hyperpigmentation. Death can occur due to cirrhosis, diabetes, cardiomyopathy or hepatocellular carcinoma. Hereditary hemochromatosis is associated with a gene on the short arm of chromosome 6 near the MHC gene complex. This gene is known as HFE and codes for HFE protein. The roles of HFE protein along with /~2-microglobulin in the regulation of intestinal iron absorption and iron sequestration in the form of ferritin and hemosiderin have been discussed previously. Gene knockout mice for either HFE protein or/~2-microglobulin develop an iron overload disorder similar to human hemochromatosis, thus substantiating the roles of HFE protein and/~2-microglobulin in iron homeostasis. Two missense mutations (C282Y and H63D) in the HFE gene have been identified in hereditary hemochromatosis. The substitution of a tyrosyl residue for a cysteinyl residue at position 282 results in the loss of formation of a

683

disulfide linkage necessary for the proper association with /~2-microglobulin (Figure 29-1). In the absence of this disulfide linkage, HFE protein fails to reach the normal membrane location and is rapidly degraded. Thus, C282Y is a loss of function (knockout) mutation. The H63D mutation has no effect on the HFE protein's association with flz-microglobulin. However, this mutation may compromise the protein regulation of the interaction between transferrin and its receptor. Population studies among Caucasians have shown that about 1 in 10 are heterozygous for the C282Y mutation. Homozygosity of C282Y has been observed in 85-90% of hereditary hemochromatosis patients, In about 4% of the hereditary hemochromatosis patients, heterozygosity of C282Y and H63D has been observed. There are still unanswered questions concerning hereditary hemochromatosis. For example, some C282Y homozygotes exhibit neither biochemical nor clinical evidence of iron accumulation. On the contrary, some hemochromatosis patients do not possess a C282Y mutation. Thus, other yet-to-beidentified genetic and environmental factors must play a role in the development of hemochromatosis. Other forms of hemochromatosis include neonatal and juvenile types in which biochemical defects have not yet been identified. Patients with aceruloplasminemia resulting from mutations in the ceruloplasmin gene exhibit accumulation of iron in neural and glial cells in the brain, in hepatocytes, and in pancreatic islet cells. Ceruloplasmin, a copper-containing protein, has ferroxidase activity and participates in the release of iron from cells. Aceruloplasminemia associated with iron overload is a different disorder from that of Wilson's disease (hepatolenticular degeneration) in which biliary excretion of copper and incorporation of copper into ceruloplasmin are defective (Chapter 37). Porphyria cutanea tarda, a disorder of porphyrin biosynthesis (discussed later), usually is accompanied by iron accumulation. Thirty percent of patients with porphyria cutanea tarda are either homozygous or heterozygous for the C282Y mutation affecting the HFE protein. Treatment of hereditary hemochromatosis is therapeutic phlebotomy (discussed earlier). This method is safe, effective, and life saving, and ideally should begin before symptoms develop. Serum ferritin levels are used as a surrogate marker for estimating total-body iron stores. Morphologic studies and quantitative determination of iron in liver tissue obtained by biopsy have been used in the assessment of early hereditary hemochromatosis and the degree of liver injury. Finally, hereditary hemochromatosis is a treatable disease. Biochemical screening for the identification of the

CHAPTER29 Metabolism of Iron and Heme

684

disease in the general population has been suggested. The biochemical tests include the measurement of serum levels of iron, transferrin saturation, and ferritin.

29.2 Heme Biosynthesis The principal tissues involved in heme biosynthesis are the hematopoietic tissues and the liver. Biosynthesis requires participation of eight enzymes, of which four (the first and the last three) are mitochondrial and the rest are cytosolic (Figure 29-4). The reactions are irreversible. Glycine and succinate are the precursors of porphyrins. Formation of 6-Aminolevulinic Acid 6-Aminolevulinic acid (ALA) formation is catalyzed by mitochondrial ALA synthase, which condenses glycine and succinyl-CoA to ALA. The enzyme is located on the matrix side of the inner mitochondrial membrane. It is encoded by a nuclear gene and is synthesized in the cytosol on the free polyribosomes as a

COOH I OH2 (~OOH

I

COOH I OH2 Pyridoxal

I

,

I + CH2 phosphate=--CH2 + CO2+ COASH CH2~NH2 I I CmSCoA C~O II I O CH2 I NH2 Glycine Succinyl-CoA 5-Aminolevulinic acid

isonicotinic acid hydrazide; Chapter 17) can prevent heme synthesis and cause anemia. Heme synthesis also requires a functional tricarboxylic acid cycle and an oxygen supply. The primary regulatory step of heme synthesis in the liver is apparently that catalyzed by ALA synthase. The regulatory effects are multiple. The normal end product, heme, when in excess of need for production of heme proteins, is oxidized to hematin, which contains a hydroxyl group attached to the Fe 3+ atom. Replacement of the hydroxyl group by a chloride ion produces heroin. Hemin and heme inhibit ALA synthase allosterically. Hemin also inhibits the transport of cytosolic ALA synthase precursor protein into mitochondria. ALA synthase has a turnover rate of 70 minutes in adult rat liver and is inducible. Its induction is suppressed by hemin and increased by a variety of xenobiotics (e.g., environmental pollutants) and natural steroids. In erythropoietic tissues, where the largest amount of heme is synthesized, regulation of heme biosynthesis may also involve the process of cell differentiation and proliferation of the erythron, which occurs to meet change in requirements for the synthesis of heme. The differentiation and proliferation are initiated by erythropoietin.

Formation of Porphobilinogen Two molecules of ALA are condensed by cytosolic zinc containing ALA dehydratase to

COOH I

CH2

I

2

precursor. The precursor protein is processed to active form during its translocation into mitochondria (Chapter 25). Pyridoxal phosphate is the required coenzyme. The reaction mechanism consists of formation of a Schiff base by pyridoxal phosphate with a reactive amino group of the enzyme; entry of glycine and formation of an enzyme-pyridoxal phosphate-glycine-Schiff base complex; loss of a proton from the r carbon of glycine with the generation of a carbanion; condensation of the carbanion with succinyl-CoA to yield an enzyme-bound intermediate (ol-amino-fl-ketoadipic acid); decarboxylation of this intermediate to ALA; and liberation of the bound ALA by hydrolysis. ALA synthesis does not occur in mature erythrocytes. In experimental animals, deficiency of pantothenic acid (needed for CoASH and, hence, succinyl-CoA synthesis), lack of vitamin B6, or the presence of compounds that block the functioning of pyridoxal phosphate (e.g.,

CH2 I C~O I

-2.~o ~,~ ._ -

OH2 I NH2 ALA

COOH/ ICOOH a c i ~ H2 d /

Acetic

acid

substituent (A)

L(~H2 I C II

Propionic substituent

CH2

(P)

I C II

/C H H2C ~ N / C I H NH2 Porphobilinogen yield porphobilinogen (PBG). There are four zinc ions per

SECTION 29.2

Heine Biosynthesis

685

F I G U R E 29-4 Biosynthetic pathway of heme. The pathway consists of eight irreversible reactions, four each in the mitochondrion and the cytosol. The primary site of regulation is the ALA synthase step.

octamer of the enzyme, and they are bound via the reduced thiol groups. Zinc is required for enzyme activity. The reaction mechanism consists of Schiff base formation by the keto group of one molecule of ALA with the s-amino group of a lysyl residue of the enzyme, followed by nucleophilic attack by the enzyme-ALA anion on the carbonyl group of a second ALA molecule with elimination of water. Then, a proton is transferred from the amino group of the second ALA molecule to the s-amino group of the lysyl residue with formation of PBG. Lead is a potent inhibitor of ALA dehydratase, presumably by displacement of zinc by lead because the leadinhibited enzyme can be reactivated by the addition of zinc. ALA dehydratase is inhibited competitively by succinyl acetone (HOOC-CHz-CHz-CO-CHz-CO-CH3), which occurs in urine and blood in hereditary tyrosinemia (Chapter 17). Genetic deficiency of ALA dehydratase is known to occur.

tetrapyrrole, followed by its cyclization to form uroporphyrinogen III. In the absence of uroporphyrinogen III synthase (e.g., in congenital erythropoietic porphyria), the hydroxymethylbilane cyclizes spontaneously to the uroporphyrinogen I isomer, which is not a precursor of heme (Figure 29-5).

Formation of Coproporphyrinogen III Cytosolic uroporphyrinogen decarboxylase catalyzes successive decarboxylation of the four acetic groups to yield four methyl groups (Figure 29-6).

Formation of Protoporphyrinogen IX Mitochondrial coproporphyrinogen oxidase is localized in the intermembrane space and is probably loosely bound to the outer surface of the inner membrane. It catalyzes the successive conversion of propionic acid groups of ring A and ring B to vinyl groups (Figure 29-7).

Formation of Uroporphyrinogen III Uroporphyrinogen III formation occurs in the cytosol and requires the successive action of porphobilinogen deaminase (or methylbilane synthase) and uroporphyrinogen III synthase. Porphobilinogen deaminase catalyzes condensation of four porphobilinogen molecules in a symmetrical head-to-tail arrangement to form a straight-chain tetrapyrrole, hydroxymethylbilane. Uroporphyrinogen III synthase catalyzes the rearrangement of one of the pyrrole rings (ring D in Figure 29-5) to form an asymmetrical

Formation of Protoporphyrin IX and Heine Both of these steps occur in mitochondria (Figure 29-8). Porphyrinogen oxidase removes six hydrogen atoms (four from methane bridge carbons and two from pyrrole nitrogens) from protoporphyrinogen to yield protoporphyrin. The oxidase has an absolute requirement for oxygen. Protoporphyrinogen can also be oxidized nonenzymatically to protoporphyrin at physiological pH, temperature, and aerobic conditions. Protoporphyrin oxidase is bound to the

686

CHAPTER29 Metabolismof Iron and Heme Ac

P

Porphobilinogen NH2

H J

4NH3~-"I

P

PGBdeaminase Ac

[

Ac

P

Hydroxymethylbilane HO-"-z/T"- NH p

~

A

HN~ c

Ac Uroporphyrinogen-III / synthase / A c

P ~pontaneous p "~

P Ac

P

Ac/ ~ ~Ac ,,/ \ r p UroporphyrinogenIII

(biologicallyusefulisomer)

Ac

Ac

P

P

~

A

c

UroporphyrinogenI (biologicallynot usefulisomer)

FIGURE29-5 Synthesis of uroporphyrinogen I and III. The latteris the biologicallyuseful isomer,and its formationrequiresthe action of uroporphyrinogen-IIIsynthase.Ac,-CH2COOH;P,-CH2CH2COOH. inner mitochondrial membrane, and its active site faces the cytosolic side of the membrane. Formation of heme is accomplished by ferrochelatase (or heme synthase), which incorporates Fe 2+ into protoporphyrin and is inhibited by lead. Zinc can function as a substrate in the absence of iron.

Disorders of Heme Biosynthesis The porphyrias are a group of disorders caused by abnormalities in heme biosynthesis. They are inherited and acquired disorders characterized by excessive accumulation and excretion of porphyrins or their precursors. Defects in any one of the eight enzymes involved in heme biosynthesis may cause inherited porphyrin-related disorders (Figure 29-9). Porphyrins have a deep red or purple color (Greek porphyra - purple). Porphyrins are

excreted by different routes, depending on their water solubility. For example, uroporphyrin with its eight carboxylic group substituents is more water-soluble than the porphyrins derived from it and is eliminated in the urine, whereas protoporphyrin (which contains two carboxylic groups) is excreted exclusively in bile. Coproporphyrin has four carboxylic groups and is found in bile and urine. These disorders are associated with acute or cutaneous manifestations (or both). In the acute state, the presentation may include abdominal pain, constipation, hypertension, tachycardia, and neuropsychiatric manifestations. Cutaneous problems consist of photosensitivity (itching, burning, redness, swelling, and scarring), hyperpigmentation, and sometimes hypertrichosis

SECTION 29.2

HeineBiosynthesis

687

Ac

Porphyria maybe classified as hepatic or erythropoietic. However, enzyme defects are sometimes common to both tissues. Porphyrias can be induced by alcohol, stress, infection, starvation, hormonal changes (e.g., menstruation), and certain drugs. These drugs presumably precipitate acute manifestations in susceptible subjects since they are inducers of cytochrome P-450 and increase the need for synthesis of heme as they deplete the mitochondrial pool of free heme. Major hepatic porphyrias include acute intermittent porphyria, variegate porphyria, hereditary coproporphyria, andporphyria cutanea tarda. The principal erythropoietic porphyrias are hereditary erythropoietic porphyria and erythropoietic

P

Ac

P

~ ' ~ n I~H

H~ 1 ~

UroporphyrinogenIII

AC~Ac P

p

Uroporphyrinogen-III decarboxylase "-'4CO2 M

protoporphyria.

P

M B

~ N H

P

Hepatic Porphyrias

P

HN~

CoproporphydnogenIII

P

FIGURE 29-6 Formationof coproporphyrinogenIII from uroporphyrinogenIII. Acetic acid side chains (Ac) are decarboxylatedto methylgroups (M), sequentially, startingclockwisefromring D. P, -CH2CH2COOH. (an abnormally excessive growth of hair). Four porphyrias can manifest as acute disorders: 6-ALA dehydratase deficiency porphyria, acute intermittent porphyria, hereditary coproporphyria, and variegate porphyria.

p.

M

M

V. P

Acute intermittent porphyria is associated with excessive urinary excretion of ALA and porphobilinogen. The lack of polymerization of porphobilinogen is due to deficiency of porphobilinogen deaminase in several cell types (e.g., hepatocytes, erythrocytes, fibroblasts, lymphocytes). Acute clinical manifestations include neuropsychiatric disorders and abdominal pain. The cause of these manifestations is not clear, but accumulation of porphyrin precursors (ALA and porphobilinogen) in pharmacological amounts has been implicated. Since afflicted subjects cannot make porphyrins to any great extent, they are not photosensitive. This disorder is inherited as an autosomal dominant trait. Porphyria cutanea tarda is the most common form. It is inherited as an autosomal dominant trait and is due to deficiency of uroporphyrinogen III decarboxylase. Clinical

M

M

V. P

M

M

V

P

ff-~-'- NH M

M

P

P

CoproporphyrinogenIII

M

HN"----~

M

P

P

Harderoporphyrinogen

p

p

ProtoporphyrinogenIX

FIGURE 29-7 Formation of protoporphyrinogenIX from coproporphyrinogenIII by coproporphyrinogenoxidase. Sequential oxidative decarboxylationof the propionic acid (P) sidechains of rings A and B produces vinyl (V) groups (V = -CH=CH2). The reactionproceeds via the stereospecificloss of one hydrogenatom and decarboxylationof the propionic acid group. Molecularoxygenis the oxidant, and/~-hydroxypropionateis a probable intermediate. M, CH3.

688

CHAPTER29 Metabolism of Iron and Heme

V,

M rVrotopor.

M /

.......

~NH

M

V, M

V

ox a e. ~~...~.NH -~) phydnogen HN'~

V,

"N_.~

M

M

V

ForrFe~=h: ,a,aso ~ ? e ( N .

~

OH

P

P ProtoporphyrinogenIX

P P ProtoporphyrinIX

P

P Heme

F I G U R E 29-8 Formation of heme. In the reaction catalyzed by protoporphyrinogen oxidase, six hydrogens are removed and the primary electron acceptor is not known, but oxygen is required for enzyme activity. In the terminal step of heme synthesis, only Fee+ is incorporated into protoporphyrin.

manifestations are mild to severe photosensitivity and liver disease. Most affected individuals have increased hepatic iron stores, which can be successfully decreased by phlebotomy. Acute episodes can be precipitated by overindulgence of alcohol or, less frequently, by therapy with estrogen. Hereditary coproporphyria and variegate porphyria are inherited as autosomal dominant traits. They are caused by deficiency of coproporphyrinogen oxidase and protoporphyrinogen oxidase, respectively. In most of these disorders, increased hepatic ALA synthase activity is due to decreased heme synthesis. There are also increased amounts of ALA and porphobilinogen in liver, plasma, and urine and specific

metabolites produced before the metabolic block. ALA synthase is regulated by the heme by a feedback process and by gene repression. Hematin has been used to treat acute attacks with marked success. Although often inadequate, carbohydrate feeding has been associated with improvement in acute intermittent porphyria. This "glucose effect" may depend on repression of the gene for ALA synthase, but the mechanism is not known.

Erythropoietic Porphyrias A defect in synthesis of type III isomers from hydroxymethylbilane, due to deficiency of uroporphyrinogen III

Inheritance & Clinical Manifestations

Glycine + Succinyl-CoA ALAsynthase ALAdehydratase Porphobilinogen deaminase Uroporphyrinogen III synthase Uroporphyrinogen decarboxylase Coproporphyrinogen oxidase ~ Protoporphyrinogen oxidase Ferrochelatase

5-aminolevulinic acid (ALA)

I ALA"yporphyria rataee ciency II acute A" Neur~ episodes

Porphobilinogen

I cute intermittentP~

[Hydroxymethylbilane] ~, UroporphyrinogenIII

I C~

Coproporphyrinogen III

[

II acutel eurovi Y' cera,.ep,soae [

a nerythr~176 d p o r pI [ hsAR' kni yPh~176 ,esions r i a

Porphyria cutanea tarda

[ [AD, Photosensitivityand skin lesions

[ Hereditary c~176176

[ [AD' l eosm se.iNeur~ ~ acuteepisodes skni

ProtoporphyrinogenIX

[

] [AD, l eosm se.iNeurovisceral. o nacute S l i nepisodes skni

Protoporphyrin IX Fe2+ Heme

[Erythropoietic protoporphyria]]AD, Photosensitivity

Variegateporphyria

I

F I G U R E 29-9 Heme biosynthetic pathway and the enzyme defects in various porphyrias. AD, autosomal dominant; AR, autosomal recessive.

I [

[

SECTION29.3 Heme Catabolism

synthase, produces congenital erythropoietie porphyria. Type I porphyrins (principally uroporphyrin I) are formed, accumulate in the tissues, and are excreted in the urine. The deficiency in production of the type III isomer further increases levels of the type I isomers by reducing the regulatory effect on ALA synthase. Excessive amounts of porphyrins in erythrocytes may produce hemolysis. A compensatory increase in hemoglobin formation can then exaggerate the already increased production of type I porphyrins. Their accumulation produces a pink to dark red color in teeth, bones, and urine. Red-brown teeth and urine are pathognomonic. Patients are sensitive to long-wave ultraviolet light and sunlight. The abnormality is transmitted as an autosomal recessive trait. Erythropoietic protoporphyria results from deficiency of ferrochelatase in reticulocytes in bone marrow. It is transmitted as an autosomal dominant trait with variable penetrance and expressivity. In general, it is a benign disorder whose most prominent symptom is photosensitivity. Occasionally, it leads to liver disease. Reduced ferrochelatase activity results in accumulation of protoporphyrin in maturing reticulocytes and young erythrocytes. When a smear of these cells is exposed to fluorescent light, they exhibit red fluorescence. The protoporphyrin appears in the plasma, is picked up by the liver, and is excreted into the bile. Protoporphyrin accumulation in the liver can lead to severe liver disease. In contrast to individuals afflicted by other porphyrias, these patients have normal urinary porphyrin levels. High levels of protoporphyrin are found in erythrocytes, plasma, and feces. The photosensitivity may be caused by stimulation of protoporphyrin in dermal capillaries to an excited (triplet) state by visible light. This in turn converts molecular oxygen to singlet oxygen, which produces cell damage. Oral administration of/3-carotene decreases the photosensitivity, possibly owing to a quenching effect on singlet oxygen and free-radical intermediates.

29.3 Heme Catabolism When heme proteins are degraded in mammals, the polypeptides are hydrolyzed to amino acids while the heme groups are freed of their iron, which is salvaged, and are converted to bilirubin. After transport to the liver, bilirubin is coupled to glucuronic acid and the conjugated bilirubin is excreted into bile as the principal bile pigment. When increased production or decreased excretion of bilirubin causes its plasma concentration to exceed 0.11.0 mg/dL (2-17 #mol/L), it diffuses into tissues and

689 produces jaundice. Although jaundice is relatively harmless unless due to extremely high concentrations of unconjugated bilirubin, it indicates the presence of a disease process that requires medical attention. The yellow coloration of jaundiced skin and sclerae has aroused much interest and has made bilirubin the subject of extensive research. Fractionation and quantitation of serum bilirubin are now widely used for diagnosis and prognosis of hepatobiliary disease. B ilirubin is a waste product and has no known beneficial physiological function. However, both the conjugated and the unconjugated forms of bilirubin show antioxidative properties (e.g., inhibition of lipid peroxidation). The physiological role of the antioxidative property of bilirubin is not known. Bilirubin is a yellow-orange pigment that in its unconjugated form is strongly lipophilic and cytotoxic. It is virtually insoluble in aqueous solutions below pH 8 but readily dissolves in lipids and organic solvents and diffuses freely across cell membranes. Bilirubin toxicity is normally prevented by tight binding to serum albumin. Only when the binding capacity of albumin is exceeded can a significant amount of unconjugated bilirubin enter cells and cause damage. Conjugated bilirubin is hydrophilic and does not readily cross cell membranes, even at high concentrations. Of the 250-300 mg (4,2755,130 #mob of bilirubin normally produced in 24 hours, about 70-80% is derived from hemoglobin. The remainder comes from several sources, including other heme proteins (e.g., cytochromes P-450 and bs, catalase), ineffective hemopoiesis (erythrocytes that never leave the marrow), and "free" heme (heme never incorporated into protein) in the liver. Hemoglobin heme has a life span equal to that of the red cell (about 125 days), whereas heme from other sources (with the exception of myoglobin, which is also quite stable) turns over much more rapidly. Hepatic P-450 enzymes have half-lives of 1-2 days. When radioactively labeled glycine or ALA is injected and radioactivity in fecal bile pigments is monitored, two peaks are seen. The rapidly labeled bilirubin (early bilirubin) peak appears 3-5 days after injection and contains about 15-20% of the injected label. It is increased by drugs that induce hepatic P-450 oxygenases and in erythropoietic porphyria and anemias associated with ineffective erythropoiesis (lead poisoning, thalassemias, and some hemoglobinopathies). Thus, the bilirubin in the early peak is partly derived from these sources. The slowly labeled bilirubin (late bilirubin) peak appears at approximately 120 days, contains about 80-85% of the label, and is due to heine released from senescent erythrocytes.

690

CHAPTER 29

Heme )roteins Proteins-."

Amino[adds (reutilized)

Heme Heine oxygenase system

Mononuclear phagocytic cells of spleen, bone marrow, and liver

Fe=+,~--J" - - C O (reutilized) Biliverdin

NAD(P)H + H+ Urinary urobilinogen (up to ~ rag/d) / Kidney

T

Systemic circulation

Biliverdin reductase "----..BAD(P)+ Bilirqbin (250-300 rag/d) ITransported to the liver complexed to albumin in the plasma

Bilirubinin the hepatocytes (bound predominantly to ligandin)

~

UBG in liver

UDP-glucumnicacid

bin UDPronyltransferase UDP Bilirubin monoglucuronide

UDP-glucuronic acid Bilirubin UDPronyltransferase UDP Secretion into bile

~,~ Enterohepatic circulation

Bilrubin diglucuronide in bile and gall bladder

l

UBG in small bowel

l

Bilirubin diglucuronide in small bowel

l

Bilirubin large bowel ]Metabolism by 1fecal flora ~Ur0bilino~len (UBG) and other compounds Excretion in the feces (50-250 mg of urobilinogen per day)

F I G U R E 29-1(t Catabolic pathway for the heme group from hemoproteins (predominantly hemoglobin).

Formation of Bilirubin A summary of the pathway for bilirubin metabolism and excretion is shown in Figure 29-10. Release of heme from heme proteins and its conversion to bilirubin occur predominantly in the mononuclear phagocytes of liver, spleen, and bone marrow (previously known as the reticuloendothelial system), sites where sequestration of aging red cells occurs. Renal tubular epithelial cells, hepatocytes, and macrophages may also contribute to bilirubin formation under some conditions. Structures of the intermediates in the conversion of heme to bilirubin are shown in Figure 29-11. The initial step after the release of heme is its binding to heme oxygenase, a microsomal enzyme distinct from the microsomal P-450 oxygenases. Heme oxygenase catalyzes what appears to be the rate-limiting step in catabolism of heme. It is induced by

Metabolism of Iron and Heme

heme and requires 02 and NADPH for activity. The activity of the inducible isoenzyme form of heme oxygenase is highest in the spleen, which is involved in the sequestration of senescent erythrocytes. The constitutive form of heme oxygenase is mainly localized in the liver and brain. After binding, the oe-methene carbon of heme is oxidized (hydroxylated) to ol-hydroxyhemin, which undergoes autoxidation to biliverdin (a blue-green pigment) with consumption of O2 and release of iron and carbon monoxide (derived from oxidation of the ot-methene bridge). Since CO production in mammals occurs primarily by this pathway, measurement of expired CO has been used to estimate heme turnover. Values obtained exceed those derived from plasma bilirubin measurements by about 15%, probably because of bilirubin produced in the liver and excreted into the bile without entering the circulation. A potent competitive synthetic inhibitor of heme oxygenase is tin (Sn) protoporphyrin, which has a potential therapeutic use in treatment of neonatal jaundice (see below). In nonmammalian vertebrates, biliverdin is the final metabolite in heme catabolism. Transport of biliverdin is much easier than that of bilirubin because biliverdin is water-soluble. Conversion of biliverdin to bilirubin may have evolved in mammals because, unlike biliverdin, bilirubin readily crosses the placenta. In this way, the fetus can eliminate heme catabolites via the mother's circulation. However, this explanation may not be complete, since the rabbit (a placental mammal) excretes biliverdin as the major bile pigment. B iliverdin is reduced to bilirubin by NAD(P)Hdependent biliverdin reductase, a cytosolic enzyme that acts at the central methene bridge. Although both molecules have two propionic acid groups, the polarity of biliverdin is greater than that of bilirubin. B ilirubin can form six internal hydrogen bonds between the carboxylic groups, the two lactam carbonyl oxygens, and four pyrrolenone ring nitrogens, and thus prevents these groups from hydrogen-bonding with water (Figure 29-12). Biliverdin cannot form these hydrogen bonds because of the lack of free rotation imposed by the double bond at the central methene bridge. Esterification of the propionyl side chains of bilirubin with glucuronic acid disrupts the hydrogen bonds and increases its solubility and lability. "Activators," such as ethanol and methanol, used in the van den Bergh test to measure "indirect bilirubin," and phototherapy for neonatal jaundice also act by disrupting the hydrogen-bonded structure of unconjugated bilirubin. Hemoglobin and heme released from intravascular hemolysis or blood extravasations (e.g., subcutaneous hematomas) are bound, respectively, by haptoglobin and

SECTION 29.3 HemeCatabolism M

691

M

M

P

V

M

p

V N

.'9

~N"'" P~

C

' HeNADPY/_lgenase '~ ,O2

..... ~.

~N-~: H

~

,

M

'

N

I-

M~ Of,-Methene unuge

~N'"'" P~

V

~N~ /

h e m ~ . ~i ~Dn c M M

Heme

OH o~-Hydroxy-

V

Probablyk 02 CO(from~-methene carbon),Fe'+ M M H

nonenzymatic M

M

C P

Central methene bridge, P

V

H2C

NH

V

O Biliverdinreductase ~H~,.,.~ -~~O NAD(P) + r- - - NAD(P)H~ +H+ -~ HN~ N M

M

O Biliverdin _.~O HN

p

V

M

M

V

Bilirubin

FIGURE 29-11 Conversion of heme to bilirubin in the monocytic phagocytic cells. Carbon monoxide and bilirubin are generated. Fe3+ released is conserved and reutilized. Biliverdin and bilirubin are lactams. P, Propionic acid; M, methyl; V, vinyl.

hemopexin V

g H"

M

C--O

"..

~)15

M M

-. M

~0

ii:::o=

................ H / 0

FIGURE 29-12 Conformation of bilirubin showing involutedhydrogenbonded-structure between NH/O and OH/O groups. Despite the presence of polar carboxyl groups, bilirubin is nonpolar and lipophilic. Disruption of hydrogen bonds by glucuronidation or by conversion of bilirubin to configurational or structural isomers yields water-solublepigments.

to form complexes that cannot be filtered by the kidney. This action prevents renal loss of the heme iron and protects the renal tubules from possible damage by precipitated hemoglobin. Haptoglobinhemoglobin and hemopexin-heme complexes are processed in mononuclear phagocytic cells in a way similar to that for hemoglobin. Haptoglobin and hemopexin are glycoproteins synthesized in the liver. The former is an o~2-globulin and an acute-phase reactant (i.e., its synthesis and release into the circulation are augmented during an acute insult to the body); the latter is a ill-globulin but not an acute-phase protein (see also Appendix VI).

C i r c u l a t o r y T r a n s p o r t of Bilirubin B ilirubin formed in extrahepatic tissues is transported to the liver for excretion in bile. Since bilirubin is virtually insoluble in aqueous media, it is transported to the liver bound noncovalently to serum albumin. The bilirubinalbumin complex increases the amount of bilirubin carried

692

per volume of plasma and minimizes diffusion of bilirubin into extrahepatic tissues, thereby preventing bilirubin toxicity. Because of formation of this complex, bilirubin does not normally appear in urine. Urinary bilirubin is almost invariably conjugated bilirubin (see below) and signifies the presence of a pathological process. An albumin molecule binds two molecules of bilirubin at one high-affinity site and at one to three secondary sites. Bilirubin conjugated with glucuronic acid also binds to albumin but with much lower affinity. Another form of bilirubin (probably conjugated), very tightly (probably covalently) bound to albumin, has been described. The mechanism of its formation is not known, although blockage of biliary flow associated with an intact hepatic conjugating system releases a chemically reactive form of bilirubin into the circulation. If the capacity of albumin to bind bilirubin is exceeded because of increased amounts of unconjugated bilirubin or decreased concentration of albumin, bilirubin readily enters extrahepatic tissues. In neonates, this can cause kernicterus, a serious condition associated with permanent neurological damage (see below). Bilirubin can be displaced from binding to albumin by sulfonamides, salicylates (notably aspirin), and cholangiographic contrast media. Use of these substances in jaundiced newborn infants increases the risk of occurrence of kernicterus. Medium-chain fatty acids increase and short-chain fatty acids decrease bilirubin binding to albumin. Binding, at least to the primary site, is independent of pH. Estimation of reserve bilirubin binding capacity has been used to evaluate the risk of bilirubin toxicity in icteric patients.

Hepatic Uptake, Conjugation, and Secretion of Bilirubin Hepatocytes take up bilirubin from the sinusoidal plasma and excrete it after conjugation with glucuronic acid across the canalicular membrane into the bile. The entry and exit steps and the transport of bilirubin within the cell are not completely understood. The following is a plausible interpretation of the available data. Since binding of bilirubin to albumin is usually reversible, a small amount of free bilirubin is present in plasma in equilibrium with albumin-bound bilirubin. It is probably this free bilirubin that is taken up at a rate determined by its plasma concentration. As this free bilirubin concentration decreases, more bilirubin is released from albumin and becomes available for uptake. Alternatively, the albumin-bilirubin complex may bind to specific hepatocyte plasma membrane receptors, and thereby bilirubin is released to enter the cell. Both models are consistent

CHAPTER 29

Metabolism of Iron and Heme

with the finding that albumin does not accompany bilirubin into the hepatocyte. The entry step seems to be carrier-mediated, is saturable, is reversible, and is competitively inhibited by sulfobromophthalein, indocyanine green, cholecystographic agents, and several drugs. Bile salts do not compete with bilirubin for hepatic uptake. After it enters hepatocytes, bilirubin is transported to the smooth endoplasmic reticulum for glucuronidation bound to a protein. Two cytosolic proteins, Z protein (fatty acid-binding protein) and ligandin (Y protein), bind bilirubin and other organic anions. Ligandin, which constitutes about 2-5% of the total soluble protein in rat and human liver, has lower capacity but higher affinity for bilirubin than Z protein. Ligandin (M.W. 47,000) has two subunits, A and B, which appear to be identical except for a 30-amino-acid extension at the carboxyl terminus of the B subunit. Bilirubin is bound entirely to the A subunit (two molecules per A subunit). Ligandin also has glutathione S-transferase, glutathione peroxidase, and ketosteroid isomerase activities, which depend on both subunits. Glutathione S-transferases catalyze detoxification reactions for a number of substances. Binding of bilirubin and other organic anions to ligandin occurs at sites unrelated to its enzyme activities. Under normal conditions, ligandin is probably the principal hepatic bilirubin-binding protein and may serve intracellularly the same protective and transport functions as albumin in plasma. It may also help limit reflux of bilirubin into plasma, since its affinity for bilirubin is at least five times greater than that of albumin. Z protein (M.W. 11,000) becomes important at high plasma bilirubin concentrations. The concentration of ligandin in the liver does not reach adult levels until several weeks after birth, whereas neonatal and adult levels of Z protein are the same. This lack of ligandin, together with low hepatic glucuronyltransferase activity, is the probable cause of transient, "physiological," nonhemolytic, neonatal

jaundice. Glucuronidation of bilirubin in the endoplasmic reticulum by UDP-glucuronyltransferase produces an ester between the 1-hydroxyl group of glucuronic acid and the carboxyl group of a propionic acid side chain of bilirubin (Figure 29-13). In bile, about 85% of bilirubin is in the diglucuronide form and the remainder is in the monoglucuronide form. Glucuronidation increases the water solubility of several lipophilic substances. There appear to be many UDP-glucuronyltransferases in the endoplasmic reticulum, which differ in substrate specificity. (Biosynthesis of UDP-glucuronic acid was described in Chapter 15.)

SECTIO29.3 N HemeCatabolism

693

COOH

HOc'~~'O

O%C O"~OH "~,COOH OH HO

O%cO /H O / H O~Oc'~O

#

~ H HN~ :u~AUr~

v@o M

ranSu::"

~ H HN-~/~

V%ooX

V

M

Bilirubin

V

Bilirubin diglucuronide

FIGURE 29-13 Formation of bilirubin diglucuronide. Glucuronidation occurs in two steps via formation of monoglucuronide. Monoand diglucuronides are more water-soluble and less lipophilic than bilirubin. Conversion of bilirubin to water-soluble products is obligatory for excretion of bilirubin from hepatocytes. M, Methyl; V, vinyl; UPD-GA, UDP-glucuronic acid.

natural isomer of bilirubin (Figure 29-12). Thus, they are more polar and readily excreted in the bile without the requirement for glucuronidation. Lumirubin, a structural isomer of bilirubin, is formed by light-induced intramolecular cyclization of the vinyl side group of C-3. It contains a seven-membered ring, is stable, is polar, and is excreted without conjugation. These observations explain the mechanism of phototherapy commonly used for treatment of neonatal hyperbilirubinemia.

Secretion across the canalicular membrane into bile appears to be the rate-limiting step in hepatic bilirubin metabolism. It is probably carrier-mediated, requires energy, is saturable, and is unaffected by bile salts. Bilirubin can be made water-soluble by conversion to its configurational isomers. These photobilirubins are formed when bilirubin is exposed to blue light of the 400- to 500-nm wavelength (Figure 29-14). Photobilirubins cannot form the intramolecular hydrogen bonds characteristic of the

HOOC

COOH

H

.o/o \OOH

H

H

H 4E, 15Z

4Z, 15Z (natural

bilirubin bilirubin)

H HOOC

bilirubin

H HOOC

4Z, 15E

COOH

bilirubin

FIGURE 29-14 Photoisomers of bilirubin. The presence of two methene bridges containing double bonds (colored areas) gives rise to configurational (geometrical) isomers of bilirubin. Each double bond can exist in the Z or E configuration. The naturally occurring, most stable, water-insoluble form is the Z, Z isomer. It undergoes photoisomerization to configurational isomers (Z, E; E, Z; and E, E), which are more polar owing to inability to form intramolecular hydrogen bonds and are excretable from the liver without glucuronidation. Some excretion of photoisomers in urine also occurs.

COOH

H

H 4E, 15E bilirubin

H

694

Bilirubin in the Intestinal Tract

CHAPTER 29

Metabolism of Iron and Heme

The plasma of normal subjects contains 0.1-1 mg of bilirubin per deciliter (2-17 #mol/L), mostly in the unconjugated form. Unconjugated bilirubin is known as indirect-reacting bilirubin and conjugated bilirubin as direct-reacting bilirubin (see Table 29-2). Jaundice occurs when plasma becomes supersaturated with bilirubin (>2-2.5 mg/dL) and the excess diffuses into the skin, sclera, and other tissues. The sclera is particularly affected because it is rich in elastin, which has a high affinity for bilirubin. Reddish yellow pigments, particularly carotene and lycopene, may give a yellowish tinge to the skin but they do not usually produce scleral coloration. Hyperbilirubinemia may result from elevation of unconjugated or conjugated bilirubin levels.

of these cells to store, transport, or conjugate bilirubin. Bilirubinuria does not accompany these disorders. Except in infancy or when pigment gallstones form, unconjugated hyperbilirubinemias are benign. Gilbert's syndrome may be the most common cause of mild, persistent, nonhemolytic, unconjugated hyperbilirubinemia. Serum bilirubin concentration rarely exceeds 5 mg/dL and usually fluctuates between 1.3 and 3 mg/dL. Other liver function tests are normal. The syndrome is usually asymptomatic and is detected during routine laboratory testing or examination for other disease. Family studies suggest that Gilbert's syndrome is an autosomal dominant disorder. The unconjugated hyperbilirubinemia in Gilbert's syndrome is due to decreased UDP-glucuronyltransferase activity resulting from an insertion mutation found in the promoter region of the enzyme. The wild-type promoter [TA]6TAA is mutated to [TA]vTAA. Mutations affecting the coding region of the enzyme, although rare, also occur. In Crigler-Najl'ar syndrome type/activity of hepatic bilirubin UDP-glucuronyltransferase is undetectable and bilirubin conjugates are absent from the serum, bile, and urine, but biliary secretion of sulfobromophthalein and indocyanine green is normal. The disease is apparent shortly after birth, kernicterus develops, and death commonly occurs during the neonatal period. The effectiveness of phototherapy is often transient. The enzyme is not inducible by phenobarbital. This autosomal recessive defect occurs in all races. The Gunn strain of Wister rats has a similar genetic defect and has been used to study the syndrome. Crigler-Najj'ar syndrome type H (Arias syndrome) is milder, usually benign, and caused by partial deficiency of bilirubin UDP-glucuronyltransferase. Jaundice may not appear until the second or third decade of life. The monoglucuronide is the predominant pigment in bile. Phenobarbital induces the enzyme. Dominant and recessive inheritance patterns have been described. An accurate diagnosis of type 1, as opposed to type 2 Crigler-Najjar syndrome, is essential since orthotopic liver transplantation is an important therapy for type 1 patients.

Unconjugated Hyperbilirubinemias

Conjugated Hyperbilirubinemias

Unconjugated hyperbilirubinemias result from imbalance between the rates of production of pigment and of its uptake or conjugation in the liver. Because of the large reserve capacity of the liver for conjugation and excretion of bilirubin, increased production seldom elevates unconjugated serum bilirubin to more than 3-4 mg/dL. If a greater increase occurs, some degree of liver dysfunction probably also occurs. These disorders are usually due to decreased uptake of pigment by hepatocytes or to failure

Conjugated hyperbilirubinemias are due to intra- or extrahepatic reduction to bile flow (cholestasis) with spillage of conjugated bilirubin into the bloodstream, which may occur from injury to the endothelial cells lining bile ductules or from reverse pinocytosis, by the hepatocytes. Since the serum bilirubin is mostly the water-soluble glucuronide, bilirubinuria is usually present. Abdominal tumors, gallstones, strictures, hepatitis, and cirrhosis can mechanically block the biliary canaliculi or

Most bilirubin entering the intestine in bile is in the diglucuronide form, which is very poorly absorbed in the small and large intestines. In the lower small intestine and colon, bacteria remove glucuronic acid residues and reduce bilirubin to the colorless urobilinogen and stercobilinogen. Exposure to air oxidizes these to urobilin and stercobilin, respectively, (i.e., red-orange pigments that contribute to the normal color of stool and urine). Other degradation products of bilirubin are present in minor amounts in feces. Urobilinogen is excreted mostly in the feces, but a small fraction is absorbed from the colon, enters the portal circulation, is removed by the liver, and is secreted into bile. That which is not removed from the portal blood by the liver enters the systemic circulation and is excreted by the kidneys. Urobilinogen excretion in urine normally amounts to 1-4 mg per 24 hours, as opposed to the 40-280 mg (67-470 #mob excreted in feces. Lack of urobilinogen in the urine and feces indicates biliary obstruction; stools are whitish ("clay-colored") owing to the absence of bile pigment. Urinary and fecal urobilinogen excretion increases in hemolytic anemia.

Disorders of Bilirubin Metabolism

695

SECTION29.3 Heme Catabolism

ducts. If obstruction affects only intrahepatic bile flow, hyperbilirubinemia occurs when 50% or more of the liver is involved. Extrahepatic obstruction elevates serum bilirubin only if it increases the pressure in the canaliculi above the maximum secretion pressure of the hepatocytes (about 250 mm Hg). Nonmechanical cholestasis can be caused by bacterial infection, pregnancy, and sex steroids and other drugs, or it may be genetically determined. In cholestasis, bile salts and bile pigments are retained and appear in the circulation, and steatorrhea and deficiencies of fat-soluble vitamins may occur. These deficiencies are often manifested as hypoprothrombinemia (from lack of vitamin K) and osteomalacia (from lack of vitamin D). The magnitude depends on the degree of obstruction. If blockage is complete, urinary urobilinogen will be absent and the stools will have a pale, clay-like color. Familial diseases include Dubin-Johnson syndrome, Rotor's syndrome, and benign familial recurrent cholestasis. Serum bilirubin values in Dubin-Johnson syndrome are the same as those found in Rotor's syndrome (Table 29-2). Very little is known about benign familial recurrent cholestasis. All three disorders are uncommon or rare, and all are benign. The liver in Dubin-Johnson

syndrome appears black on direct examination. The pigment is not identical to melanin but may be a catecholamine (perhaps epinephrine) polymer. The total urinary coproporphyrin excretion is normal but about 80% is the I isomer and the rest is the III isomer, the reverse of the normal ratio. This abnormality seems to be diagnostic for the syndrome, provided the history and physical examination are consistent with the diagnosis. Obligate heterozygotes have urinary coproporphyrin patterns intermediate between those of normal and affected individuals, indicating autosomal recessive inheritance. The hyperbilirubinemia, hepatic pigment, and urinary coproporphyrin abnormality may result from a defect in porphyrin biosynthesis. Studies of sulfobromophthalein clearance show normal storage but greatly decreased secretion. Investigation of a mutation in Corriedale sheep, which causes a similar condition, may help clarify this poorly understood disorder. In Rotor's syndrome, patients lack the hepatic pigment but have urinary coproporphyrin levels 2.5-5 times greater than normal. Coproporphyrin type I is increased relative to the type III isomer but not as much as in Dubin-Johnson syndrome. Intrahepatic storage of sulfobromophthalein is

TABLE 29-2 Serum Bilirubin Levels in Normal and Some Abnormal Conditions

Bilirubin Values in mg/dL of Serum* Condition Newborn 1 day 3 days 7 days Normal (adult) Hemolytic disorders (in adults) Gilbert's disease (probably a heterogeneous group of diseases) Glucuronyltransferase deficiency Type I: Crigler-Najjar syndromem complete deficiency Type II: Crigler-Najjar syndromem partial deficiency Dubin-Johnson syndrome Cholestasis (severe form) Cirrhosis (severe) Hepatitis (acute/severe) *To convert mg/dL to/xmol/L, multiply by 17.1.

Unconjugated (indirect reacting)

Conjugated (direct reacting)

0.1-0.3 0.2-0.4 Normal

(rarely >5)

0.2-0.7 2-3 1.1-2.7 (rarely >5)

15-48

15-48

Negligible

6-22 (up to 40 with physiological stress) 2.5-20 10 11 10

6-22 (up to 40 with physiological stress) < 50% of total 19 56 1.5

Trace

Total 0.2-2.9 0-6 0.3-11 0.1-9.9 0.1-1 2.2-3.4

1.2-3

>50% of total

8.5

696

markedly decreased, but the rate of secretion is only moderately depressed. Inheritance appears to be autosomal recessive.

Neonatal Hyperbilirubinemia Normal neonates are frequently hyperbilirubinemic (Table 29-2). Birth interrupts normal placental elimination of pigment, and the "immature" liver of the neonate must take over. Normally serum bilirubin levels rise on the first day of life, reaching a maximum (rarely greater than 10 mg/dL) by the third or fourth day. This type is mostly unconjugated. If the placenta is functioning normally, jaundice will not be present at birth. If jaundice is present at birth, a cause other than hepatic immaturity must be sought. The primary blocks to bilirubin metabolism are low activity of bilirubin glucuronyltransferase and low concentration of ligandin in the liver at birth. Secretion of conjugated bilirubin into the bile is also reduced. Hepatic immaturity may be partly due to diversion in utero of blood from the liver by the ductus venosus. When this channel closes shortly after birth and normal hepatic blood flow is established, concentrations of a number of substances rise within the hepatocytes and may induce enzymes needed for their metabolism. Accumulation ofbilirubin in plasma may play an important role in hastening the maturation. Although the liver normally matures within 1-2 weeks after birth, hypothyroidism can prolong this process for weeks or months. The neonate is at risk for kernicterus if the serum unconjugated bilirubin level is higher than 17 mg/dL. Kernicterus is characterized by yellow staining of clusters of neuronal cell bodies in the basal ganglia, cerebellum, and brain stem, leading to motor and cognitive deficits or death. Immaturity and perhaps hypoxia make the bloodbrain barrier permeable to bilirubin and contribute to the likelihood of kernicterus. The biochemical basis of bilirubin encephalopathy is due to many causes: inhibition of RNA and protein synthesis, carbohydrate metabolism (both cAMP-mediated and CaZ+-activated), phospholipiddependent protein kinases, enzymes involved in the electron transport system, and impaired nerve conduction. A major complicating factor can be hemolytic anemia such as that of erythroblastosis fetalis caused by Rh incompatibility between mother and child. The hemolysis increases the rate of bilirubin formation, which soon overwhelms the liver and produces severe jaundice and kernicterus. Sickle cell anemia has a similar effect. Congenital absence of bilirubin UDP-glucuronyltransferase (CriglerNajj ar syndrome type 1) usually causes a kernicterus that is fatal shortly after birth. Inhibition of glucuronyltransferase

CHAPTER 29

Metabolism of Iron and Heme

by various drugs (e.g., novobiocin) or toxins can increase the severity of neonatal jaundice. "Breast milk jaundice" is due to the presence in breast milk of a substance (perhaps pregnane-3o~,20/3-diol) that inhibits bilirubin glucuronyltransferase, although the resulting unconjugated hyperbilirubinemia is seldom serious enough to cause neurotoxicity or to require discontinuation of breast-feeding. Other risk factors for pathologic hyperbilirubinemia include Gilbert's syndrome (discussed earlier) and glucose6-phosphate dehydrogenase deficiency (Chapter 15). Conjugated hyperbilirubinemia is rare during the neonatal period. It can result from impaired hepatocellular function or extrahepatic obstruction. Hepatocellular defects can be caused by bacterial, viral, or parasitic infections, cystic fibrosis, ot 1-antitrypsin deficiency, DubinJohnson and Rotor's syndromes, and other genetic disease. Extrahepatic obstruction can be congenital (biliary atresia) or acquired. Treatment of neonatal jaundice is usually by phototherapy. A decrease in bilirubin production in the neonatal period can also be achieved by inhibiting the rate-limiting enzyme of bilirubin formation from heme, namely, the heme oxygenase. A potent competitive inhibitor of heme oxygenase is the synthetic heme analogue tin (Sn 4+) protoporphyrin. When administered parenterally, the tin protoporphyrin safely decreases bilirubin formation. Exchange transfusions also rapidly decrease plasma bilirubin levels.

Supplemental Readings and References B. R. Bacon, J. K. Olynyk, E. M. Brunt, et al.: HFE genotype in patients with hemochromatosis and other liver disease. Annals oflnternal Medicine 130, 953 (1999). N. C. Andrews: Disorders of iron metabolism. New England Journal of Medicine 341, 1986 (1999). J. D. Arnold, A. D. Mumford, J. O. Lindsay, et al.: Hyperferritinaemia in the absence of iron overload. Gut 41,408 (1997). M. C. Augustine: Hyperbilirubinemia in the healthy term newborn. Nurse Practitioner 24, 24 (1999). B. R. Bacon, L. W. Powell, P. C. Adams, et al.: Molecular medicine and hemochromatosis: at the crossroads. Gastroenterology 116, 193 (1999) J. D. Bancroft, B. Kreamer, and G. R. Gourlev: Gilbert syndrome accelerates development of neonatal jaundice. Journal of Pediatrics 132, 656 (1998). T. H. Bothwell and A. P. MacPhall: Hereditary hemochromatosis: etiologic. pathologic, and clinical aspects. Seminars in Hematology 35, 55 (1998). S. S. Bottomley: Secondary iron overload disorders. Seminars in Hematology 35, 77 (1998). N. Chalasani, N. R. Chowdhury, J. R. Chowdhury, et al.: Kernicterus in an adult who is heterozygous for Crigler-Najjar syndrome and homozygous for Gilbert-type genetic defect. Gastroenterology 112, 2099 (1997). M. E. Conrad: Introduction: iron overloading disorders and iron regulation. Seminars in Hematology 35, 1 (1998). R. W. I. Cooke: New approach to prevention of kernicterus. Lancet 353, 1814(1999).

Supplemental Readings and References E A. Dennery, D. S. Seidman, and D. K. Stevenson: Neonatal Hyperbilirubinemia. New England Journal of Medicine 344, 581 (2001 ). C. Q. Edwards, L. M. Griffen, R. S. Ajioka, et al.: Screening for hemochromatosis: phenotype versus genotype. Seminars in Hematology 35, 72 (1998). G. H. Elder, R. J. Hift, and E N. Meissner: The acute porphyrias. Lancet 349, 1613 (1997). B. M. Gaston: Emissions testing for children, chapter 2: carbon monoxide. Journal of Pediatrics 135, 537 (1999). Y. Haimi-Cohen, E Merlob, T. Marcus-Eidlits, et al.: Dubin-Johnson syndrome as a cause of neonatal jaundice: the importance of coporphyrins investigation. Clinical Pediatrics 37, 511 (1998). R. Kauppinen and E Mustajoki: Prognosis of acute porphyria: occurrence of acute attacks, precipitating factors, and associated diseases. Medicine 71, 1 (1992). Y. Maruo, H. Sato, T. Yamano, et al.: Gilbert syndrome caused by a homozygous missense mutation (Tyr486Asp) of bilirubin UDPglucuronyltransferase gene. Journal of Pediatrics 132, 1045 (1998). J. M. McCord: Iron: free radicals and oxidative injury. Seminars in Hematology 35, 5 (1998). G. Monaghan, A. McLellan, A. McGeehan, et al.: Gilbert's syndrome is a contributory factor in prolonged unconjugated hyperbilirubinemia of the newborn. Journal of Pediatrics 134, 441 (1999).

697 E. Pollitt: Early iron deficiency anemia and later mental retardation. American Journal of Clinical Nutrition 69, 4 (1999). P. Ponka, C. Beaumont, and D. R. Richardson: Function and regulation of transferrin and ferritin. Seminars in Hematology 35, 35 (1998). R. D. Press: Hemochromatosis: a simple "genetic" trait. Hospital Practice 34, 55 (1999). Report of a Meeting of Physicians and Scientists at the Royal Free Hospital School of Medicine: Genetic hemochromatosis. Lancet 349, 1688 (1997). E E Rubaltelli, A. Novello, L. Zancan, et al.: Serum and bile bilirubin pigments in the differential diagnosis of Crigler-Najjar disease. Pediatrics 553, (1994). A. Tefferl, L. Solberg, and R. Ellefson: Porphyrias: clinical evaluation and interpretation of laboratory tests. Mayo Clinic Proceedings 69, 289 (1994). C. G. Uasuf, A. Jatakanon, A. James, et al.: Exhaled carbon monoxide in childhood asthma. Journal of Pediatrics 135, 569 (1999). J. N. Umbreit, M. E. Conrad, E. G. Moore, et al.: Iron absorption and cellular transport: the mobilferrin/paraferritin paradigm. Seminars in Hematology 35, 13 (1998). C. Utzel and M. E. Conrad: Absorption of heme iron. Seminars in Hematology 35, 27 (1998).

CHAPTER

30

Endocrine Metabolism 1: Introduction

Survival of multicellular organisms depends on integration and coordination of differentiated cell functions and the ability to react appropriately to internal and external influences that threaten to disrupt homeostatic conditions. These requirements are fulfilled by a form of intercellular communication in which chemical signals (messengers) released by one cell evoke a receptor-mediated response in another. There are two types of chemical messenger: neurotransmitters and hormones. Neurotransmitters convey signals from one neuron to another or from a neuron to an effector cell, travel very short distances to reach their target sites, and function within the specialized regions of synapses and junctions. Hormones are usually defined as messengers that are transported by the blood to distal target cells. Because they are released into the interstitial space and thence into blood, they are called "endocrine" (ductless; "secreted within") secretions to distinguish them from those which are released into the external environment ("exocrine" or ductal secretions). This terminology and these definitions, though not always appropriate and possibly misleading, are used for want of better alternatives. The term "hormone" comes Endocrine topics not discussed in Chapters 30-34 that are covered elsewhere in the text are as follows: gastrointestinal hormones, Chapter 12; eicosanoids, Chapter 18; pancreatic hormones, Chapter 22; parathyroid hormone and vitamin D, Chapter 37; renin-angiotensin system and antidiuretic hormone, Chapters 32 and 39. A list of expanded acronyms appears in Appendix VIII.

from the Greek hormaein, "to excite." This is a misnomer because some hormones have primarily inhibitory effects, and others may stimulate or inhibit, depending on the target cell type. The term endocrine is misleading because some hormones (steroids, prostaglandins, growth factors) are released via ducts to the external environment, where they affect reproductive activities or wound healing. The designation pheromones may apply to some of these hormones (steroids, prostaglandins) because they act on cells in another organism or individual. The usual definition of a hormone excludes those messengers which affect cells in the vicinity of their release and which do not rely on transport in the blood to reach their destination. For these, the term parahormone (paracrine secretion, local hormone, autacoid) is commonly used. However, if produced in sufficiently large amounts, parahormones diffuse into the vascular compartment and behave as authentic hormones (e.g., somatomedins). Conversely, hormones frequently assume paracrine functions. For example, androgens produced by Leydig cells of the testes diffuse and exert their effect on nearby Sertoli cells. Parahormones, therefore, should be regarded as authentic hormones that exert mainly local effects. Finally, neurotransmitters and hormones differ in respect to the systems that use them. Thus, the nervous and endocrine systems share some messengers (norepinephrine, dopamine, enkephalins, endorphins, cholecystokinin, bombesin, etc.) that exert receptor-mediated effects. The different events that are

699

700

CHAPTER 30

triggered by the same substance clearly manifest the different specialized cell functions that are affected. Hormones and neurotransmitters may have evolved from the same, or similar, ancestral prototypes in unicellular organisms. Substances that resemble mammalian hormones in structure and biological activity serve as neurotransmitters in some invertebrates and lower vertebrates. Several peptide and steroid hormones long considered to be evolutionarily advanced mammalian forms have been found in certain prokaryotes and unicellular eukaryotes. Several mammalian brain-gut peptides occur in the skin of amphibians. Specific receptor sites for mammalian hormones are demonstrable at all stages of phyletic evolution. Thus, the structures of hormones and neurotransmitters and their functions as chemical messengers may have been highly conserved during evolution. The fact that many prototypic messengers in unicellular organisms predate the appearance of nerve cells implies that neurotransmitters may be a specialized form of hormone. Chemically, hormones are of four types:

1. 2. 3. 4.

Endocrine Metabolism I: Introduction

Hormonal amine, Peptide, protein, or glycoprotein, Steroid, and Eicosanoid.

Amine hormones and some agents of the second type have counterparts in the nervous system that function as neurotransmitters.

30.1 Hormonal Amines Hormonal amines are derived from amino acids and, in most cases, represent simple modifications of the parent compound. Table 30-1 lists the important hormonal amines, parent amino acids, major sites of synthesis, and principal actions. All of these amines except the thyroid hormones (Chapter 33) are decarboxylated products that are synthesized both in and out of the nervous system. Within the nervous system, they are important neurotransmitters; outside the nervous system, the cells that produce

TABLE 30-1 Hormonal Amines

Hormone

Parent Amino Acid

Dopamine

Tyr

Hypothalamus; adrenal medulla

Yes

Epinephrine

Tyr

Adrenal medulla

Minor

Norepinephrine

Tyr

Peripheral nerve endings; adrenal medulla

Yes

Histamine

His

Mast cells; basophils; regenerating cells

Yes

Serotonin

Trp

Mast cells; liver; sympathetic nerve endings; stored platelets

Yes

3,5,3'-Triiodothyronine (T3)1

Tyr

Peripheral conversion from thyroxine (T4); thyroid

1An amino acid.

Major Site of Synthesis

Neurotransmitter in Nervous System

Systemic Action

Renal vasodilation (D1) inhibition of anterior pituitary hormone release (D 2); inhibition of aldosterone synthesis (D 1)" Cardiovascular and metabolic effects (via ~- and /3-adrenergic receptors). Cardiovascular and metabolic effects (via c~- and /3-adrenergic receptors). Systemic vasodilation (H l , H2); increased capillary permeability (H l); increased gastric secretion (H2); increased heart rate (H 2). Vasodilation in skin and skeletal muscle, vasoconstriction elsewhere; positive inotropic and chronotropic effects. Many effects, e.g., regulation of metabolism.

SECTION30.2 Peptide, Protein, and Glycoprotein Hormones

them are modified postsynaptic neurons (e.g., adrenal medulla), blood-derived cells (e.g., basophils, mast cells), or APUD 1 cells (e.g., enterochromaffin cells). Regardless of where they are released, hormonal amines exert their effects through specific receptor sites located in various parts of the body. These systemic receptors (Table 30-1) and their subtypes probably have identical counterparts in the central and peripheral nervous systems. All of these amines except T3 exert rapid systemic effects that usually involve smooth-muscle activity. Because the amines are hydrophilic, their receptors are located on the outer surface of target cells, and most if not all of their effects are mediated by intracellular mediators.

30.2 Peptide, Protein, and Glycoprotein Hormones Cells that produce peptide, protein, or glycoprotein hormones are derived embryologically from the entoderm or ectoblast (progenitor of ectoderm and neuroectoderm). More than 40 hormones have been identified, containing from three to over 200 amino acid residues. Table 30-2 lists the important hormones and hormonal candidates, number of amino acid residues, and sites of synthesis. All hormonal peptides and many hormonal proteins are synthesized as part of a larger molecule (preprohormone) that contains a leader sequence (signal peptide) at its amino terminal end. The leader sequence is removed as the nascent precursor enters the lumen of the endoplasmic reticulum, and the resultant prohormone undergoes posttranslational processing after being packaged into secretory granules by the Golgi complex. Posttranslational processing involves proteolytic cleavage at specific sites, usually paired basic amino acid residues (Lys-Arg, ArgLys, Arg-Arg, Lys-Lys), by endopeptidases within the secretory granule. In the synthesis of insulin, for example, removal of the 23-amino-acid leader sequence of preproinsulin results in proinsulin, which has two pairs of basic residues (Lys-Arg and Arg-Arg) that are cleaved to yield insulin and the C-peptide (Chapter 22). Likewise, the endogenous opiates arise from site-specific cleavages of their respective prohormones (Chapter 31). When the cell is stimulated to secrete, all major fragments of the prophormone, active and inactive, are released by calciumdependent exocytosis. 1APUD (amine precursor uptake and decarboxylation) cells are derived from the neuroblast (stem cells that give rise to nerve cells and neural crest cells) or the entoderm. They have the ability to synthesize and release peptide hormones and, as their name implies, take up amine precursors (e.g., dopa) and decarboxylate them, producing hormonal amines (e.g., dopamine).

701 Although several peptide hormones have multiple anatomical sites of synthesis, there is usually only one important source of the circulating hormone. The presence of the same hormone in ancillary sites (Table 30-2) may indicate that it functions as a local hormone at those sites. For example, somatostatin produced by the hypothalamus is transported by blood to the anterior pituitary, where it inhibits the release of growth hormone (GH); somatostatin produced by the hypothalamus also functions as an inhibitory neurotransmitter when released into synapses in the central nervous system; somatostatin produced by pancreatic islet cells acts locally to inhibit the release of insulin and glucagon; and somatostatin produced by the gastrointestinal mucosa acts locally to inhibit the secretion of gastrin, secretin, and gastric inhibitory polypeptide (GIP). Other "brain-gut" peptides(Chapter 31) also exemplify this point, although they are not released into the general circulation in significant amounts. Peptide hormones, because of their high information content,may have evolved from a limited number of ancestral molecules that functioned as intercellular messengers or as extracellular enzymes. Thus, some exhibit homology in their amino acid sequences, suggesting the same (or similar) evolutionary roots. Several families of peptides have been described, including the opiomelanocortin family (endorphin, adrenocorticotropic hormone, melanocyte-stimulating hormone), the somatomammotropin family (growth hormone, prolactin, human placental lactogen), the glycoprotein hormones (thyroid-stimulating hormone, luteinizing hormone, follicle-stimulating hormone, human chorionic gonadotropin), the insulin family (insulin, insulin-like growth factors, somatomedins, relaxin), and the secretin family (secretin, glucagon, glicentin, gastric inhibitory peptide, vasoactive intestinal peptide). Members in a family are related in structure and function. Thus, members of the secretin family stimulate secretory activity in target cells, while those of the insulin family promote cell growth. This preservation of structure and function can be traced to more primitive forms of life. For example, "o~-mating factor," a primitive relative of mammalian gonadotropin-releasing hormone (GnRH) with which it shares 80% sequence homology, functions as a reproductive pheromone in yeast. Moreover, mammalian corticotropin-releasing hormone (CRH) is related to urotensin I of teleosts and to sauvagine of amphibians in structure and in the ability to cause hypotension, vasodilatation, and ACTH release when administered to mammals. Certain strains of bacteria synthesize substances indistinguishable from mammalian hCG and TSH and also synthesize serine proteases that are structurally homologous to the subunits of the glycoprotein hormones. Glycoprotein hormones

702

CHAPTER 30

Endocrine Metabolism I: Introduction

TABLE 30-2

Peptide, Protein, and Glycoprotein Hormones in Humans Hormone

Released into Blood?l

Major Production Site z

Y N N Y Y

Hypoth CNS, Hypoth CNS, Hypoth Blood Circulation Hypoth

9 10 14 21 28 29 31 32 33 39

Y Y Y N Y Y Y Y Y Y

Hypoth Hypoth Hypoth Blood vessels Heart Islets AP, Hypoth Thyroid C-cell Intestinal mucosa AP

41 43 44 51 57 70 84 191 191

Y Y Y Y Y Y Y Y Y

H ypoth Intestinal mucosa H ypoth Islets Ovary, Placenta Cartilage, liver, et al. Parathyroid glands AP Placenta

198 209 215 231 236

Y Y Y Y Y

AP AP AP Placenta AP

AA Residues

Thyrotropin Releasing Hormone (TRH) Methionine Enkephalin (MENK) Leucine Enkephalin (LENK) Angiotensin II (AII) Antidiuretic Hormone (ADH); Arginine Vasopressin (AVP) Oxytocin (OT) Gonadotropin Releasing Hormone (GnRH) Somatostatin (SS)(SRIH) Endothelin- 1 (ET- 1) Atrial Natriuretic Peptide (ANP) Pancreatic Glucagon /3-Endorphin (/3-END) Calcitonin (CT) Cholecystokinin (CCK) Adrenocorticotropic Hormone (ACTH)(Corticotropin) Corticotropin Releasing Hormone (CRH) Gastric Inhibitory Polypeptide (GIP) GH-Releasing Hormone (GRH) Insulin Relaxin Insulin-like Growth Factor I (IGF-I) Parathyroid Hormone (PTH) Growth Hormone (GH)(Somatotropin) Placental Lactogen (hPL) (Chorionic Somatomammotropin) Prolactin (PRL) Thyroid Stimulating Hormone (TSH)(Thyrotropin) Luteinizing Hormone (LH) Chorionic Gonadotropin (hCG) Follicle Stimulating Hormone (FSH)

Other Production Sites 3

Islets (P)

Hypoth (S)

1y = major route of dissemination to target cells is via blood transport; N = functions mainly as paracrine or autocrine hormone, although it may enter the blood stream. 2Hypoth= hypothalamus; CNS = central nervous system; AP = anterior pituitary (Adenohypophysis); Islets = Islets of Langerhans. 3Restricted to those sites that are physiologically important;P = paracrine function; S = released into blood.

therefore may have evolved from ancestral extracellular proteases.

30.3 Steroid Hormones Four kinds of steroid hormones differ in structure and action; they are the androgens (C19), the estrogens (C18), the progestins (C21), and the corticosteroids (C21). All

are synthesized from cholesterol and are produced by mesoderm-derived cells regulated by at least one peptide hormone. Structures of the steroid hormones are shown in Figure 30-1. The hormonal form of vitamin D3 [1,25(OH)2-D3] is a sterol derived from an intermediate of cholesterol biosynthesis and is discussed in Chapter 37. Synthesis begins with cholesterol, which in most instances is acquired from circulating low-density lipoprotein (Chapter 20); however, all steroidogenic cells are

SECTION 30.3

Steroid Hormones

703

$

Cholesterol (exclusive substrate)

B H C--O

Pregnenolone (intermediate)

~H3

?I-I=OH

c=o

c=o HO

Progesterone (major progestin)

(~H2OH

Cortisol (hydrocortisone) (major glucocorticoid)

%0/"~-o

HO

Aldosterone (major mineralocorticoid) OH

Testosterone (major androgen) F I G U R E 30-1 Steroid structure and nomenclature.

Estradiol (major estrogen)

-

-

-

OH

704

Endocrine Metabolism I: Introduction

CHAPTER 30

CH3 j

/CH3

CHcI~~~

\CH3

HC-- ell2-- C1"12--CI'~ CH

H O " ~ CHOLESTEROL

CHOLESTEROL O E S M O t ~ S E (cvP~A) ....... ] . . 36-HYDROXYSTEROID

[17=-HYOROXYt~SE (CYP~r~ !Z,20-LYA~EI

PREGNEI~IOLONE-~ = =. ~

'17..OHPREGNENoLONE = =. ~ - D E HANDROSTERONE YDROEPI"-

....... ! ............................ 21-HYOROXYLASE :i16-HYDROXYLASE ' 18-HYDROXYLASE (CYP11B2) 18-OXlDASE (CYP11B2)

L(CYP~9} I' ' ~ ~

PROGESTERONE

t ............................... 17-OH PROGESTERONE

....... ! ............................

!

.

.

.

.

.

.

! ....

.

11-DEOXYCORTISOL

CORTICOSTERONE

CORl~SOLI(CYPllB1} ............... 21 ~;H2OH

.

TESTO I "'"

H ~

:

.............

ERONE

:i .........

~

HO~~

~

t

---

i

OH

OH

"OH

21 CH=OH

0

I .....

ESTRADIOL

"

I01HC=O H CH~

ESTARONE/176-HYOROXYSTEROIO1

J - ..... DIHYDROTESTOSTERONE

HC= O

ALDOSTERONE

-

ANDROSTENEDIONE ~

DEOXYCORTICOSTERONE

....... t'" 18-OH CORTICOSTERONE ....... t'"

:9

Jl

HO

V

0

F I G U R E 30-2 Biosynthesis of steroid hormones from cholesterol. The different pathways occur to varying extent in adrenals, gonads, and placenta. The systematic names for cytochrome P-450 enzymes, namely CYP followed by a number, are given in parentheses. CYPll B2 and CYP17 possess multiple enzyme activities. [Reproduced with permission from R C. White and R W. Speiser, Congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Endocrine Reviews 21,245 (2000).]

capable of d e n o v o cholesterol synthesis from acetate. Cholesterol either is stored as a fatty acid ester or is immediately utilized in steroidogenesis. The esterified cholesterol is tapped when the cell is stimulated by the activation of cholesterolesterase, a cytoplasmic enzyme that catalyzes the hydrolysis of cholesteryl esters to free cholesterol and fatty acid. The steroidogenic pathway begins in the mitochondria. The unesterified cholesterol is transported from the cytosol to the matrix side of the mitochondrial membrane; this process is mediated by a 37-kDa phosphoprotein known as stetoridogenic acute regulatory (STAR) protein. The importation of cholesterol from cytosol to mitochondria is the rate-limiting step in the steroidogenesis. Trophic stimuli such as ACTH in zona fasciculata and zona reticularis and elevated cytosolic Ca 2+ in zona glomerulosa cause increased synthesis of the StAR protein within minutes. Other proteins in addition to StAR are also involved in cholesterol transfer across the mitochondrial membrane. A number of enzymes in the steroidogenic pathway are cytochrome P-450 proteins that require molecular oxygen and NADPH (Chapter 14). These enzymes

catalyze the clevage of carbon-carbon bonds and hydroxylation reactions. The P-450 enzymes are distinguished by specific notations. For example, C Y P l l A (also known as cholesterol desmolase, side chain clevage enzyme, P450scc) is a mitochiondrial enzyme and catalyzes the conversion of cholesterol (a C27 compound) to pregenolone (a C21steroid). CYP21 refers to the hydroxylase that catalyzes the hydroxyation at carbon 21. The pathways of steroidogenesis are shown in Figure 30-2. A true steroidogenic cell is equipped with the enzyme CYP11A, which catalyzes the initial step of steroidogenesis; the conversion of cholesterol to the steroid intermediate, pregnenolone. CYP1 I A catalyzes the hydroxylation of carbons 20 and 22 of cholesterol, followed by the cleavage of the bond between these two hydroxylated carbons. The resultant steroid, pregnenolone, is a 21-carbon derivative of cholesterol with a two-carbon remnant side chain. It retains the A 5 configuration and the 3/~-hydroxyl group of cholesterol, and is lacking in biological activity; however, it is the universal intermediate in all steroid hormone synthesis. Because mitochondria have no enzyme that recognizes pregnenolone as a

SECTION 30.3

Steroid Hormones

substrate, pregnenolone enters the smooth endoplasmic reticulum (SER) and is processed along whatever steroidogenic pathway is active in the given tissue. Because no single steroidogenic tissue normally expresses all of the steroidogenic enzymes, the presence or absence of enzymes in a given tissue will determine what sort of steroid hormone the tissue can produce from pregnenolone. Pregnenolone serves as a substrate for either of two enzymes; the SER:3/~-hydroxysteroid dehydrogenase/ isomerase (3/3HSD) and 17oe-hydroxylase/17,20-1yase (CYP17). In cells that contain both activities, pregnenolone is first processed by 3/3HSD, followed by CYP17.

T h e / k 4 Pathway: 3/3HSD 3r is a single, nonheme protein that catalyzes a two-step conversion of a aS,3/~-hydroxylated steroid to a A4,3-ketosteroid in the SER of the adrenal cortex, ovary, and testes. When pregnenolone is the substrate, the product is progesterone, a biologically active steroid. In the corpus luteum and the placenta, which are lacking in progesterone-metabolizing enzymes, the nascent progesterone diffuses from the cells and enters the plasma. However, in other tissues progesterone serves as a substrate for at least one other enzyme.

Directional Flow Valve" CYP17 The presence or absence of the enzyme, CYP17, is a major determinant of the fate of pregnenolone (or progesterone). The presence of the enzyme allows access to either the cortisol or the androgenic (sex steroid) pathways but blocks off the aldosterone synthetic pathway. The absence of the enzyme allows for the formation of progesterone and aldosterone but precludes the formation of either cortisol or androgens. (See following subsections on corticosteroid pathway and sex steroid pathway.) CYP17 is a single protein that catalyzes the 17oehydroxylation of either pregnenolone or progesterone in the SER. In some tissues, such as the gonads, this enzyme catalyzes an additional reaction, involving lysis of the bond between carbons 17 and 20 of the 17o~-hydroxylated product. This results in the formation of a 19-carbon androgenic steroid: dehydroepiandrosterone (DHEA) is formed from pregnenolone and A4-androstenedione (A4-A) is formed from progesterone. Thus, the lyase reaction initiates the sex steroid pathway. However, in other tissues that express the CYP17 gene, such as the zona fasciculata of the adrenal cortex, the enzyme does not catalyze the 17, 20-1yase reaction to a significant extent, and the product is a 17oe-hydroxylated pregnenolone or progesterone, which

705

enters the cortisol synthetic pathway. In both tissues, the enzyme is identical, with the same 17oe-hydroxylation activity; and it is not clear what confers the 17,20-1yase activity. In tissues that have CYP17 but are lacking in 3/~HSD, such as the fetal zone and the zona reticularis (Chapter 32), the 17o~-hydroxylation of pregnenolone favors the lyase reaction and the formation of dehydroepiandrosterone (DHEA), probably because no other options are available in the A 5 pathway.

The Corticosteroid Pathway Is Initiated by 2 1 - H y d r o x y l a s e (CYP21) The valve that allows entry of 17-OH progesterone into the corticosteroid pathway is CYP21, a SER enzyme that catalyzes the hydroxylation of carbon 21. In the absence of CYP21, neither aldosterone nor cortisol can be produced. In the presence of this enzyme, two corticosteroid pathways become available, each with a different substrate requirement. Normally, the gene encoding CYP21 is expressed only in the adrenal cortex, primarily in the outer two zones (glomerulosa and fasciculata). The kidney and other tissues are also capable of 21-hydroxylation of steroids; however, these tissues contain an enzyme that is not CYP21 and its activity is too low to interfere with the normal endocrine function of the adrenals. The mineralocorticoidpathway is catalyzed by two enzymes and utilizes progesterone as the initial substrate. The first enzyme, CYP21, catalyzes the 21-hydroxylation of progesterone, which converts it to the mineralocorticoid, 11-deoxycorticosterone (DOC). This enzyme is present in the outer two zones of the adrenal cortex. The second enzyme is zone-specific in that each of the two outer zones of the cortex expresses a different gene for the enzyme. In the zona fasciculata, the CYPllB1 gene expresses a mitochondrial enzyme that catalyzes the 11/~-hydroxylation of DOC to form cortisol; in the zona glomerulosa, another gene, CYPllB2, expresses a mitochondrial enzyme (aldosterone synthase) that catalyzes the three-step conversion of DOC to aldosterone with the formation of the intermediates corticosterone and 18-hydroxycorticosterone. No other tissue in the body normally expresses either of these two genes. Therefore, production of DOC and corticosterone occurs only in the two outer zones of the adrenal cortex, and aldosterone is synthesized exclusively by the zona glomerulosa. The glucocorticoid pathway is confined to the zona fasciculata of the adrenal cortex and it is catalyzed by the same CYP21 enzyme that is involved in the mineralocorticoid pathway in this zone along with CYP11B1. The principal difference between the two pathways in the zona fasciculata is the initial substrate, and this explains

~06

why the intermediates such as DOC and the products (corticosterone and cortisol) are different. Thus, whereas in the mineralocorticoid pathway the initial substrate is progesterone, in the glucocorticoid pathway it is 17o~hydroxyprogesterone. The only difference between the two substrates is the presence of a 17ot-hydroxy group in the latter. Note that this same difference is seen between the two pathways at all three stepsmsubstrate, intermediate, and product. The salient features here are: 1. The presence of 17ot-hydroxy group confers maximal glucocorticoid activity to cortisol (Chapter 32); and 2. The zona fasciculata is the exclusive site of cortisol formation.

The Sex Steroid Pathway Is Initiated by 17~-Hydroxylase/17,20-Lyase ( C Y P I 7) The valve that allows entry of 17-OH prognenolone into the "sex steroid" pathway is the 17,20-1yase component of CYP17, the SER enzyme that catalyzes the 17othydroxylation of steroids. As noted above, the lyase component of the enzyme is active in certain tissues for reasons that are not currently clear. Both A 4 and A 5 steroids are known to feed into the sex steroid pathways although the absence of 3/3HSD precludes the former. It should be emphasized that the formation of androgenic steroids is necessary before estrogen formation is possible; therefore, the initial reaction in the sex steroid pathway invariably produces an androgenic steroid.

Synthesis of Androgens: 17,20-1~yase In the adrenal cortex, the sites of sex steroid production (fetal zone and zona reticularis) are lacking in 3/3HSD, and pregnenolone is processed by both components of CYP 17 to become DHEA, as discussed above. Most of the DHEA is sulfated at the 3-hydroxyl group and the product, DHEAS, is secreted into the plasma. This occurs due to the paucity or absence of any other steroid-modifying enzymes in the tissue. In the Leydig cells of the testes and the theca interna cells of the ovaries, both components of CYP17 are active and will accept either pregnenolone and progesterone as a substrate; radioactive tracer studies indicate that the A 4 substrate (progesterone) is preferred. Progesterone processing by the enzyme results in the formation of androstenedione, which is secreted as such (by the theca interna) or converted to the biologically active androgen, testosterone (T) by a 17/3-hydroxysteroid dehydrogenase (17flSDH)-catalyzed reaction (by the Leydig). Androstenedione and testosterone are 19-carbon steroids are quantitatively the major androgens produced by the

CHAPTER 30

Endocrine Metabolism I: Introduction

gonads; however, the two biologically active androgens are testosterone and dihydrotestosterone (DHT). Androstenedione is inactive because its 17-keto group is not recognized by the androgen receptor. In the peripheral tissues, androstenedione can be converted to T because of the presence of 17/3SDH and many androgen target tissues can convert either androstenedione or T into DHT because of the presence of 5o~-reductase. CYP17 activity (both 17o~hydroxylase and 17,20-1yase activities) in the ovary and testis is inhibited by estrogen.

Synthesis of Estrogens: Aromatase (CYP19) Formation of estrogen is catalyzed by the enzyme aromatase (CYP19) which is present in the SER of a few tissues (gonads, brain, placenta, and adipose). The enzyme is expressed by a single gene ( c y p l 9 ) on chromosome 15; it catalyzes a multistep reaction in which a 19-carbon androgen undergoes hydroxylations at carbon 19 followed by the removal of carbon 19 and the aromatization of ring A. The overall aromatization reactions require an appropriate ring A as substrate (either testosterone or androst-4-ene-3,17dione), 3 NADPH molecules, and 3 oxygen molecules. The regulation of CYP19 activity differs among tissues; FSH induces the enzyme in Sertoli and granulosa cells of the gonads, whereas glucocorticoids induce the enzyme in adipose tissue. The tissues that contain CYP19 are of two varieties; one that produces estrogen de novo from cholesterol (e.g., corpus luteum), and the other that lacks the ability to produce its own 19-carbon substrate and relies on a supply of androgens from other sources (e.g., Sertoli cells of testis, granulosa cells of ovary, placenta, adipose cells, hypothalamus, liver). CYP19 activity is inhibited by the nonaromatizable androgen DHT and is stimulated by the aromatizable androgen testosterone. Steroidogenic enzymes are located mainly, but not exclusively, in the three tissues that are authentically steroidogenic (i.e., capable of converting cholesterol to pregnenolone): the adrenal cortex, the gonads, and, in pregnancy, the placenta. Steroidogenic enzymes are also present in a few tissues that are not capable of de novo steroid synthesis from cholesterol but are able to take up intermediates from plasma and convert them to active hormones; for example, fat cells and the hypothalamus contain CYP19, which enables them to convert circulating androgens into estrogens. The kidney, one of the few extra-adrenal sites with CYP21, converts progesterone to the mineralocorticoid DOC. A "steroid-modifying" enzyme catalyzes a reaction that alters some structural feature of a steroid, resulting in the formation of an active hormone or an inactive metabolite. These enzymes are present in many tissues, although

707

SECTION30.3 Steroid Hormones

in most the result of steroid modification is inactivation; for example, the liver contains a large number of steroidmodifying enzymes, virtually all of which are involved in steroid inactivation. The kidney is another tissue that contains several steroid-modifying enzymes, most of which serve to inactivate steroids. On the other hand, at least three steroid-modifying enzymes are physiologically important, as demonstrated in the disorders resulting from a deficiency in any one of them: 5t~-Reductase, an enzyme that is present in a few tissues, serves to convert testosterone and other androgens to DHT, a reduced form that is most active in those tissues. There are two types of the enzyme (type 1 and type 2), which are products of different genes. The two types of 5ot-reductase do not differ in substrate specificity but differ in enzyme kinetics and tissue distribution. Tissues that express the type 1 enzyme appear to be those contributing to secondary sex characteristics (the hair follicles, sebaceous glands), while tissues that express the type 2 enzyme are structures of the reproductive system (external genital tissues of the fetus, prostate gland, and seminal vesicles). A deficiency of 5o~-reductase type 2 enzyme results in a disorder of sexual differentiation in the male (Chapter 34). The synthesis of 5o~-reductase activity in prostate and skin is regulated by androgens; activity in liver is regulated by thyroid hormone. 17r dehydrogenase (17~HSD) (also called 17-ketosteroid reductase; 17/3-hydroxysteroid ketoreductase) catalyzes the reversible conversion of unconjugated 17-ketosteroids to 17-hydroxysteroids and supplies the physiologic mechanism for generating estradiol from estrone and testosterone from androstenedione, conversions that lead to the formation of biologically active hormones from inactive precursors. This enzyme is present in the testes and ovaries and its activity is critical for the gonads' ability to produce the biologically active form of androgen (testosterone, DHT) and estrogen (estradiol); a deficiency of this enzyme is known to cause disturbances in sexual differentiation. The gene encoding gonadal and placental 17/3HSD is on chromosome 17. The same enzyme is also present in nonreproductive tissues such as liver, lung, erythrocytes, and platelets, but the gene encoding this enzyme is not subject to the same regulation as its gonadal counterpart. Accordingly, in pseudohermaphroditism due to hereditary 17/3HSD deficiency, nongonadal 17/3HSD activity is normal. ll~-Hydroxysteroid dehydrogenase (ll~HSD) catalyzes the conversion of l l-hydroxysteroids into l lketosteroids, a physiologically important reaction that converts the biologically active cortisol to cortisone, an inactive metabolite. The presence of this enzyme in tissues that respond to aldosterone is critical in the hormonal

regulation of blood volume (Chapter 32). It is not clear whether the reverse reaction, one that converts cortisone to cortisol, is catalyzed by a separate enzyme, 11-oxidoreductase. From the clinical standpoint, this is an important reaction because the effectiveness of cortisone treatment relies on it (Chapter 32). Evidence that 11-oxidoreductase and 11/3HSD are different enzymes comes from the observation that patients with a deficiency of 11-oxidoreductase activity may have normal 11/3HSD activity; however, there is evidence that the same enzyme catalyzes both reactions but that different isoforms of the enzyme exist. The activity of 1 l flHSD (cortisone formation) is inhibited by licorice and by carbenoxolone while the activity of 11-oxidoreductase is inhibited by carbenoxolone. A number of drugs have an inhibitory effect on one or more steroidogenic/steroid modifying enzymes, and some of these are used clinically to inhibit the production of certain steroid hormones. Some of the important drugs used for this purpose are broad spectrum while others are hormone-specific; a few are listed below along with their principle therapeutic use: 9 Aminoglutethimide--inhibits CYP11A and CYP19 (breast cancer, Cushing's syndrome) 9 Trilostaneminhibits 3/3HSD (Cushing's syndrome) 9 Ketoconazole--inhibits CYP 17 enzymes (17ol-hydroxylase and Cl7,20-1yase), particularly those involved in testosterone synthesis. After 6 months of treatment in hirsute women, serum testosterone declines without a change in serum cortisol or DHEAS. 9 Mitotane (o,p'-DDD)minhibits adrenal CYP11A and CYP11B 1, but not CYP11B2 (neoplasms of adrenal cortex) 9 Metyrapone--inhibits CYP11B 1 (hypercorticism due to adrenal neoplasms or ectopic ACTH production) 9 Finasteride--inhibits 5o~-reductase type 2 (prostatic hyperplasia) 9 Testolactone~inhibits CYP19 (breast cancer) 9 Fadrozole~inhibits CYP19 (breast cancer) 9 Spironolactone~primarily an aldosterone antagonist but inhibits CYP17 in both ovary and testis. It is also an inhibitor of androgen binding to the AR (K+-sparing diuretic)

Steroid-Binding Serum Proteins Steroid hormones are hydrophobic molecules that convey information to diverse regions of the body by way of the bloodstream. Because of their very limited solubility in water, however, steroid hormones would precipitate

~08

CHAPTER 30

Endocrine Metabolism I: Introduction

TABLE 30-3

Steroid-Binding Serum Proteins CBG 1

Albumin

Site of production Molecular weight Normal concentration

Liver 66,000 550/~M

Liver 58,000 710 nM

Cortisol-binding capacity Testosterone-binding capacity Estradiol-binding capacity Steroid binding sites/molecule Affinity (Kd)

>200 mg/L >400 mg/L >3,000 mg/L >10 -2 • 10--4 M (cortisol) ---4 x 10-4 M (testosterone) Glucocorticoids Thyroid hormone

250 ~g/L

Factors that increase concentration

Factors that decrease concentration

Hepatic disease Renal disease Protein malnutrition

TeBG 2

Liver 94,000 25 nM in men 40 nM in women 6/.~g/L 2/zg/L

1

1

3 x 10-8 M (cortisol)

-2• M (testosterone)

Increased estrogens Increased thyroid hormone

Increased thyroid hormone Increased estrogen (5-10• Decreased androgens Stress Aging High carbohydrate diet Prolonged stress Cirrhosis of liver Increased androgens (2• Obesity Hyperprolactinemia Increased GH Menopause Progestins Glucocorticoids

Increased glucocorticoids Septic shock Pernicious anemia High carbohydrate diet

lCorticosteroid-bindingglobulin (also called transcortin). 2Testosterone-estradiol-binding globulin.

out of blood if it there were no means of rendering them water-soluble. Two processes normally operate to solubilize steroids in blood conjugation and protein binding. Steroids that are conjugated, either with sulfate or glucuronic acid, are unable to enter target tissues because of their water-soluble (hydrophilic) property; they are usually destined for excretion by the kidney. Although a few tissues (e.g., placenta) have sulfatases that can liberate a steroid from its sulfoconjugated state, most tissues are lacking in this ability and are therefore unresponsive to the high levels of conjugated steroids in the blood. The physiologically important means by which steroid hormones are solubilized for transport in blood is the use of certain serum proteins that solubilize steroids. The liver produces three serum proteins that serve to solubilize steroid hormones (Table 30-3): albumin, a nonspecific adsorber

of hydrophobic substances that binds steroids with low affinity; corticosteroid-binding globulin (CBG, also called transcortin), which binds Czl steroids but has the greatest affinity for cortisol; and testosterone-estradiol-binding globulin (TeBG, also called sex hormone-binding globulin, SHBG), which binds steroids of the androstane (C19) and estrane (CIs) series but has greater specificity for androgens than estrogens. Because of the high-affinity binding, steroids are not accessible for tissue uptake while bound to either CBG or TeBG; therefore, the greater the fraction of the steroid that is bound to these proteins, the longer the half-life of the steroid. Unlike that of albumin, the concentrations of CBG and TeBG in plasma are normally, near the concentration of the hormone they preferentially bind, and each protein has only one steroid binding site per molecule. The CBG concentration in plasma is

SECTION 30.4

Mechanism of Hormone Action

~0.71 #M, with a cortisol binding capacity of 250 #g/L (mean cortisol levels (_~ 100 #g/L); while the plasma TeBG concentration is ~ 25 nM in men and -~ 40 nM in women (mean testosterone levels in adult men is ~ 22 nM). An increase in steroid hormone production will result in an increase in the fraction of the hormone bound by the protein until all of the sites on the protein are saturated (i.e., maximal binding capacity); any increase in hormone production beyond this will result in a pathological situation of hormone excess because the cells will be exposed to supraphysiological levels of albumin-bound and unbound hormone. The concentrations of CBG and TeBG in plasma have important influences on both the production and bioeffectiveness of steroid hormones; thus, factors that affect the plasma levels of these binding proteins will indirectly affect the physiology of the steroid hormones they bind. Serum albumin is a quantitatively important binder of hydrophobic molecules that makes possible the transport of these otherwise insoluble molecules in blood; however, it binds very loosely, and this enables the molecules (ligands) to dissociate very rapidly. This means that the albumin-bound fraction serves primarily to solubilize the steroid in plasma and does not oppose tissue uptake of the steroid. For this reason, the albumin-bound fraction should be regarded as being functionally "free." Thus, plasma steroid hormones can be viewed as existing in only two fractions: those that are bound to specific binding protein and those that are not (albumin-bound + unbound). For the sake of simplicity, when dealing with specific steroid hormones, the fractions will be designated relative to the specific binding protein only; for example, when dealing with testosterone, the two plasma fractions will be designated "TeBG-bound" and "non-TeBG-bound." Likewise, plasma cortisol fractions will be referred to as "CBGbound" and "non-CBG-bound." Although the unbound form of steroid hormones is the "active" form, it is also the form that is more susceptible to rapid metabolism. The major site of steroid metabolism is the liver, which inactivates steroids by conjugation and by structural modification. Hepatic A4-hydrogenase reduces the double bond at positions 4 and 5; 17-HSD oxidizes the hydroxyl group at position 17, thereby forming 17ketosteroids; 3o~-hydroxysteroid dehydrogenase reduces the 3-keto to a 3~-hydroxy group. The efficacy of this liver function explains why steroid hormones are ineffective when taken by mouth: they are transported via the hepatic portal system to the liver, where they are promptly metabolized. Minor modifications in the steroid structure, however, can render the hormone more resistant to liver inactivation and, hence, orally active. For example, the introduction of a double bond at position 1 of cortisol yields prednisolone, of a 17ot-ethinyl group in estradiol gives

709

ethinyl estradiol, and of a 17o~-ethinyl group and removal of the 19-carbon in testosterone yields the orally active progestin, norethindrone.

Eicosanoids The eicosanoids are discussed in Chapter 18.

30.4 Mechanism of Hormone Action A target cell is equipped with specific receptors that enable it to recognize and bind a hormone selectively and preferentially. Usually there is one type of receptor for a hormone, but in some cases more than one type of receptor may exist. For example, ol 1-, orz-,/71-, and/Jz-adrenergic receptors are available for catecholamines. The number of receptors for a given hormone in a given cell varies from about 10,000 to more than 100,000. Hormone recognition (and binding) by a receptor initiates a chain of intracellular events that ultimately lead to the effect of the hormone. Binding can be described as a lock-and-key interaction, with the hormone serving as the key and the receptor as the lock. The structural attributes of a hormone allow it to bind to its receptor and to unlock the expression of receptor function. The receptor, which frequently is a hormone-dependent regulatory protein, is functionally coupled to key enzyme systems in the cell, such that hormone binding initiates a receptor-mediated activation of enzymatic reactions, or it is functionally coupled to a region on chromatin, such that hormone binding initiates expression of one or more structural genes. Stated in another way, a hormone-responsive cell is programmed to carry out certain functions when a sensor (receptor) receives the appropriate signal (hormone). In general, the number of receptors for a hormone determines how well the cell responds to that hormone. Several factors influence the number of receptors in a cell. 1. The genotype of the cell determines whether the cell is capable of receptor synthesis and, if so, how much and of what type. (Some endocrinopathies involve receptor deficiency or a defect in receptor function.) 2. The stage of cellular development. 3. Hormones themselves are important regulators of the number of receptors in a cell; this regulation may be homologous or heterologous. Homologous regulation occurs when a hormone affects the number of its own receptors. "Downregulation" (decrease in receptor number) is seen when a target cell is exposed to chronically elevated levels of a hormone. Downregulation involves a decrease in receptor synthesis and

710

is a means by which a cell protects itself against excessive hormonal stimulation. Insulin, the catecholamines, GnRH, endogenous opiates, and epidermal growth factor downregulate their receptors. "Upregulation" (increase in receptor number) also occurs. Prolactin, for example, upregulates its receptors in the mammary gland. Heterologous regulation is more widespread and is a mechanism by which certain hormones influence the actions of other hormones. Some hormones diminish the production of receptors for another hormone thereby exerting an antagonistic effect. Growth hormone, for example, causes reduction in the number of insulin receptors. More prevalent, however, is augmentation of receptor number by a heterologous hormone. Estrogen, for example, increases the number of receptors for the progesterone, oxytocin, and LH, while thyroid hormone increases the number of /~-adrenergic receptors in some tissues. Hormone receptors can be classified into three types on the basis of their locations in the cell and the types of hormone they bind: 1. Nuclear receptors, which bind triiodothyronine (T3) after it enters the cell; 2. Cytosolic receptors, which bind steroid hormones as they diffuse into the cell; and 3. Cell surface receptors, which detect water-soluble hormones that do not enter the cell (peptides, proteins, glycoproteins, catecholamines). The mechanism of action of each of these receptor types is different because each is associated with different postreceptor events in the cell.

30.5 Types of Hormone Receptors Nuclear Receptors Receptors for thyroid hormone (TR), 1,25-dihydroxyvitamin D (VDR), and retinoic acid (RAR) are called nuclear receptors because they are located in the nucleus already bound to DNA (nuclear chromatin) even in the absence of their respective hormone or ligand. These nuclear receptors closely resemble the steroid hormone receptors and belong to the same "superfamily" of DNAbinding proteins. The similarities among the members of this superfamily are striking, particularly in their DNA recognition domains and in the corresponding receptor recognition segment of DNA. The receptor molecule consists of three domains: a carboxy terminal that binds the ligand (hormone/vitamin), a central DNA binding domain (DBD), and an amino terminal that may function as a gene enhancer. The ligand binding domain has high specificity for the ligand, and is the trigger that initiates

CHAPTER 30

Endocrine Metabolism I: Introduction

receptor-mediated regulation of gene expression by the ligand. The receptor is bound to DNA by way of two "zinc fingers" (Chapter 26) in the DBD, which associates with a specific nucleotide sequence in DNA called the "response element" for the ligand. The response element is usually located "upstream" (at the 5' end) of a promoter for the gene that is regulated by the ligand, such that binding of the ligand to the receptor activates the response element (via the DBD), which then activates ("transactivates") transcription of the gene. Transactivation of gene transcription involves the binding of RNA polymerase II to the promoter region of the gene and construction of an RNA transcript (hnRNA) from the DNA gene sequence (Figure 30-3). The hnRNA is then spliced to yield mature mRNA (messenger RNA), which translocates to the cytoplasmic compartment and becomes associated with ribosomes. Ribosomal translation of the mRNA results in the synthesis of a nascent polypeptide, the primary sequence of which is encoded in the gene that was activated by the ligand.

Thyroid Hormone Receptors Thyroid hormone receptors (TRs) are nonhistone proteins that function as transducers of the effects of thyroid hormone on gene expression. There are four isoforms of TR (TRot 1, TRot2, TR/31, TR/32) that are products of two different genes, c-erbAot and c-erbAfl. There is some tissue specificity in the distribution of these isoforms. Three of these isoforms (TRot 1, TRot2, TRill) are found in almost all cells (although in different relative amounts); however, the TR/32 isoform is found only in the brain. The TRot2 is unique because it does not bind thyroid hormone but it does bind to DNA. Its function is unclear. The other three TR isoforms are single proteins with three regions (domains): a carboxy terminal region that binds T3, a central region that binds a specific region of DNA, and an amino terminal region that may function as a gene enhancer. The TR is synthesized on ribosomes and is actively transported through the nuclear membrane pores into the nucleus, where the DBD of the TR binds to a specific segment of DNA called the "thyroid hormone response element" (TRE). The TR binds to a TRE half-site as a monomer, a homodimer (ot/ot, Ot/fl,/3//3, etc.) or a heterodimer, in which a TR isoform dimerizes with another protein, e.g., the retinoid X receptor (RXR). In the absence of thyroid hormone, the TR association with the TRE does not result in any changes in gene expression. Thus, TRs exist in the nucleus, are bound to DNA, and await the arrival of thyroid hormone. Thyroid hormones T4 and T3 arrive at their target cells by transport in plasma, for the most part bound to plasma proteins (Chapter 33). The very small fraction of T4 and

SECTION 30.5

Types of Hormone Receptors

711

Ligand

S

RNA Transcription

DNA

\

v Hormone Response Element

/

Promoter Region

F I G U R E 30-3 Thyroid hormone, 1,25-dihydroxyvitamin D, and retinoic acid receptor regulation of transcription. The hormone receptor (HR) is dimerized at site (3) and is bound to DNA at hormone response element site (2). Without the ligand, transcription is inactive due to the interaction of HR with corepressor at site 4. When the ligand (hormone) binds to HR, the bound corepressor dissociates leading to an interaction between the coactivator and HR. These regulatory changes result in increased transcription.

T3 that is unbound at any given instant is accessible for cellular "uptake," i.e., the diffusional passage of the hydrophobic hormone through the lipid matrix of the cell membrane and into the cell. Once in the cell, the hormone associates loosely with cytoplasmic proteins that prevent its escape back into plasma. Microsomal 5'-deiodinase, which is present in thyroid target cells, catalyzes the removal of the 5'-iodine from T4, converting it to T3. This creates a concentration gradient that favors the movement of hormone into the nuclear compartment, in which TR (monomer or dimers) exists in association with the TRE sequence of DNA. Monomeric TR appears to bind to a TRE half-site, while dimeric (homodimer or heterodimer) TR binds to two half-sites. When activated by T3 binding to TR, the TRE promotes transcription of the gene into hnRNA. After removal of the introns from hnRNA, the resultant mRNA is translated into a protein at the ribosomes. The protein, which may be regulatory or structural, is the final molecular expression of thyroid hormone action. The outcome of T3 binding to TR is not a stimulation of transcription in all cases. In the pituitary thyrotrophs, T3 inhibits transcription of both the TSH-ot and TSH-/3 genes by binding to TR-TRE complexes. The TR mediating this inhibitory effect appears to be a monomer that binds to a TRE half-site, in contrast to the usual dimeric TR that binds to two TRE half-sites and mediates the stimulatory effects of T3 on transcription in other cell types. In some instances, the actual cell response to thyroid hormone is a manifestation of the activity of the induced

protein; therefore, the induced protein would first need to be activated (e.g., by phosphorylation) before the thyroid hormone effect on the cell can be seen. Steroid H o r m o n e Receptors

Steroid hormone receptors belong to a large family of DNA-binding proteins that include receptors for thyroid hormone, retinoic acid, and 1,25(OH)2 vitamin D. There are specific receptors for glucocorticoids (GRs), mineralocorticoids (MRs), estrogens (ERs), androgens (ARs), and progestogens (PRs), all of which are coded for by different genes. Unlike the TR, there appears to be only one functional receptor protein for a given steroid that is encoded on a single gene. Like the TR, the steroid receptor is a single protein with three regions (domains): a carboxy terminal region that binds the steroid specifically, a central DBD, and an amino terminal region that may function as a gene enhancer. The DBD of the steroid receptor projects two zinc fingers that allows both recognition of and binding to the hormone response element (HRE) of DNA. A generic depiction of the mechanism for steroid hormone activity at a target cell is shown in Figure 30-4. The first step consists of dissociation of the hormone from the plasma transport protein and entry into the cell by diffusion across the plasma membrane. In the second step the hormone binds with the receptors in the cytoplasm and nucleus. Receptors for glucocorticoid and aldosterone are found in the cytoplasm and receptors for estrogen and progesterone are found in the nucleus. Recall that receptors for

712

CHAPTER30

Plasma transport ~ = protein with bound steroid hormone Heat shock protein

STEP 1 ,~ . ~ =

-.

Free steroid hormone

~ ,, ' ~ "

~ J

'1- ~

J'~

STEP2 ~ " " O

..

7

~STEP3

Non-DNA / binding cytoplasmic receptor

+ ,....,fSTEP 4 +

Target cell

.........

+!~-

~'i" 4 ~ ~ ++.+:'"Activa~on ~!'.i \t;~'; / ::..'-:.~

Nucleus

~~~receptor

~

Endocrine Metabolism I: Introduction

!1

-:7+

TEP4

+?'i:k~ ';'.i':+:!

v t '~!I)~

r

~l ~ ' STEP 6

_

++

+++++l] .":i!!

on"++

~,._J . Trenslocetion ~...';i..... ~ .:~'~ "Activated" ?fece~p?gl%Smipiex~':~:.:.+:.i . I ~ ~ + /

/ \ response /

~ STEP 7 i

mRNA

";~"

mRNA

Protein synthesis ,

.

.

~

j

,

FIGURE 30-4 A genericdepictionof the mechanismof steroidhormoneaction(seetextfor details). [Reprintedwithpermissionfrom A. W.Normanand G. Liwack,Hormones, 2nded. San Diego;AcademicPress, 1997,p. 40.] thyroid hormone, retinoic acid, and 1,25(OH)2 vitamin D are all found in the nucleus. Activation of the receptor occurs in steps 3 and 4. The receptors exist in cells complexed with a dimer of heatshock protein (HSP, 90-kDa), which conserves the functional status of the receptor molecule in the absence of the hormone. In addition, HSPs may function to prevent the unoccupied receptor from binding to DNA by masking the DNA binding domain. HSPs bound to the hormone binding domain of the receptor are displaced by the hormone for which the receptor is specific. Once formed, steroid-receptor complexes aggregate into homodimers, and in this form the receptor is said to be "activated." The activated cytoplasmic receptors are translocated via a nucleopore to the nucleus (step 5). In step 6, the activated receptor triggers (or inhibits) transcription of the hormone-regulated gene. In the event of mRNA production, the newly synthesized mRNA is translocated to the cytoplasm where it is translated (step 7). The biological effect of the hormone reflects the function of the protein (e.g., enzyme, transporter, hormone, receptor), and the duration of the effect is a function of the half-life of the mRNA (minutes to hours) and of the protein (hours to days).

The steroid receptor homodimer may also regulate gene expression by interacting with transcription factors that are associated with either the HRE or the promoter. By binding to certain transcription factors, the activated hormone receptor may 1. Stimulate gene expression by removing an inhibitory transcription factor; 2. Stimulate gene expression by facilitating the activation of the promoter by a transciption factor; or 3. Inhibit gene expression by removing a stimulatory transcription factor. One important transcription factor with which steroid receptors interact is AP-1 (accessory/auxiliary/adapter protein-l), which promotes the expression of genes involved in cell proliferation. The growth-suppressing effect of glucocorticoids is probably mediated by the binding of the hormone-GR complex to one component of AP-1 (c-jun) which, in effect, removes the AP-1-mediated promotion of cell proliferation. Another transcriptional factor involved in steroid hormone action is the cAMP response element binding protein (CREB), which mediates the genomic effects of peptide hormones that utilize the cAMP second messenger system (see below). A

SECTION 30.5

Types of Hormone Receptors

number of steroid-regulated genes are subject to modulation by CREB and are thus subject to the regulatory influence of both steroid and peptide hormones.

Cell Surface Receptors Many hormones, such as the hormonal amines and all peptidic hormones, are unable to penetrate the lipid matrix of the cell membrane, and thus depend on the presence of receptor sites at the surface of target cells. As listed in Table 30-4, there are several types of cell membrane receptors for these hormones, each of which is coupled to a distinct set ofintracellular postreceptor pathways. The surface receptors all initiate postreceptor events that involve the phosphorylation of one or more intracellular proteins, some of which are enzymes whose activities depend on the state of phosphorylation. In two of these cases, an intracellular second messenger is utilized to implement the hormonal action and involves G-protein-coupled receptors. One is coupled to the adenylate cyclase-cAMP system and the other is associated with the phosphatidylinositol-Ca 2+ pathway (IP3 pathway).

G-Protein-Coupled Adenylate Cyclase-cAMP System The effect on adenylate cyclase of ligand binding to specific receptors mediated by heterotrimeric G-proteins is shown in Figure 30-5. The G-proteins belong to a family of regulatory proteins each of which regulates a distinct set of signaling pathways. The cell membrane receptors coupled to intracellular G-proteins share a common seven transmembrane-spanning o~-helical serpentine structure; however, they do not show a common overall sequence homology. The oe-helical segments are linked with alternating intracellular and extracellular peptide loops with the N-terminal region located on the extracellular side and C-terminal region located on the intracellular side (Figure 30-6). Receptors coupled to G-proteins have been divided into three subfamilies and significant sequence homologies do exist within the families, the receptors belonging to the three families are described in Figure 30-6. Extracellular signals detected by the membrane receptors are diverse and include hormones, growth factors, neurotransmitters, odorants, and light. Examples of G-proteins are Gs, Gi, products of ras oncogenes (Chapter 26), and transducin. Transducin is a constituent of the light-activated cGMP-phosphodiesterase system in the retina (Chapter 38). This system is similar to the adenylate cyclase system except that light is the ligand, rhodopsin is the membrane receptor, cGMP-phosphodiesterase is the effector, and the G-protein is the transducer. The two types

713

of receptors, Rs and Ri, can be functionally linked to adenylate cyclase (Figure 30-5). Binding of an appropriate ligand to Rs stimulates the adenylate cyclase whereas binding to Ri inhibits the enzyme. The G-proteins are heterotrimeric consisting of an o~ subunit and a tightly coupled fl ~, subunit. The ot subunit is the unique protein in the trimer. It possesses sites for interaction with cell membrane receptors, fl V subunit, intrinsic GTPase activity, and ADP ribosylation. In the unstimulated state, the G-protein is present in the heterotrimeric (otfi V) form with GDP tightly bound to the ot subunit. Upon activation of the receptor by the bound ligand, GDP is exchanged for GTP on the ot subunit followed by dissociation of Gs~Tp from the f i t subunit. The el subunits Gs and Gi are designated G~ and Gia. The G-protein subunits undergo posttranslational covalent additions of lipids such as palmitoyl, farnesyl, and geranyl groups. These hydrophobic groups linked to G-protein subunits are necessary for proper anchoring in the cell membrane. Figure 30-7 shows the cyclic functioning of the G~ protein. Binding of a hormone to R~ permits binding of GTP to Gs, to form a Gsc~.GTPcomplex and release of the G~ and G• subunits. The Gso~.GTPcomplex activates adenylate cyclase which remains active as long as GTP is bound to Gs~. Binding of GTP to Gs~ also activates a GTPase intrinsic to Gs~ that slowly hydrolyzes the bound GTP (GTP turnover is one per minute), eventually allowing reassociation and formation of the trimer and inactivation of adenylate cyclase. A similar series of events is initiated by binding of a ligand to Ri. In this instance, dissociation of the Gi trimer by formation of Gi~.GTP inhibits adenylate cyclase and may allow the/3,g subunit to bind to Gs~, preventing Gs~ from activating adenylate cyclase. GTP hydrolysis by GTP-activated Gi~-GTPase allows re-formation of the Gi trimer. GDP is displaced from Gi~ by binding of another molecule of GTP with reactivation of Gic~. The inhibitory effect occurs because the target cell contains much more Gi than G~. Gic~ may also interact with membrane calcium transport and the phosphatidylinositol pathway. The function of/3 g dimers is not completely understood but they may participate in the regulation of downstream effectors in the signaling pathway. The G-protein-coupled receptor signaling pathway is regulated by several different mechanisms. Negative regulation of the signaling pathway can occur at both the receptor and G-protein levels. Receptor phosphorylation by G-protein-coupled receptor kinase and arrestin binding can lead to inability of the ligand to activate the signaling system (e.g.,/3-adrenergic receptor-G-protein transducing system). Negative regulation also occurs after the activation of G-protein, by promoting the GTPase activity of G~. This is achieved by regulator of G-protein signaling

,m

e~

,m

""

~

~9

His or Arg 2~ --+ Cy in G~ of affected endocrine glands. Some of the clinical manifestations of McCune-Albright syndrome include hyperthyroidism, polyostotic fibrous dysplasia, and hyperpigmented irregularly shaped skin lesions (known as cafd-au-lait lesions). The cutaneous hyperpigmentation

~1~

is caused by hyperfunctioning of MSH receptors coupled to Gs~ in the melanocytes. An example of decreased function due to impaired activation or loss of Gs~ and resistance to hormone action is pseudohypoparathyroidism (Albright's hereditary osteodystrophy). Patients with this syndrome exhibit generalized resistance to the action of those hormones dependent on Gs~ for their function despite elevated serum hormone levels. Several dysmorphic features are also characteristic of these patients. A well-understood disorder of hypofunctioning Gs~ is pseudohypoparathyroidism type la. This disorder exhibits a dominant inheritance pattern with one normal and one abnormal Gs~ allele. In different families affected by pseudohypoparathyroidism Ia, two missense mutations have been identified in G~: Arg 385 --+ His and Arg 231 ~ His. The former mutation prevents receptormediated Gs stimulation and the latter mutation prevents receptor-mediated binding of GTP to Gs. A paradoxical finding consisting of both gain and loss of function due to identical mutations in Gs~ (Ala 366 ~ Ser), in two unrelated boys with pseudohypoparathyroidism Ia. Both of these patients, while exhibiting hormone resistance in some tissues, showed autonomous production of testosterone by testicular Leydig cells due to gain of function in the receptors of luteinizing hormone. This results in precocious puberty (testotoxicosis). This particular mutation in the G~ gene involves a gain in function property in G~ enhancing the rate of release of GDE However, the paradoxical effect is due to temperature stability. The mutant protein is unstable at 37~ which causes pseudohypoparathyroidism; however, the mutant protein is stable at lower temperature and thus is active in testes, which are about 3~176 cooler than the rest of the body. G-protein abnormalities also may arise from mutations in genes encoding proteins that regulate G-protein signaling. Several RGSs have been identified. RGS proteins bind to G~ and accelerate the hydrolysis of GTE The physiological responses of G-proteins regulated by RGSs are fast and include vagal slowing of the heart rate (Gi), retinal detection of photones (Gt), and contraction of vascular smooth muscle (GQ).

G-Protein-Coupled PhosphatidylinositolCa z+ Pathway The calcium system is more complex than the cAMP system and contains a variety of mechanisms for transducing the Ca 2+ signal into changes in cellular function. It is also sensitive and responds to relatively small, transient changes in free Ca 2+ concentration. This sensitivity is desirable from a regulatory point of view, but it also

CHAPTER30 Endocrine Metabolism I: Introduction

reflects the toxicity of even low concentrations of free Ca 2+ (Chapter 37). In a resting cell, the pool of cytosolic calcium is very small, and most of the intracellular Ca 2+ is bound in mitochondria or the endoplasmic reticulum or is bound to the plasma membrane. The cytosolic calcium ion concentration is -~0.1 /~mol/L, in contrast to the high concentration (~ 1000 #mol/L) outside cells. This 10,000-fold gradient is maintained by the plasma membrane, which is relatively impermeable to calcium; by an ATP-dependent Ca 2+ pump (which extrudes Ca 2+ in exchange for H +) in the plasma membrane; and by "pump-leak" systems in the endoplasmic reticulum and inner mitochondrial membranes. A change in the permeability of any of these membranes alters the calcium flux across the membrane and causes a change in cytosolic calcium concentration. This change is thought to act as the messenger in the calcium system. Plasma membranes regulate cytosolic calcium concentration by their role as a diffusion barrier and by containing the ATP-dependent Ca 2+ pump and receptors for extracellular messengers. Activation of some of these receptors increases the Ca 2+ permeability of the plasma membrane and thus the level of cytosolic calcium. In other cases, the activated receptor produces one or more second messengers that increase cytosolic Ca 2+ concentration from the calcium pool in the endoplasmic reticulum. Calcium then becomes a "third messenger." The endoplasmic reticulum is the source of Ca 2+ in the initial phase of cell activation by some hormones in many systems. Since the plasma membrane can bind Ca 2+, the rise in cytosolic calcium following binding of some extracellular messengers may be due to release from this intracellular calcium pool. The inner mitochondrial membrane may function primarily as a calcium sink, taking up excess calcium in the cytosol that results from hormonal activation of the cell. At cytosolic Ca 2+ concentrations greater than 0.6 #mol/L, the mitochondrial calcium pump is activated and stores calcium in the mitochondrial matrix as a nonionic, rapidly exchangeable, phosphate salt. At low cytosolic calcium concentrations, the inner mitochondrial membrane allows Ca 2+ to "leak" into the cytosol. The capacity of the active influx pathway (the pump) is much greater than that of the passive efflux route (the leak). The mitochondrial pumpleak system may serve to fine-tune the cytosolic calcium concentration while the plasma membrane is the principal safeguard against entry of toxic amounts of calcium into the cell.

Mechanism of the Calcium Messenger System Most extracellular messengers that cause a rise in cytosolic Ca 2+ concentration also increase the turnover

SECTION 30.5

Types of Hormone Receptors

719

Receptor . Hormones

~ Cell membrane

DAG

PIP2

Go-Protein

DAG

PS

II

.V.oop os,a ~"-GTP

p

p

Protein Kinase C /P IP3

~ p

p ~

Calmodulin (CoM) Protein Kinase ~

:~[/') }

.)

CaM/Ca2+ ~

,./.,//ReceptOrsmoothforIP3

/////

endoplasmic recticulum(SER)

FI G U R E 30-8 " 2+ pathway. The binding of a hormone at a specific receptor site results in GQ protein-coupled phosphatidylinosito 1-tJa the activation of G-protein which, in turn, activates phospholipase C via GQ~.OTp-protein. The action of phospholipase C on phosphatidylinositol 4,5-bisphosphate (PIP2) yields inositol trisphosphate (IP3) and diacylglycerol (DAG) which, along with phosphatidylserine (PS), activates protein kinase C. IP3 binds to receptors on SER, releasing Ca 2+ which, in turn, activates another set of protein kinases. +, Activation.

of phosphoinositides in the plasma membrane. The pathway is initiated by the binding of extracellular messengers to specific receptors located on the plasma membrane and activating G-proteins (e.g., GO). The activated GQ~.CTp-protein, in turn, activates phospholipase C, which catalyzes hydrolysis of phosphatidylinositide 4,5-bisphosphate, initiating the intracellular secondmessenger effects (Figure 30-8). The action of phospholipase C results in the formation of two products, inositol 1,4,5,-trisphosphate (IP3) and diacylglycerol (DAG), which function as intracellular messengers (Figure 30-9). Most of the DAG released in response to stimulation of calcium-mobilizing receptors contains arachidonic acid esterified to C2 of the glycerol. This arachidonate may

O"

I

O ~ p _ O"

oII

I

H2 - O - C - R1

R2-

O II C - O - CH

I

CH 2 - O

i

O H

o

P- O - Cl

I O"

I"

H~?

H

HJ ( ~ / -

C

C

OH

I

I

OH~c~HI

\

~.?C

/

H

o

II

0"

.-P-

I

0"

O"

F I G U R E 30-9 Structure of phosphatidylinositol 4,5-bisphosphate. Hydrolysis at the dotted line is catalyzed by phospholipase C and releases diacylglycerol and inositol 1,4,5-trisphosphate, which are the intracellular second messengers.

be released from DAG by diacylglycerol lipase or from phosphatidic acid by phospholipase A2, following phosphorylation of DAG. It can then serve as a substrate for the synthesis of eicosanoids (prostaglandins, prostacyclins, thromboxanes, and leukotrienes; Chapter 18), which also modulate a number of physiological functions. These reactions are part of lipid and phosphoinositide cycles (Figure 30-10). Many extracellular messengers in a variety of cells activate this "leak" pathway. They include acetylcholine (many tissues); epinephrine, angiotensin, and vasopressin (liver); angiotensin (adrenal cortex); glucose (pancreatic islets); photons (photoreceptor cells); and several different substances in neutrophils, leukocytes, and platelets. Many of the membrane receptors linked to phosphoinositide metabolism also activate guanylate cyclase, but the mechanism and function of this activation remain unknown. The starting point of these reactions is phosphatidylinositol 4,5-bisphosphate (PIP2; Figure 30-10), which is synthesized from phosphatidylinositol (PI) in two steps catalyzed by specific kinases. The phosphates are removed by specific phosphomonoesterases setting up futile cycles. Stimulation of phospholipase C by binding of an agonist to its receptor hydrolyzes PIP2, releasing IP3 and DAG (usually 1-stearoyl-2-arachidonyl-sn-glycerol). IP3 stimulates the release of Ca 2+, predominantly from the endoplasmic reticulum, and may increase plasma membrane permeability to Ca 2+ in some cells. In most cells studied, the rise in level of cytosolic Ca 2+ is preceded by an increase in that

720

CHAPTER30 Endocrine Metabolism I: Introduction

99 L,p,d c y c l e f . . . . ~

CDP-DG ~

P~ CTP ~-(~)~~.

Futile cycles i

(

\

myo-lno

(~ADP

ATP ATP L ADP L 6 DP Agonist

P'

-,o P~[

itol

..-.

I\ATP I.~---'~--~ MG

H,O

,

P~

H=O

PMG t (C~ I~.Arachidonic acid

]X,R

~

P~

H=O

Inositolphosphate cycle

F I G U R E 30-10 Inositol metabolism. An inositol phosphate cycle converts inositol 1,4,5-trisphosphate (IP3) to free inositol, which can then combine with CDP-diacylglycerol (CDP-DAG) to re-form phosphatidylinositol (PI). Inositol 1,4-bisphosphate (IP2) and inositol l-phosphate (IP) are intermediates in this cycle. The CDP-DAG produced in the lipid cycle returns diacylglycerol for resynthesis of PI. It is uncertain whether arachidonic acid is released by hydrolysis of diacylglycerol, with release of a monoglyceride (MG), or of phosphatidic acid (PA), with release of a phosphomonoglyceride (PMG). The futile cycling of PI, phosphatidylinositol 4-phosphate (PIP), and phosphatidylinositol 4,5-bisphosphate (PIP2) maintains a supply of PIP2 to generate diacylglycerol and IP3 in response to agonist binding. The enzymes involved are l, phosphatidylinositol kinase; 2, PIP kinase; 3, PIP2 phosphomonoesterase; 4, PIP phosphomonoesterase; 5, PIP2 phosphodiesterase (phospholipase C); 6, IP3 phosphatase; 7, IP2 phosphatase; 8, IP phosphatase; 9, CDP-diacylglycerol inositol phosphatidate transferase (phosphatidylinositol synthase); 10, diacylglycerol kinase; l I, CTP-phosphatidate cytidyl transferase; 12, diacylglycerol lipase; and 13, phospholipase A2. [Modified and redrawn with permission from M. J. Berridge and R. E Irvine, lnositol triphosphate, a novel second messenger in cellular signal transduction. Nature 312, 315 (1984). @ 1984 by Macmillan Magazines Ltd.]

of IP3, making IP3 the second messenger and calcium the third messenger. In the endoplasmic reticulum, IP3 seems to increase efflux of Ca 2+. Amplification occurs since an estimated 10-20 calcium ions are released from liver endoplasmic reticulum by each IP3 molecule. IP3 is rapidly hydrolyzed to free inositol, which is used for resynthesis of PI (Chapter 19). Removal of the 1-phosphate is blocked by lithium, with accumulation of inositol 1-phosphate and depletion of m y o - i n o s i t o l pools. The effect is most marked in the brain, where lithium also inhibits de n o v o synthesisof m y o - i n o s i t o l and plasma inositol is unable to cross the blood-brain barrier. Lithium carbonate, used to treat bipolar affective disorders, may have its therapeutic effect by inhibition of the phosphatidylinositol system. In several types of cells, stimulation of plasma membrane receptors coupled to phosphoinositide turnover not only triggers a rapid mobilization of intracellular Ca 2+ as discussed above, but also a prolonged mobilization.

The latter is dependent extracellular Ca 2+, and another metabolite of inositol, namely, inositol tetrakisphosphate (IP4),or Ins(1,3,4,5)P4, has been implicated in the transport of extracellular Ca 2+. The molecular mechanism of this process is nuclear. IP4, unlike IP3, cannot mobilize Ca 2+ from endoplasmic reticulum. IP4 metabolism consists of its synthesis from IP3 by a novel, specific ATPdependent kinase and its conversion to inactive metabolite Ins(1,3,4)P3 by a phosphatase. The regulation of kinase and phosphatase activity may be of importance in controlling the intracellular events. Diacylglycerol released by phospholipase C may increase protein phosphorylation. Protein kinase C (C-kinase) is a monomeric enzyme (M.W. 82,000) found free in the cytosol in most tissues except brain, kidney, and liver, where it is predominantly membrane-bound. Diacylglycerol increases both the binding of protein kinase C to the inner surface of the plasma membrane and the sensitivity of the kinase to activation by Ca 2+ and phosphatidylserine. The effects of DAG related to protein phosphorylation are thought to be mediated by this kinase. Tumor-promoting phorbol esters such as 12-O-tetradecanoylphorbol- 13-acetate (ingredient in croton oil that promotes skin tumor production by carcinogens) can directly activate protein kinase C, mimicking DAG and bypassing the receptors. When activated by phorbol esters, protein kinase C phosphorylates cytosolic myosin light-chain kinase (Chapter 21) and other proteins, particularly those related to secretion and proliferation. When activated by DAG, the kinase phosphorylates a number of proteins in vivo, but its physiological substrates are unknown. DAG is removed by phosphorylation to phosphatidic acid or by hydrolysis to monoacylglycerol and free fatty acid. In addition to the effects of DAG and phorbol esters on protein kinase C, some evidence links malignant transformation by some oncogene products to phosphoinositide metabolism. Platelet-derived growth factor, the product of the sis oncogene, stimulates inositol lipid metabolism. Products of the src and ras oncogenes may be phosphoinositide kinases, which increase the concentration of PIP3 and provide more substrate for phospholipase C. The product of the ras gene is a GTP-binding protein (G-protein) involved in receptor activation of PIP2 hydrolysis. A point mutation activates the ras oncogene, and the resulting oncogene product binds GTP but lacks GTPase activity. This abnormal protein could activate PIP2 hydrolysis independently of receptor occupancy. Following these early events initiated by binding of a calcium-mediated hormone to its receptor, a transient rise in cytosolic calcium concentration and a more prolonged increase in DAG concentration occur. There is also a prolonged four- to fivefold increase in Ca 2+ flux across the

SECTION 30.5

Typesof Hormone Receptors

plasma membrane, which may maintain an elevated Ca 2+ concentration at the cytoplasmic face of the membrane. This increase, together with the diacylglycerol, may continue to activate the membrane-bound protein kinase C. In this way, myosin light chains may remain phosphorylated and active during smooth muscle contraction (Chapter 21) despite a fall in myoplasmic Ca 2+ to near-basal levels. The rise in intracellular Ca 2+ level also alters the activity of metabolic pathways by calcium-binding proteins. Of these proteins, calmodulin (M.W. 17,000), found in the cytoplasm of every eukaryotic cell, has received the most attention. Parvalbumin in skeletal muscle, troponin C in skeletal and cardiac muscle, and myosin light chain in smooth muscle serve similar functions. The affinity of these proteins for Ca 2+ is high, as reflected by dissociation constants of 10-6-10 -8 M. Occupation of at least three of four equivalent Ca 2+ binding sites on calmodulin induces a conformational change and, in most cases, increases its affinity for proteins having a calmodulin binding site. A major function of CaZ+-calmodulin is regulation of the phosphorylation states of intracellular proteins. Systems affected include those involved in secretion (of insulin, for example), neurotransmitter release and neuronal function, cellular contractile processes (Chapter 21), and several protein kinases involved in regulation of glycogen and glucose metabolism. A phosphatase activated by Ca 2+ and calmodulin, known as calcineurin, functions as a negative modulator of some of the effects of Ca 2+ related to protein phosphorylation. Calcineurin is a CaZ+-calmodulin-dependent serine/threonine protein phosphotase and it participates in many CaZ+-dependent signal transduction pathways. The immunosuppressant drugs cyclosporin A and FK506, after binding to their respective cytoplasmic binding proteins (cyclophilin and FK506-binding protien), inhibit the action of calcineurin. This blocks the translocation of a factor into the nucleus of activated T lymphocytes and prevents allograft rejection by blocking T cell cytokine gene transcription (Chapter 35).

Receptors for Insulin and Growth Factors Insulin exerts its effects by altering the state of phosphorylation of certain intracellular enzymes by a mechanism that does not involve cAMP but that requires specific binding to surface receptors with tyrosine kinase activity. Insulin exerts acute (minutes), delayed-onset (hours), and longterm (days) effects entirely by way of a single receptor. The insulin receptor is a transmembrane heterotetramer consisting of one pair each of two dissimilar subunits linked by disulfide bridges (Chapter 22). Both subunits

721

are expressed entirely by a single gene, which is located on chromosome 19. The two subunits, ot and/~, are formed posttranslationally from a single proreceptor following glycosylation and proteolytic cleavage by an endopeptidase. The o~ subunit, which projects into the extracellular space, is a glycosylated 135-KDa protein that specifies the insulin binding site (one binding site per o~ subunit). The transmembrane/~ subunit is a glycosylated 95-KDa subunit that contains four domains, one of which has tyrosine kinase activity. The tyrosine kinase activity of the subunit is suppressed by the o~ subunit in the absence of insulin; insulin binding to the ot subunit removes the inhibition and allows the/~ subunit tyrosine kinase to phosphorylate itself (autophosphorylation of the fl subunit). Autophosphorylation is followed by phosphorylation of one or more key intracellular proteins, one of which is insulin receptor substrate-1 (IRS-1) that initiates a cascade of intracellular phosphorylations mediated by a number of intracellular enzymes that are incompletely characterized, but which are critical in bringing about all of the known cellular effects of insulin (Chapter 22). Autophosphorylation of the insulin receptor may also be required for inactivation of the insulin signal ("OFF" signal). Within minutes after autophosphorylation, the insulin-activated receptor complex undergoes lateral movement toward clathrin-coated pits, the site at which a number of activated insulin receptors congregate or "cluster." This clustering stimulates endocytosis of the congregation, a process referred to as "internalization" or "endocytosis," which is followed by fusion of the internalized insulin receptor membrane vesicle (endosome) with a lysosome, i.e., an endolysosome formation. This allows breakdown of both insulin and its receptor by lysosomal proteases. It has been suggested that insulin fragments resulting from this digestion process are mediators of some of the actions of insulin within the cell, but further evidence in support of this is needed. In states of chronically elevated blood levels of insulin, there is a substantive decrease in the density of insulin receptors in insulin-dependent cells due to a decrease in the synthesis of insulin receptors. This phenomenon, which is referred to as "downregulation," represents a means by which a cell autoregulates its receptivity to the hormone, and is also detected by other hormones such as the catecholamines, endorphins, and GnRH. The precise mechanisms involved in downregulation are not known. The insulin receptor is expressed in most cells and tissues examined to date, including the non-insulindependent tissues such as brain, kidney, and blood cells (both red and white). Although no specific cellular response to insulin has been described in these tissues, it is conceivable that insulin regulates one or more intracellular

722

CHAPTER 30

Endocrine Metabolism I: Introduction

F I G U R E 30-11 Mechanism of prolactin receptor activation. Activation of prolactin receptor consists of ligand-induced sequential receptor homodimerization driven by the two binding sites of prolactin. In the intracellular domain of the homodimer of the ligand-receptor complex, a tyrosine kinase [known as Janus kinase 2 (Jak-2)] is activated. Jak-2 kinase causes autophosphorylation and phosphorylation of the receptor. [Reproduced with permission from M. E. Freeman, B. Kanyicska, A. Lerant, and G. Nagy, Physiological Reviews 80, 1530 (2000).]

activities that are unrelated to glucose uptake and utilization. The IGF-I receptor is a n otzfl 2 heterotetramer that is structurally similar to the insulin receptor but is coded for by a different, single-copy gene located within bands q25-26 of chromosome 15. Like the insulin receptor, with which it shares 50-60% sequence homology, the IGF-I receptor has a cysteine-rich domain in the c~ subunit for ligand binding and a tyrosine kinase domain in the /3 subunit that catalyzes transphosphorylation of contralateral fl-subunit sites. The receptor has highest affinity for IGF-I, a lesser affinity for IGF-II (about 2- to 15-fold less), and low affinity for insulin (about ~ 100- to 1000-fold less) due to the absence of insulin-binding determinants in the ot subunit.

Receptors for Growth Hormone and Prolactin Growth hormone (GH) and prolactin (PRL) belong to the "helix bundle peptide" (HBP) hormone family, which includes human placental lactogen (hPL), erythropoietin (EPO), and many interleukins and cytokines. All members of this family have similar mechanisms of action; they utilize membrane receptors that are straight-chain glycoproteins with extracellular, transmembrane and intracellular domains. The membrane receptors for this group of hormones when bound with their respective receptors undergo dimerization. Each dimerized receptor is bound to one molecule of hormone. The intracellular events of the ligand-mediated activation of the receptor consists of activation of a constitutively associated tyrosine kinase known

as Janus kinase 2 (Jak-2). Jak-2 kinase transphosphorylates itself and phosphorylates the intracellular domains of the receptor. Figure 30-11 illustrates the mechanism of prolactin receptor activation. The signal transduction pathways for GH and PRL are discussed in Chapter 31.

30.6 Organization of the Endocrine System The nervous and endocrine systems function in a coordinated manner to promote growth, homeostasis, and reproductive competence. The need to rapidly adjust physiological processes in response to impending disturbances in homeostasis is met by the nervous system, while the endocrine system effects the more prolonged, fine-tuned adjustments. Together, they effect appropriate organismic adaptation. Some interdependence also exists between the two systems. The nervous system, for example, relies heavily on a continuous supply of glucose, the circulating levels of which are under multiple hormonal control, while maturation of the nervous system is regulated by thyroid hormone, and maintenance of mental acuity in adults depends on the availability of both thyroid hormone and glucocorticoids. On the other hand, hormone production is generally dependent on the nervous system, albeit to varying extents. The synthesis of insulin, glucagon, calcitonin, parathyroid hormone, and aldosterone appears to require little or no neural regulation, whereas that of the hypothalamic peptides and adrenal medullary hormones is totally dependent on it. It may be significant that those hormones which are relatively independent of neural control serve

SECTIO30.6 N Organizationof the EndocrineSystem

723

Nervoussyste m_.~ .... Level I (neuroendocrine)

IHypothalamus[ I Adrena' I\~ ' l " ~ " [medlulla ] ~ ~ CRH ADH. TRH Oxytoc,nEpin!phrine ~ SS/GRH Norepinephrine \

)

LevelII

] Antedor~,~pGHLPituitary ] ACTH TSH

LevelIII

ic-c ,,I Gastro-

~/

' int~rStic~al ~ ' '$ ~'X~ alcitOnin Many \

[ ,,,Paintresati ~i cI' l Paratiyr~ i Insulin

LH

Glucagon

Parathyroid

hormone

]Adrenal'cortexj I mhyroidi [Gonads[

L

Cortisol

1

T, ,Ts

t

Progestins Androgens Estrogens

F I G U R E 30-12 Organization of the endocrine system. The endocrine system can be classified into three levels. At level I are those endocrine tissues that are, or were embryologically derived from, nervous tissue; these include the hypothalamus, the adrenal medulla, the thyroid C-cell, and several of the gastrointestinal mucosal cells. Hormones produced by these tissues resemble neurotransmitters to the extent that their effects are rapid in onset and short term. At level II are those endocrine tissues that are directly or indirectly influenced by the nervous system; these include the anterior pituitary, the pancreatic islets, and the parathyroids. Hormones produced by these tissues, which are polypeptides, proteins, or glycoproteins, exert both short- and long-term effects. At level III are the adrenal cortex, the thyroid, and gonads; these endocrine tissues exhibit strong dependence on the anterior pituitary. Hormones produced by these tissues are small, hydrophobic molecules that ultimately affect gene expression in the target cells; their effects are thus slow in onset but long term. Although the level III tissues are not directly regulated by the central nervous system (CNS), they are, by virtue of their control by the anterior pituitary, indirectly regulated by the hypothalamus. Neural regulation by the CNS is critical; - - , neural regulation by the CNS exists but is not critical; . . . . , little or no regulation by CNS; - - - > , hormonal regulation.

to regulate the level of substances that profoundly affect nerve function (glucose, calcium, sodium, potassium). The organization of the endocrine system can best be described in relation to the central nervous system. Three levels of endocrine tissues can be distinguished on the basis of their association with the central nervous system (Figure 30-12). The first level consists of those that are (or were) derived from nerve cells; these include the hypothalamus, adrenal medulla, thyroid C-cell, and gastrointestinal enterochromaffin cells. The hypothalamus and adrenal medulla still retain their neural connections and can therefore be regarded as endocrine extensions of the nervous system. The C-cell and the gut cells, however, are APUD cells and lack neural connections. These four tissues produce hormonal peptides or amines having, like neurotransmitters, rapid-onset, short-term effects.

The second level of endocrine tissues are the anterior pituitary, pancreatic islets, and parathyroids, which show varying degrees of dependence on neural regulation. Because it is highly dependent on the hypothalamus for control of hormone synthesis and release, the anterior pituitary is highly dependent on the nervous system. On the other hand, the pancreatic islets and parathyroids synthesize and release their hormones in the absence of neural signals; however, the rate of hormone release can be influenced by autonomic nerve stimulation. These three tissues produce polypeptide, protein, or glycoprotein hormones having both short- and long-term effects, often involving the regulation of cellular protein synthesis. The third level of endocrine tissues includes the adrenal cortex, thyroid follicles, and gonads, all of which

724

depend heavily on trophic stimulation by anterior pituitary hormones; therefore, although these peripheral endocrine tissues are anatomically and functionally separated from the central nervous system, they are indirectly dependent on the hypothalamus and, thus, on the nervous system. They produce low-molecular-weight hormones (steroids, iodinated amines) having long-term effects that involve regulation of gene expression. The hierarchical organization of the endocrine system emphasizes the substantial influence of the nervous system on endocrine function; however, the influence of the central nervous system is not always obligatory. Thus, although C-cells and gut cells are of neuroblastic origin, they function well in the absence of neural input; similarly, the neural influence on hormone release from pancreatic islets and parathyroid glands is relatively unimportant. Nevertheless, the hypothalamus and adrenal medulla fail when their neural connections are severed, leading to atrophy of the anterior pituitary, adrenal cortex, thyroid, and gonads. However, two hormones are produced by these tissues in normal or elevated amounts following neural disconnection: prolactin from the anterior pituitary, and aldosterone from the adrenal cortex. Release of aldosterone is regulated by the renin-angiotensin system and by extracellular potassium levels, while that of prolactin normally is under tonic inhibition by the hypothalamus (Chapter 31 ). A unidirectional scheme of endocrine control has been alluded to above in the discussion of three levels of endocrine tissues: the nervous system-hypothalamus controls the anterior pituitary, which in turn controls the peripheral endocrine tissues. Closer examination of how hormonal secretions are regulated reveals that, in many instances, a closed-loop feedback (servo-mechanism) circuit operates to ensure that the correct amount of hormone is released at any given time. In its simplest form, a circuit involves an endocrine cell that can monitor changes in the blood level of the substance it regulates. This sensing ability is coupled to a discriminator with an imprinted "set point" such that if the circulating level deviates significantly from the set point, the hormonal output is appropriately modified to counteract that deviation. This type of simple negative feedback regulation of hormone release-usually involving one endocrine tissue, one hormone, and one substance that is monitored--is the primary means by which secretion of insulin, glucagon, parathyroid hormone, and (to some extent) aldosterone is regulated (Figure 30-13). Another way in which hormonal secretion is regulated involves a neuroendocrine reflex, which differs from a neural reflex in that the efferent neuron is replaced by a

CHAPTER30

[.,~L.'.dl TOO

Endocrine Metabolism I: Introduction ,

i

I substance I

j

Too

,

lEndocrlnecel:

["]

Glucose

,.=,ofl substance I

Hormone

l Uver

I Muscle, others

Too h~p

1

Insulin

6-C911

I

o,K- ] ~ Adrenalcortex | zon8 glomerulosaX

i

1

i oon.,1

Aldosterone~

F I G U R E 30-13 Simple negative feedback control of hormone release. (Top) A discriminator associated with an endocrine cell is capable of monitoring blood levels of a regulated substance. If the level of the substance deviates from the "set point," the endocrine cell adjusts its hormonal output to restore that level. The hormone may act either on the source of the substance or on its removal. (Middle) If the blood level of glucose rises above the set point imprinted in the # cell of the pancreatic islet, insulin secretion is increased. Insulin acts on the liver (source of glucose) to inhibit ( G ) further production of glucose, thereby diminishing the input of glucose into the blood glucose pool (broken line). Insulin also acts on several tissues (muscle, others) to promote ( ~ ) glucose utilization, thereby facilitating the removal of glucose from the blood glucose pool (solid line). (Bottom) Plasma K + concentration is monitored by the zona glomerulosa of the adrenal cortex, and an elevation will stimulate the release of aldosterone. Aldosterone acts only on the disposal of K +, thereby serving to control the level of K + in blood. ECF, Extracellular fluid.

neuroendocrine cell (Table 30-5). As in a neural reflex arc, a sensory (afferent) neuron conveys, via synaptic connections, signals emitted from a sensory receptor to an efferent (neuroendocrine) cell, feedback which then affects the function of an effector organ or tissue. Release of oxytocin, ADH (vasopressin), and adrenal medullary hormones is regulated in this way (Table 30-5). No direct negative feedback comparable to that in the previous model operates; however, the efferent signal (hormone) counteracts, alleviates, or compensates for the stimulus that initiated the reflex. Several examples follow. By increasing water reabsorption, ADH promotes osmodilution and volume expansion, which counteract the dehydration that triggered distress signals from osmoreceptors and baroreceptors; by causing milk ejection, oxytocin helps appease the suckling infant. The adrenal medullary catecholamines produce

SECTION 30.6

Organizationof the Endocrine System

725

TABLE 30-5 Neuroendocrine Reflex

Sensory receptor

Sensoryneuron

Sensory Receptor

Sensory Neuron

Cardiovascular baroreceptors

Cranial nerves IX and X

Osmoreceptors

? Osmoreceptor neuron Ascending touch pathways Unclear

Touch receptors in nipple Uterine stretch receptors Various receptors of noxious stimuli

Many

o~

Neuroendocrinecell HormoneEffectorcell / ~ ~

Neuroendocrine Cell

Hormone

Effector; Effects

Magnocellular neurons, hypothalamic PVN*, SON* (As above)

ADH

Kidney; water reabsorption

(As above)

(As above)

(As above)

Oxytocin

(As above)

(As above)

Adrenal medulla

Epinephrine, norepinephrine

Mammary gland; milk ejection Uterus; smooth muscle contraction Cardiovascular augmentation and liberation of energy substrates

*PVN = paraventricular nucleus. *SON = supraoptic nucleus.

cardiovascular and metabolic changes that enable the individual to cope with the stressful situation that triggered their release, while the adrenal medullary enkephalins probably serve to suppress the sensation of traumatic pain. The neuroendocrine reflex is usually polysynaptic, i.e., one or more interneurons connect the sensory neuron to the endocrine cell. Thus, the system is more amenable to modulation by other neurons or hormones, serving to fine-tune the system. For example, under conditions of normal extracellular fluid volume and osmolality (i.e., when ADH release is suppressed), the sensation of pain stimulates the release of ADH and promotes water retention. Pain-induced release of ADH may be due to neural discharge of endorphins or enkephalins in the central nervous system, which act on the hypothalamus. The peripheral hormones (level III, Figure 30-12) also influence the neuroendocrine reflex. Cortisol, for example, amplifies the adrenal medullary response to stress by increasing the excitability of interneurons in the reticular formation and by stimulating the synthesis of adrenal medullary phenylethanolamine N-methyltransferase (PNMT), which catalyzes the formation of epinephrine (Chapter 32). Estrogen is an important regulator of hypothalamic pro-oxyphysin synthesis

(Chapter 31) and thus promotes the formation of oxytocin. Some hormones are under complex feedback regulation involving both a neuroendocrine reflex and a negative feedback circuit. Invariably, the neuroendocrine reflex component involves the hypothalamus, which in one instance is also the site of positive feedback (Chapter 34). The "substance" that is monitored and exerts negative feedback may be a substrate (e.g., glucose, amino acid) or the hormone that is being regulated. Generally, two or more hormones are involved, which is not surprising, since most hypothalamic and anterior pituitary hormones subserve the function of regulating the release of another hormone (Figure 30-14). As examples, release of growth hormone is regulated by growth hormonereleasing hormone and somatostatin; that of prolactin by prolactin-releasing factor and prolactin-inhibiting factor (dopamine); that of thyroid hormone by thyrotropinreleasing hormone and thyroid-stimulating hormone; that of cortisol by corticotropin-releasing hormone and corticotropin; and that of the sex steroids by gonadotropinreleasing hormone, luteinizing hormone, and folliclestimulating hormone.

726

CHAPTER30 Endocrine Metabolism I: Introduction

F I ( ; U R E 30-14 Complex feedback regulation of hormone secretion. Hormones of level III endocrine tissues are regulated by a complex system having both a neuroendocrine reflex component and a simple negative feedback component. Invariably, the former involves the extrahypothalamic central nervous system (sensory neurons), the hypothalamus (endocrine cell 1), and the anterior pituitary (endocrine cell 2), while the latter involves the anterior pituitary (endocrine cell 2) and a level III endocrine tissue (endocrine cell 3). Hormone 3 exerts negative feedback on endocrine cell 2 and hence on its own secretion. Hormone 3 may also feed back to endocrine cell I and to higher centers, although such cases of feedback may be positive ones.

Supplemental Readings and References M. K. Bagchi, M. J. Tsai, B. W. O'Malley, and S. Y. Tsai: Analysis of the mechanism of steroid hormone receptor-dependent gene activation in cell-free systems. Endocrine Reviews 13, 525 (1992). D. M. Berman and A. G. Gilman: Mammalian RGS proteins: Barbarians at the gate. Journal of Biological Chemistry 273, 1269 (1998). D. L. Bodenner and R. W. Lash: Thyroid disease mediated by molecular defects in cell surface and nuclear receptors. American Journal of Medicine 105, 524 (1998). G. A. Brent: The molecular basis of thyroid hormone action. New England Journal of Medicine 331, 847 (1994). D. L. Cadena and G. N. Gill: Receptor Tyrosine kinases. FASEB Journal 6, 2332 (1992). M. A. Carson-Jurica, W. T. Schrader, and B. W. O'Malley: Steroid receptor family: structure and functions. Endocrine Reviews 11,201 (1990) L. De Vries, B. Zheng, T. Fischer, et al.: The regulator of G protein signaling family. Annual Review of pharmacology and Toxicology 40, 235 (2000).

Z. Farfel, H. R. Bourne, and T. Iiri: The expanding spectrum of G protein diseases. New England Journal of Medicine 340, 1012 (1999). M. E. Freeman, B. Kanyicska, A. Lerant, et al.: Prolactin: structure, function and regulation of secretion. Physiological Reviews 80, 1523 (2000). U. Gether: Uncovering molecular mechanisms involved in activation of G protein-coupled receptors. Endocrine Reviews 21, 90 (2000). C. K. Glass: Differential Recognition of target genes by nuclear receptor monomers, dimers and heterodimers. Endocrine Reviews 15, 391 (1994). M. A. Lazar: Thyroid hormone receptors: Multiple forms, multiple possibilities. Endocrine Reviews 14, 184 (1993). W. L. Miller: Molecular biology of steroid hormone synthesis. Endocrine Reviews 9, 295 (1988). M. Pfahl: Nuclear receptors/AP- 1 interactions. Endocrine Reviews 14, 651 (1993). J. W. Putney, Jr. and G. S. Bird: The inositol phosphate-calcium signalling system in nonexcitable cells. Endocrine Reviews 14, 610 (1993). M. D. Ringel, W. F. Schwindinger, and M. A. Levine: Clinical implications of genetic defects in G proteins: The molecular basis of McCune-Albright

Supplemental Readingsand References Syndrome and Albright Hereditary Osteodystrophy. Medicine 75, 171 (1996). W. Rosner: The functions of corticosteroid-binding globulin and sex hormone-binding globulin: Recent advances. Endocrine Reviews 11, 80 (1990). E Rusnak and P. Mertz: Calcineurin: form and function. Physiological Reviews 80, 1483 (2000).

727 E. R. Simpson, M. S. Mahendroo, G. D. Means, and others: Aromatase cytochrome P450, the enzyme responsible for estrogen biosynthesis. Endocrine Reviews 15, 342 (1994). M. Truss and M. Beato: Steroid hormone receptors: Interaction with DNA and transcription factors. Endocrine Reviews 14, 459 (1993). E C. White and E W. Speiser: Congenital adrenal hyperplasia due to 21hydroxylase defeciency. Endocrine Reviews 21, 245 (2000).

CHAPTER

31

Endocrine Metabolism lI: Hypothalamus and Pituitary

31.1 Hypothalamus The hypothalamus is a small region of the brain in the ventral aspect of the diencephalon. In the adult human, it is about 2.5 cm in length and weighs about 4 g. Ventromedially, it surrounds the third ventricle and is continuous with the infundibular stalk of the pituitary (hypophysis). This cone-shaped region of the hypothalamus, the median eminence, consists mainly of axonal fibers from hypothalamic neurons, which either terminate in the median eminence or continue down into the posterior lobe of the pituitary, and it is perfused by a capillary network (primary plexus) derived from the carotid arteries. Blood from the primary plexus is transported by portal vessels (hypophyseal portal vessels) to another capillary network (secondary plexus) in the anterior lobe of the pituitary (adenohypophysis) (Figure 31-1). The hypothalamus contains a high density of nerve cell bodies clustered into nuclei or areas. Neurons in each of these nuclei tend to send their axons to the same regions in the form of tracts. These nuclei innervate the median eminence, other hypothalamic nuclei, the posterior pituitary, and various structures in the extrahypothalamic central nervous system. All of the hypothalamic neurons are presumably monoaminergic (i.e., they A list of expanded acronyms appears in Appendix VIII.

synthesize and release the neurotransmitter amines norepinephrine, serotonin, or dopamine); however, many are also peptidergic (i.e., they synthesize and release neuropeptides). At least 12 hypothalamic neuropeptides have been identified, and it is e:~pected that many more will be discovered. The hypothalamus is an important integrating area in the brain. It receives afferent signals from virtually all parts of the central nervous system (CNS) and sends efferent fibers to the median eminence, the posterior pituitary, and certain areas of the central nervous system. In the median eminence, a number of peptidergic fibers terminate in close proximity to the primary plexus, which transports their neuropeptide secretions via the portal blood flow to the anterior pituitary. Since these neuropeptides affect the function of the anterior pituitary cells, they are called "hypophysiotropic." Two hypothalamic nuclei, the paraventricular nucleus (PVN) and the supraoptic nucleus (SON), consist of two populations of neurons that differ in size: the parvicellular (small-celled) neurons and the magnocellular (large-celled) neurons. The parvicellular neurons of the paraventricular nucleus produce a peptide (CRH; see below) that is transported by axoplasmic flow to the median eminence and then released into the hypophyseal portal blood. The magnocellular neurons of the paraventricular and supraoptic nuclei send their long axons directly into the posterior pituitary (neurohypophysis),

729

730

CHAPTER31 Endocrine Metabolism I1: Hypothalamus and Pituitary

F I G U R E 31-1 Hypothalamic-pituitary regulatory system. Hypothalamic releasing hormones and release-inhibiting hormones are synthesized in various neurons. These hypothalamic hormones are transported to the anterior pituitary via a portal venous system and the anterior pituitary target cells either release or inhibit the release of specific hormones into the general circulation. The posterior pituitary hormones are synthesized and packaged in cell bodies in the hypothalamus and then are transported to nerve endings in the posterior pituitary. Afterward they are released following appropriate stimuli. The explanation for the abbreviations appears in the text. [Slightly modified and reproduced with permission from A. W. Norman and G. Litwack, Hormones, 2nd ed. New York: Academic Press, 1997, p. 89.1

where their dilated nerve endings are closely positioned next to capillaries that drain the tissue. Peptides synthesized in these magnocellular neurons are transported to the neurohypophysis by axoplasmic flow, then released into the general circulation. These neuropeptides are called "neurohypophyseaL" Other hypothalamic neurons make synaptic contact with other neurons, and their neuropeptide products function as neurotransmitters or as neuromodulators. They are called "neuroregulatory."

Hypophysiotropic Peptides (Table 31-1) Thyrotropin-releasing hormone (TRH) is a tripeptide amide with the following structure:

o O H

o

C--N--C--C--N H I CH 2

C--O

I

NH2

Pyroglutamic acid

Histidine

Proline amide

All of its components are essential for activity. TRH is resistant to intestinal peptidase action and is active when taken orally. TRH is produced by the parvicellular neurons of the PVN and the periventricular nucleus, and is

e~

r~

.,e -~

0

c.~

0

Cq

~

Cq

~o~

9,.~

,~ o~

~

rO

9

I.~

0

~

rO

o~

~

Z

>

~

..,= . 0 ,= ,~

o~

0

~

-,~

Z

O0

~;:>

oo

;:>

~

~ -

m

0

~ ~o ~

~

c~

O0

~ ~

c,.; +

~

~

0

o

;:> ~

~

..~. ~ ~,=,~

rO

.,..~

~~~.~ ~o

Z

.~. ~

9

..~

o

It

z ;>

.,-.q

.,.,~

o

o

o~

ZN

II

2~

~.~ g

it N

0

Z'~

o,..~

~32

released at the median eminence. TRH principally stimulates the synthesis and release of thyroid-stimulating hormone (TSH, thyrotropin) in the anterior pituitary, and also stimulates the release of prolactin (PRL). Both effects are mediated by membrane receptors coupled to the GQ~-phospholipase C-/~-calcium-protein kinase C second-messenger system (Chapter 30). Gonadotropin-releasing hormone (GnRH) or luteinizing hormone-releasing hormone (LHRH) is a decapeptide which, like TRH, has a pyroglutamic acid residue in its N terminus. The presence of histidine and tryptophan at positions 2 and 3, respectively, is required for biological activity, while the replacement of glycine at position 6 by D-Trp increases activity up to 100-fold. The potent synthetic GnRH agonist shown below lacks the glycinamide terminal and has instead an ethylamide residue and D-Trp at position 6:

CHAPTER31

EndocrineMetabolism I1: Hypothalamus and Pituitary

anterior pituitary. CRH stimulates the release of ACTH and/~-endorphin by the anterior pituitary corticotrophs by stimulating the synthesis and posttranslational processing of their prohormone, pro-opiomelanocortin (POMC). Growth hormone-releasing hormone (GHRH) is a 44amino-acid polypeptide produced in the tuberoinfundibular dopaminergic system (TIDA) of the hypothalamus, which includes the arcuate and ventromedial nuclei. GHRH stimulates the synthesis and release of GH in anterior pituitary somatotrophs by a cAMP-mediated mechanism. The efficacy of GHRH in promoting GH release is strongly modulated by hypothalamic somatostatin.

Somatostatin(growth hormone release-inhibiting hormone) is a tetradecapeptide with an intrachain disulfide bridge. Somatostatin neurons are located in the parvicellular neurons of the PVN, the periventricular nuclei

O I! pGlu l~His2~Trp3~Ser4~Tyr5~D-Trp6~Leu7~Arg8~ Pr09~C~ NH~CH2~CH3 GnRH-producing neurons are located in the preoptic area and in the organum vasculosum of the lamina terminalis but are known to have migrated to these areas from the olfactory placode during fetal development. Failure of these neurons to migrate into the hypothalamus during embryogenesis results in infertility secondary to GnRH deficiency (Kallmann's syndrome). GnRH stimulates the release of luteinizing hormone (LH) and follicle-stimulating hormone (FSH), the two gonadotropic hormones produced by the pituitary. GnRH also occurs in other regions of the brain and in the placenta. Its pivotal role in the regulation of gonadotropin release, and thus in reproductive competence, has led to the formulation of GnRH antagonists for contraceptive purposes. Paradoxically, however, synthetic GnRH agonists, when administered chronically in humans, cause a downregulation of GnRH receptors that results in suppression of gonadal steroid production. GnRH action on pituitary gonadotropes begins by occupancy of GnRH receptors by GnRH and involves intracellular mobilization of calcium ions through the action of G-protein, phospholipase C, and phosphatidylinositol pathways (Chapter 30). This suppression of gonadal function has been referred to as "medical castration." Corticotropin-releasing hormone (CRH) is a 41amino-acid polypeptide produced in the parvicellular neurons of the paraventricular nucleus and released into the hypophyseal portal system in the median eminence. Antidiuretic hormone (ADH, AVP) is also synthesized and released by the same neurons, but in humans, unlike the situation in rats, the amount of ADH released at the median eminence is too small to influence the function of the

and the preoptic nuclei. Somatostatin inhibits the synthesis and release of GH from the somatotropes by neutralizing the effect of GHRH, an effect mediated by Giot inhibition of adenylyl cyclase. Within the hypothalamus, somatostatin inhibits the release of GHRH by the same mechanism and therefore exerts an inhibitory effect at two levels of GH control. Somatostatin is also produced outside of the CNS, particularly in the D cells of the islets of Langerhans and in the gastrointestinal tract. Somatostatin produced at these extra hypothalamic sites does not normally influence GH secretion by the anterior pituitary because it is greatly diluted by systemic blood. Prolactin-inhibiting hormone (PIH), also known as dopamine, is not a peptide (Chapter 17). It functions as a neurotransmitter in the CNS and as a precursor of norepinephrine and epinephrine in the adrenal medulla. In the hypothalamus, it originates in the TIDA and is released at the median eminence. Dopamine is a potent inhibitor of PRL release by the lactotropes (and mammosomatotropes) of the anterior pituitary, and this effect is mediated by D2 receptors that are coupled to Gia inhibition of adenylate cyclase. The lactotropes are unique in that they do not require stimulation by the hypothalamus to secrete PRL; in fact, blockage of the blood flow from the hypothalamus to the anterior pituitary results in elevated serum levels of PRL, due to withdrawal of dopamine. Thus, unlike somatostatin, the effectiveness of dopamine does not depend on the presence of a stimulating hormone (Chapter 34). The existence prolactin-releasing factor (PRF), a yetto-be-characterized stimulator of PRL release, remains controversial, particularly in view of the fact that TRH subserves this role. What is clear, however, is that the

733

SECTION31.1 Hypothalamus

TABLE 31-2

Neurohypophyseal Hormones Structure

Hormone S-S

Antidiuretic hormone (ADH) (arginine vasopressin)

I ]6 -Pro 7-Arg 8-Gly 9- N H 2 Cysl-TyrZ-Phe3-Gln4-AsnS-Cys

Oxytocin

C~sl -TyrZ-Ile3-Gln4-AsnS-Cys S-S ]6 -Pro 7-Leu 8-Gly 9- N H 2

principal effect of the hypothalamus on PRL secretion is inhibitory. A stimulating hormone from the hypothalamus may be important in tempering the inhibition by dopamine.

Neurohypophyseal Peptides The magnocellular neurons of the paraventricular and supraoptic nuclei synthesize antidiuretic hormone (ADH, vasopressin, or arginine vasopressin) and oxytocin, neuropeptides that are released into the general circulation in significant amounts (Table 31-2). These neuropeptides are synthesized in separate cells and transported by axoplasmic flow to nerve endings in the neurohypophysis. Cholinergic signals to the magnocellular neurons stimulate the release of ADH and oxytocin via nerve impulse propagation along the axon and CaZ+-dependent exocytosis of secretory granules. Release mechanisms for ADH and oxytocin are under separate control. ADH is a nonapeptide that contains an intrachain disulfide bridge. ADH is synthesized as part of a precursor glycoprotein (propressophysin), whose gene is located on chromosome 20. Propressophysin is packaged into secretory granules and transported to the neurohypophysis. During transit, propressophysin is split by proteases in the granule wall, yielding ADH and a "carrier" polypeptide called neurophysin II or "nicotine-sensitive neurophysin" (NSN), whose release is stimulated by nicotine. Upon neural stimulation, ADH and NSN are released in equimolar amounts. NSN has no known biological activity. In the collecting ducts of the kidney, ADH acts by a Gs-protein coupled cAMP-mediated mechanism to increase the permeability of ductal cells to water by mobilizing aquaporin proteins to the apical membrane which prevents diuresis (see also Chapter 39). At relatively high concentrations, ADH is a potent vasoconstrictor. Such concentrations are attained after massive hemorrhage, when constriction of blood vessels prevents further blood loss. Major stimuli for ADH release are an increase in plasma osmolality, monitored by hypothalamic osmoreceptors, and a decrease in blood volume, monitored by baroreceptors in the carotid sinus and in the left atrial wall. Afferent

fibers to the vasomotor center in the hindbrain sense a fall in blood volume, and diminished noradrenergic signals from this center to the hypothalamic magnocellular neurons stimulate ADH release. Because norepinephrine inhibits ADH release, decrease of this inhibition constitutes a stimulation. Oxytocin is a nonapeptide that differs from ADH at the residues in positions 3 and 8. It is synthesized in separate magnocellular neurons from a gene that codes for prooxyphysin, the precursor of oxytocin and its "carrier" (neurophysin I or estrogen-sensitive neurophysin). Neurons in the paraventricular and supraoptic nuclei synthesize prooxyphysin, which is packaged and processed during transit to the neurohypophysis. The principal action of oxytocin is ejection of milk from the lactating mammary gland ("milk let-down"), and it also participates in parturition (Chapter 34). Oxytocin is released by a neuroendocrine reflex mechanism and stimulates contraction of estrogenconditioned smooth muscle cells. The mechanism of action of oxytocin does not involve cAMP but may involve regulation of increased intracellular Ca 2+. The oxytocin receptor belongs to a seven-membrane-spanning receptor family.

Neuroregulatory Peptides Neuronal projections from the hypothalamus to other regions of the brain relay important output information that influence blood pressure, appetite, thirst, circadian rhythm, behavior, nociception (pain perception), and others factors. Although many of these neurons release neurotransmitter amines at synapses, some of them are known to release neurotransmitter peptides. These include, among others, peptides that closely resemble hormones formed in the gastrointestinal system as well as the endogenous opiates (Table 31-3).

Brain-Gut Peptides Gut hormone-like neurotransmitters have been detected in the brain and are believed to function in a manner that

b

om

olll~

Z

"~

~ ~

~--~

~

~

9

=

o~~

.(...a

~=~

~

r.~

~

0~

=

r..~

o~

r..~

r~

o~

r.~

~

~

~

~

~
InSl000

Ins inhib GH inhib

IGF-I inhib Starvation inhib

differences of IGFs with insulin and among themselves are summarized in Table 31-5. Six serum proteins produced mainly in the liver that bind IGFs in the circulation have been identified; these are designated IGF-binding proteins (IGFBPs) 1 through 6. IGFBP- 1 appears to retard target tissue uptake of IGF-I, while IGFBP-3 appears to enhance it. The latter accounts for most of the IGF found in blood. About 80-90% of the IGF-I in circulation is bound to IGFBP-3, along with an 88-kDa acid-labile subunit (ALS); this complex confers protection on the IGF and prolongs its half-life to 12-15 hours. About 5% of the IGF is unbound, and the remaining 5-15% is bound to IGFBP-I, 2, or 4 as smaller complexes. Unlike insulin and other peptide hormones that are released from storage granules, IGFs are released as they are produced. Most tissues produce IGFs in small amounts that are sufficient for local (paracrine/autocrine) effects and do not contribute significantly to the circulating pool of IGFs. The highest concentration of IGF-I and II is found in circulation and is primarily derived from the liver. GH and insulin are positive regulators of IGF-I synthesis in the nonfasting state. During mild starvation when GH levels increase, there is a decline in hepatic production

IGF-I I

+

13 mg/day 700 ng/ml Nonpulsatile Steady, nonvariable 6 (types 1-6) 15h Muscles, connective tissue, cartilage, bone IGF-I receptor; plasma membrane IGF-II/Man-6-P receptor (monomeric) IGF-II >> I500 IGF-I inhib (IGF-IR); Starvation inhib (IGF-IR); Insulin, IGF-I stim (IGF-II R)

of IGF-I due, in part, to the decline in insulin. Refeeding, which causes insulin to rise and GH to fall, promotes IGF-I production only if the diet is adequate in caloric and protein content. Note that circulating IGFI does not mediate the growth-promoting effect of GH but is an important feedback regulator of GH. The indispensability of IGF-I for linear growth and the fact that it mediates this effect of GH are firmly established. IGF-I, locally produced in endochondral bone in response to GH, promotes skeletal growth by stimulating clonal chondrocyte expansion of the distal proliferative zone of the epiphyseal plate. Under physiological conditions, IGF-I is a relatively minor regulator of fuel homeostasis. It does not mediate the effects of GH on intermediary metabolism; in fact, many of the effects of IGF-I resemble those of insulin, not GH. IGF-I promotes production and actions of erythropoietin and is responsible for the increased packed volume of blood that results from elevated GH levels. Disturbances in G H and I G F

Only rarely has a condition of IGF deficiency or excess been described that is not accompanied by a disturbance in

SECTION31.2 Pituitary 61and (Hypophysis)

741

FIGURE 31-6 Structure of human prolactin. [Reproduced with permission from R. K. Murray, D. K. Granner, P. A. Mayes, and V. W. Rodwell, Harper's Biochemistry, 21st ed. Norwalk: Appleton & Lange, 1988. @1988 Appleton & Lange.]

GH production or responsiveness. Thus, a GH deficiency results in IGF-I deficiency, whereas a GH excess results in IGF-I excess. GH deficiency in children results in reduced growth velocity and growth retardation (dwarfism) due to secondary IGF-I deficiency, whereas in adults there are no dramatic signs or symptoms. Adults who are deficient primarily in GH have decreased lean body mass, increased adiposity, and are at increased risk of cardiovascular disease. GH hypersecretion in children causes increased growth rate and can result in gigantism. However, GH excess occurs infrequently in childhood, and most frequently in middle-aged adulthood, which leads to acromegaly (acral -- extremities + megas -- large), a condition in which the cartilaginous tissues proliferate, resulting in distorted overgrowth of the hands, feet, mandibles, nose, brow, and cheek bones. Because epiphyseal cartilage is absent in long bones, there is no gain in height. Acromegaly promotes insulin resistance and cardiovascular complications. Resistance to the effects of GH results in a condition similar to GH deficiency. The major cause of GH resistance is a genetic defect in the growth hormone receptor (GHR) and the resultant condition is known as Laron-type dwarfism. This is an autosomal recessive disorder characterized by normal to high levels of serum growth hormone and low levels of IGF-I. Analysis of the GHR gene

in Laron-type dwarfism has shown point mutations, deletions, and splicing defects. Growth hormone deficiency in children can be treated by administration of recombinant GH or, in some cases, growth hormone-releasing hormone. Potential targets for recombinant GH that are undergoing clinical investigation include children with idiopathic short stature, persons with wasting syndrome associated with human immunodeficiency virus infection, critically ill patients, and elderly individuals. Excess production of GH due to somatotroph adenomas may be treated by surgical resection, irradiation, or in some cases with somatostatin analogues (e.g., octreotide) that suppress GH secretion, or by a combination of these above.

Prolactin Human prolactin (PRL) contains 199 amino acid residues (M.W. 23,500) and three intramolecular disulfide bridges (Figure 31-6). In healthy adults the anterior pituitary releases very little PRL under nonstressed conditions, primarily because PRL release is under hypothalamic inhibition. This inhibition is exerted by dopamine or PIH. The hypothalamus also secretes PRF but the major regulator appears to be PIH.

742

CHAPTER 31

Endocrine Metabolism I1: Hypothalamus and Pituitary

TABLE 31-6

Opiomelanocortin Family Site of Secretion

Peptide Length

Location of Gene

Adrenocorticotropi c Hormone (ACTH) (Corticotropin)

Anterior Pituitary (Corticotroph)

39 aa

2p25

/3-Endorphin

Anterior Pituitary (Corticotroph)

31 aa

2p25

Hormone

The biological actions of prolactin are initiated by its membrane receptor, followed by the associated intracellular signal transduction pathways. These actions are similar to growth hormone (discussed previously; also see Chapter 30). In humans, the function of PRL may be restricted to promotion of lactation, but there is some evidence that PRL suppresses gonadal function in females. There is no consensus regarding a physiological role for PRL in males. During late pregnancy, the maternal pituitary releases increasing amounts of PRL in response to rising levels of estrogen, a stimulator of PRL synthesis. Elevated levels of PRL stimulate milk production in the mammary gland (Chapter 34). After parturition, PRL promotes milk secretion via a neuroendocrine reflex that involves sensory receptors in the nipples (Chapter 34). In mammary tissue, it binds to alveolar cells and stimulates the synthesis of milk-specific proteins (casein, lactalbumin, and lactoglobulin) by increasing production of their respective mRNAs. Accordingly, there is a lag of several hours before this effect of PRL is seen. In the liver, PRL stimulates the synthesis of its own receptors. PRL receptors occur in the mammary gland, liver, gonads, uterus, prostate, adrenals, and kidney.

Disturbances in Prolactin In women, hyperprolactinemia is often associated with amenorrhea, a condition that resembles the physiological situation during lactation (lactational amenorrhea). Excess PRL may inhibit menstrual cyclicity directly by a suppressive effect on the ovary or indirectly by decreasing the release of GnRH (Chapter 34). In men, hyperprolactinemia is not associated with altered testicular function but is often attended by diminished libido and impotence. This finding suggests that PRL may serve an important role in regulating certain behavior patterns. Prolactinomas are the

Regulation of Secretion

Function Stimulates steroidogenesis in all three zones of the adrenal cortex, thereby increasing secretion of cortisol, DHEAS, and aldosterone. Possible role in stress analgesia,

Stimulated by CRH and inhibited by cortisol.

Stimulated by CRH and inhibited by cortisol.

most common hormone-secreting pituitary adenomas and, in addition to the above-mentioned clinical characteristics, the patient may exhibit visual field defects. Prolactinoma may be treated by surgery, radiotherapy, or pharmacotherapy. This last method consists of using a dopamine D2 receptor agonist such as bromocriptine or cabergoline to suppress prolactin secretion.

The Opiomelanocortin Family All members of this family are derived from a single prohormone, pro-opiomelanocortin (POMC). The prohormone molecule contains three special peptide sequences: enkephalin (opioid), melanocyte-stimulating hormone (MSH), and corticotropin (ACTH). These result from posttranslational cleavage of the prohormone but do not share the same biological actions (Table 31-6). In humans, the gene encoding POMC is normally expressed in the adenohypophysis, hypothalamus, and brain, as well as in other sites. It may also be expressed as a result of neoplastic transformation, notably in the lung. POMC is a large glycoprotein that contains three MSH sequences and one MENK sequence. Several paired basic amino acid residues exist along the molecule and are sites of potential cleavage by proteases during posttranslational processing (Figure 31-7). In humans, the important products of POMC that are secreted by the adenohypophysis are ACTH and fl-endorphin, although other products (e.g., v3-MSH, y-LPH) may also be significant (see later). After translation POMC is processed by proteases contained in tissue. In the adenohypophyseal corticotrophs there is no further processing of ACTH and fi-endorphin, whereas in the intermediate lobe of other species, ACTH can be processed to form ot-MSH, and v-LPH can be processed to form fi-MSH. The human hypothalamus

SECTION31.2 Pituitary Gland (Hypophysis)

743

=m

m

eX

0 ~9

~~

~

o

.,..~

o

._~ ~ ~.~ raN2

SECTION 32.2

763

Adrenal Medulla

chronic, intense cholinergic stimulation increases the activity of tyrosine hydroxylase and dopamine /~-hydroxylase by increased protein synthesis. Epinephrine production is catalyzed by PNMT, which is induced by glucocorticoids (cortisol). The venous drainage of the adrenal cortex, which contains very high concentrations of newly released cortisol, bathes the adrenal medulla before entering the general circulation.

Regulation of Release

utilizes S-adenosylmethionine as the methyl donor and usually initiates inactivation (Figure 32-7). Monoamine oxidase (MAO), a mitochondrial enzyme that oxidizes the amino side chain of catecholamines, acts generally (but not invariably) on methoxylated catecholamines (Figure 32-7). About 70% of the total output of urinary catecholamines is 3-methoxy-4-hydroxymandelic acid (also called vanillylmandelic acid, VMA) (Figure 32-7). Unmodified catecholamines represent 0.1-0.4% of the total.

The major regulator of catecholamine release from the adrenal medulla is cholinergic stimulation, which causes calcium-dependent exocytosis of the contents of the secretory granules. Exocytosis of the granular content releases epinephrine (E), NE, DA, dopamine jS-hydroxylase, ATE peptides, and chromaffin-specific proteins that are biologically inert. The amounts of DA and NE released are minor in comparison with that of E. Of the total catecholamine content in the granules, approximately 80% is E, 16% is NE, and the remainder is mostly DA.

Catecholamines exert their effects through specific receptors on the target cell surface. However, the effects elicited depend on the type or subtype of receptor with which they interact. There are three types of catecholamine receptor: dopamine, oe-adrenergic, and/5-adrenergic. Each of these consists of at least two or more subtypes, which differ with respect to ligand affinity, tissue distribution, postreceptor events, and drug antagonists (Table 32-3).

Transport and Metabolism of Catecholamines

Mechanism of Action

Under basal (unstimulated) conditions in the average adult, NE enters plasma at an average rate of ~5.8 ng/kg/min, almost entirely from adrenergic nerve endings that innervate peripheral tissues (liver, kidney, intestines, pancreas, skeletal muscle, heart, etc.), and with only negligible contribution by the adrenal medulla; E enters plasma at an average rate of "-.3.6 ng/kg/min, entirely derived from the adrenal medulla. Under stimulated conditions, however, the degree to which plasma NE and E levels increase may differ, depending on the nature of the stimulus. For example, whereas physical exercise increases the level of both catecholamines similarly, acute hypoglycemia causes E levels to increase 5- to 10-fold without affecting the levels of NE significantly. As discussed below, the adrenal contribution to NE is physiologically important because it can elevate the plasma level of NE to what is required for a biological effect in peripheral tissues. Circulating catecholamines have an estimated half-life of about one circulation time (20 seconds). They are rapidly taken up by various tissues (notably the liver), where they may exert their effects before they are inactivated. DA may be converted to NE after uptake by nerve endings, kidney, heart, and other tissues. Two enzymes responsible for inactivation of catecholamines are present in most tissues but are particularly abundant in the liver. Catechol-O-methyltransferase (COMT) is a cytosolic, MgZ+-dependent enzyme that catalyzes methoxylation of catecholamines at the hydroxyl group at position 3. COMT

Biological Actions of Catecholamines

Dopamine Receptors Dopamine receptors (D1 and D2) have a greater affinity for DA than for NE or E (Table 32-3). Both subtypes exert effects by altering the activity of adenylate cyciase via a G-protein. D1 receptors coupled to Gs~ activate the enzyme and cause a rise in intracellular cyclic AME D2 receptors are coupled to G~i, and inhibit the enzyme (Table 32-3). At very high concentrations, however, dopamine is capable of activating ot-adrenergic receptors, which results in an effect resembling that of NE (vasoconstriction) instead of the characteristic vasodilatation elicited by low concentrations of DA.

Adrenergic Receptors There are two types of adrenergic receptors, designated ot and/5. Two subtypes of o~-adrenergic receptors (Oel, ol2) and three subtypes of/3 adrenergic receptors (/~1,/52,/33) have been identified according to their differing affinities or susceptibilities to synthetic ligands with agonist or antagonist biological activity. Table 32-3 shows the five subtypes of adrenergic receptors, their relative affinities for E and NE, their subtypespecific agonists and antagonists, their tissue distribution, and the biological effects they mediate. Although both E and NE are capable of binding to and activating any of the adrenergic receptor subtypes (albeit to differing extents), the receptor subtypes may not be equally

|

eq

764

~m

.,,,i

0

!

9

~

r,9

~

~

~.~.~.

~

.~

~

~'~

.=.

.~=

zz

..,:

"~ r

~

~

zz

~

zzz

z

9

zzz

,~.

~ ~

"~ -~

~ "o-o~

~

~9. ~

~

~ ~

~ .,,

.~ c~ >

o~

. ~

Z~m

~'~''~E~ o ~..~'~c~ ~ c ~ - ~.=..-~ E

z

"~

~

~.~

...'-'~

~.~ . ~

~

~9 ~ 9

~~z

-~

~ ~

"~

~

~ ~ ~ o o ~ .

~

~zz

~~ =

~ ~

"o"o

~

,,~

~

: ~ ~ ~ ~

,~

~~

~

o

.,..~

mZ

~

_

.2

~

r

-~

::

.2 ~

.~

~:~::

.2 ~ _

:z:~

~ .~. ~

_~~

~ ~ _

_~=

.. ... ~

_~c~

.-

r./'~ r.,/'~

Z~

LT..,

%

.~

9- ~

~D

.=.

~

.

[',,,,1



*~,

.-

.-

~,

II

~

E~

AA

=

Z

A

=.=

~,

~~ ~

~ AA

All

9

.: ~,

Z

II

r..T..1

.~ ~.

.~.~

9

'~ < . :

~D

LT..1

Z II AI

r---

=

~1~

9

.~

e'~

t,~

9

.

~'

e~

~::~ " ~

..,..... o,..

~

.~ .~ ~ .~

~,-~

~

.,.~ 9 ~

.,..~ ~

~

~.,~

~

m

"~Z

"~ ~.~.

~

..~

m

~

~~ ~.~

"2._~

SECTION32.2 Adrenal Medulla

accessible to both circulating (E) and neurotransmitted (NE) catecholamines. In fact, thedistribution of the subtypes appears to be appropriate for the ligand that is prevalent in a given region. For example, the /~2-adrenergic receptors (E > NE) in bronchiolar smooth muscles are located in regions of the respiratory system that are poorly supplied by sympathetic nerve fibers but are accessible to agents that arrive by way of the blood supply. These receptors are most likely to respond to circulating E than to neural NE, and are responsive to inhaled synthetic adrenergic agonists (e.g., albuterol, salmeterol) that diffuse through the mucosal surface. On the other hand, the/~ 1-adrenergic receptors in the juxtaglomerular apparatus (JGA) of the kidney, the site of renin release that is richly supplied by sympathetic nerve fibers but is also well vascularized, tend to respond preferentially to neural NE rather than to plasma E, despite the fact that this subtype is known to have equal affinity for both catecholamines (Table 32-3). This suggests that the/31 receptors of the JGA are localized in the synaptic clefts as postjunctional binding sites and are not accessible to plasma-derived E. To facilitate the discussion on the effects of catecholamines (below), Table 32-3 includes a list of the "physiological ligand" (i.e., neural NE or plasma E) for the adrenergic receptor subtype in some of the tissues, based on their efferent sympathetic nerve supply. The different tissue distribution of adrenergic receptor subtypes indicates the genetic influence on the kinds of receptors; however, the density of the receptor subtypes also exhibits tissue variation. For example, although white adipose tissue contains both/31 and/33 receptors, the latter is in greater quantity in visceral adipose tissue. The density of adrenergic receptors is strongly influenced by hormones. The adrenergic catecholamines exert an important regulation on their own receptors, causing downregulation when the concentrations are raised. All subtypes of adrenergic receptors except the/33 subtype is subject to downregulation. Thyroid hormone increases the density of/3-adrenergic receptors in some tissues (heart, fat, skeletal muscle), but decreases it in the liver. Glucocorticoids (GC) increase the density of/~2-adrenergic receptors in vascular and bronchiolar smooth muscle, and has been shown to be effective in reversing agonist-induced desensitization of/32-adrenergic receptors in asthmatic patients.

Biological Effects of Epinephrine and Norepinephrine (Table 32-3) Although NE is present at higher concentrations in plasma than E (Table 32-1), plasma NE is physiologically ineffective under most conditions. Plasma NE levels above

765

100 ng/dL are required for a systemic response, and such concentrations usually are produced only during very stressful situations (e.g., strenuous exercise or myocardial infarct). In contrast, E is capable of activating adrenergic receptors at threshold concentrations of 10-15 ng/dL, levels that are attained with relatively mild stimuli (e.g., cigarette smoking or hypoglycemia). E exerts important cardiovascular, pulmonary, renal, metabolic, endocrine, and thermogenic effects, most of which are supported or complemented by neural effects of NE that are exerted at the same time.

Cardiovascular, Pulmonary, and Renal Effects E increases cardiac output by increasing both the strength and the rate of ventricular contractions (/~1). This effect is augmented by the constrictive effect of E (ot2) and NE (ot2) on the great veins, which increases venous return to the heart and leads to increased ventricular preload and increased stroke volume. The increased cardiac output is selectively directed to certain organs by the combined effects of circulating E and neurotransmitted NE. Thus, blood flow to the skin (subcutaneous) is reduced by the combined vasoconstrictive effects of E (oe2) and NE (oil), while flow to the gastrointestinal tract, spleen, pancreas and kidney are reduced by sympathetic (NE) stimulated vasoconstriction (oil). At the same time, E redirects the flow of blood through skeletal and cardiac muscles by promoting dilatation of the arterioles of the skeletal muscle (/32) and the coronary arteries of the heart (/32). Because the arteries in the brain, lungs, and liver are not directly affected by the adrenergic catecholamines, these tissues passively receive an increase in blood flow as a result of the augmented cardiac output and increased blood pressure secondary to increased peripheral resistance.

Pulmonary Respiratory System E promotes air flow through the bronchioles by causing relaxation of their smooth muscle (bronchodilatation) (/32), and thus allows for increased alveolar ventilation. The increased blood flow through the lungs (see above) and the increased alveolar ventilation ensure maximal oxygenation of blood.

Renal Urinary System Autonomic discharge results in stimulation of ADH release due to a neurotransmitted NE effect (oe1) on the magnocellular neurons in the hypothalamic paraventricular nuclei. This promotes increased retention of water, which, in turn, promotes the effective blood volume. In addition, autonomic discharge causes the release of neurotransmitter

766

NE at the JGA of the kidney and this stimulates the release of renin (/~), ultimately resulting in aldosteronemediated sodium retention at the nephron. The overall result is preservation and promotion of vascular volume due to isotonic fluid retention, decreased urine flow, and decreased glomerular filtration rate, all of which are attributable to neurotransmitted NE. Paradoxically, elevated levels of E and NE can cause a reduction in plasma volume that can lead to hemoconcentration and poor tissue perfusion; however, the mechanism of this effect is not known. This is said to be the cause of orthostatic hypotension in untreated patients with pheochromocytoma, a tumor of chromaffin tissue that produces excessive amounts of NE; most of these patients have reduced plasma volume but exhibit hypertension while in the reclining position.

Metabolic, Endocrine, and Thermogenic Effects E promotes the production and release of glucose from the liver by two related mechanisms: 1. A direct stimulatory effect on hepatic glycogenolysis (/32) mediated by cAMP-dependent phosphorylation of phosphorylase. This effect is dependent on prior storage of glycogen in the liver; therefore, both insulin and cortisol serve to condition the liver for this effect. 2. A direct activation of gluconeogenesis in the liver (/~2) via cAMP-dependent phosphorylation of the key gluconeogenic enzymes. This effect requires prior induction and maintenance of enzyme concentrations by cortisol. These two glucose-generating actions of E on the liver are enhanced in an additive fashion by glucagon which, like E, depends on cAMP mediation and cortisol conditioning. The fact that neural NE simultaneously causes a fall in the insulin/glucagon ratio indicates that the sympathetic nervous system has a supportive role. Neural NE and plasma E are both stimulators of adipocyte lipolysis, an effect that is mediated by/~ l- and /~3-adrenergic receptors and involves cAMP-mediated phosphorylation of hormone-sensitive lipase (HSL). There are regional differences in /33-adrenergic receptor density (visceral fat has more than subcutaneous fat), which determines that catecholamine is physiologically important. Note that/~3 receptors are low-affinity receptors that require high concentrations of catecholamines. They respond better to neural stimulation because of the higher local concentration of NE at the fat tissue site. Cortisol, thyroid hormone, and GH support this effect of the catecholamines, presumably by promoting the synthesis of one or more components of the lipolytic pathway.

CHAPTER32 Endocrine Metabolism II1: Adrenal Glands

This results in the release of free fatty acids (FFAs) and glycerol. Glycerol is taken up by the liver and is used for the production of glucose via gluconeogenesis (/~2). FFAs are taken up by cardiac muscle, skeletal muscle, kidney, and liver, and are oxidized for energy which obliges these tissues to consume less glucose. In the liver, where FFA uptake is related to circulating FFA levels, the oxidation of acyl-coA also is shunted to ketogenesis; ketone body production increases as a result (Chapter 18). E can promote FFA utilization and ketogenesis in the liver by inhibiting acetyl-CoA carboxylase activity (/32); this results in unrestricted entry of FFA into the mitochondria for oxidation and subsequent ketogenesis. Ketone bodies are consumed by extrahepatic tissues that possess succinyl-CoAacetoacetate-CoA transferase, also known as thiophorase (cardiac muscle, skeletal muscle, intestines, kidney, and the brain during starvation), and this also has a glucosesparing effect. E stimulates skeletal muscle glycogenolysis (/~2) and thus promotes glycolysis. The effects of E may depend on skeletal muscle activity because contraction alone is a stimulus of both glycogenolysis and glycolysis; the effects may also be influenced by the muscle fiber type (types I, IIa, or IIb). In resting muscle and presumably in type I (slow-twitch) fibers, there is preferential use of fatty acids and ketone bodies for energy, which also inhibits glycolysis; thus, the glycogenolytic and glycolytic effects of E may be minimal or overriden. In the contracting muscle and presumably in fast-twitch (types IIa and IIb) fibers, E may potentiate the active glycogenolytic and glycolytic pathways and promote the release of both lactate and alanine. Indirectly, E can inhibit skeletal muscle glycolysis by increasing the supply of FFAs and ketone bodies, which are glucose-sparing energy fuels for this tissue; however, the increased turnover of ATP in the contracting muscle probably allows the tissue to accommodate both glucose and fatty acids as fuels (Chapter 21). A potentially harmful effect is the stimulation by E of potassium ion movement from plasma into skeletal muscle and liver (/~2), which can lead to hypokalemia. This effect is accompanied by a decrease in potassium ion excretion by the kidney. The ability of E to stimulate plasma membrane Na+,K+-ATPase activity in skeletal muscle (/~2) may partially explain this hypokalemic action, as well as the thermogenic effect of the hormone (see below). The direct adrenergic innervation of the islets of Langerhans in the pancreas allows NE to stimulate the release of glucagon from the o~ cells (/~2) and to inhibit the release of insulin from the /~ cells (o~2), causing the insulin:glucagon (I:G) ratio to fall. Circulating E also exerts these effects and is probably important in maintaining the low I:G ratio following neural stimulation. Because the

767

Supplemental Readings and References

effects of a lowered I:G ratio are similar to those of NE and E on glycogenolysis, gluconeogenesis, lipolysis, and ketogenesis, the outcome is a great enhancement of the metabolic response to adrenergic discharge. In the neonate, neural NE promotes "nonshivering thermogenesis," i.e., heat production via stimulation of lipolysis and fatty acid oxidation in brown adipose tissue (/33) with the support of thyroid hormone. Brown adipose tissue contains a high density of mitochondria with an "uncoupling protein" that allows the cells to oxidize fatty acids and generate heat as a major product; NE released at nerve endings stimulates cAMP-mediated lipolysis in these cells and promotes thermogenesis by this fl3-mechanism (Chapter 14). In the adult, nonshivering thermogenesis is probably a minor source of body heat production because brown adipose tissue is sparse and limited to only a few regions of the body. Adults depend on neurogenically induced shivering thermogenesis, which does not depend on adipose lipolysis. However, even in the resting state, catecholamines stimulate thermogenesis by promoting the thermogenic effect of thyroid hormone. Thus, when administered to adults, E increases resting body temperature and oxygen consumption (fl), in part due to stimulation by E of skeletal muscle Na+,K+-ATPase activity (f12).

Disturbances in Adrenal Medullary Function Because there is considerable overlap in the functions of the sympathetic nervous system and the adrenal medulla, elimination of the adrenal medulla would be tolerated as long as the autonomic nervous system remains functionally intact. However, overproduction of adrenal medullary hormones would be disruptive. Such catecholamine excesses are seen in patients with tumors of the adrenal medullary chrornaffin cells and/or tumors of chromaffin tissue located outside of the adrenal gland. These tumors are called pheochromocytomas, and, although their incidence is rare, the pathophysiology should be understood for full appreciation of adrenergic catecholamine functions under normal conditions. One site at which pheochromocytomas often develop is the organ of Zuckerkandl, a collection of pheochromocytes at the bifurcation of the aorta. Familial predisposition to pheochromocytoma can occur due to activating mutations of the RET

proto-oncogene that cause multiple endocrine neoplasia type II (Chapter 37) and mutations in the von HippelLindau tumor suppressor gene. Pheochromocytoma is usually characterized by intermittent to permanent hypertension with potentially life-threatening consequences. The biochemical diagnosis in patients suspected of pheochromocytoma consists of measuring epinephrine, norepinephrine, and their metabolitesmnamely, metanephrine, normetanephrine, dihydroxyphenylglycol, and vanillylmandelic acidmin a 24-hour urine specimen. Measurement of plasma levels of total free metanephrine has a very high sensitivity for detecting pheochromocytoma. Tumors of the pheochromocytoma can be surgically removed.

Supplemental Readings and References E.-E. Baulieu: Dehydroepiandrosterone (DHEA): a fountain of youth? Journal of Clinical Endocrinology and Metabolism 81, 3147 (1996). S. R. Bornstein, C. A. Stratakis, and G. R Chrousos: Adrenocortical tumors: recent advances in basic concepts and clinical management. Annals of Internal Medicine 130, 759 (1999). R R. Casson, S. A. Carson, and St. J. E. Buster: Replacement dehydroepiandrosterone in the elderly: rationale and prospects for the future. Endocrinologist 8, 187 (1998). K. C16ment, C. Vaisse, B. S. J. Manning, et al.: genetic variations in the fl3adrenergic receptor and an increased capacity to gain weight in patients with morbid obesity. New England Journal of Medicine 333, 352 (1995). A. Ganguly: Primary aldosteronism. New England Journal of Medicine 339, 1828 (1998). T. L. Goodfriend, M. E. Elliott, and K. J. Catt: Drug therapy: angiotensin receptors and their antagonists. New England Journal of Medicine 334, 1649 (1996). R A. Insel: Adrenergic receptors--evolving concepts and clinical implications. New England Journal of Medicine 334, 580 (1996). M. New: The prismatic case of apparent mineralocorticoid excess. Journal of Clinical Endocrinology and Metabolism 79, 1 (1994). W. Oelkers: Current concepts: adrenal insufficiency. New England Journal of Medicine 335, 1206 (1996). D. N. Orth: Cushing's syndrome. New England Journal of Medicine 332, 791 (1995). T. M. Penning: Molecular endocrinology of hydroxysteroid dehydrogenases. Endocrine Reviews 18, 281(1997). K. Pacak, W. M. Linehan, G. Eisenhofer, et al.: Recent Advances in Genetics, Diagnosis, localization, and Treatment of Pheochromocytoma. Annals of Internal Medicine 134, 315 (2000). R M. Stewart: Mineralocorticoid hypertension. Lancet 353, 1341 (1999). R C. White: Disorders of aldosterone biosynthesis and action. New England Journal of Medicine 331, 250 (1994). E C. White and R W. Speiser: Congenital adrenal hyperlpasia due to 21hydroxylase deficiency. Endocrine Reviews 21,245 (2000).

CHAPTER

33

Endocrine Metabolism IV: Thyroid Gland

The thyroid gland consists of two lobes connected by an isthmus and is positioned on the ventral surface of the trachea just below the larynx. It receives adrenergic fibers from the cervical ganglion and cholinergic fibers from the vagus and is profusely vascularized by the superior and inferior thyroid arteries. Histologically, thyroid tissue is composed of numerous follicles lined by a single layer of epithelial cells around a lumen filled with proteinaceous material called colloid. The follicle cells, called thyrocytes, produce the thyroid hormones and are derived from the entodermal pharynx. Interspersed between follicles are specialized APUD cells derived from the neural crest, called C-cells or parafollicular cells. These cells produce calcitonin (CT), a polypeptide hormone discussed in Chapter 37.

33.1 Structure-Activity of Thyroid Hormones The generic term "thyroid hormones" refers to the iodinated amino acid derivatives T3 (3,3',5-triiodo-Lthyronine) and T4 (3,3',5,5'-tetraiodo-L-thyronine), the only iodinated hormones produced endogenously. T3 is the biologically active hormone and is, for the most part, produced from T4 in extrathyroidal tissues. T4 lacks significant bioactivity and is a hormone precursor; however, A list of expanded acronyms appears as Appendix VIII.

since thyroidal production of T4 and its handling by the body are the pivotal determinants of thyroid hormone effects, the term "thyroid hormone" includes T4. The basic structure of T3 and T4 is that of thyronine, a substituted tyrosine [0-(4-hydroxyphenyl)tyrosine] (Figure 33-1). Iodine residues at positions 3 and 5 of the inner phenolic ring confer on the outer ring a preferred orientation approximately 120 ~ to the plane of the inner ring (Figure 33-2). Hormonal activity is maximal when the following requirements are met: 1. Iodine is present at positions 3 and 5 of the inner ring (required for receptor binding). A natural analog of T3, called reverse T3 (rT3, 3,3',5'-triiodo-L-thyronine), is inactive. Substitution of iodine by bromine or a methyl group almost completely abolishes the activity. 2. A substituent is present at position 3', but it need not be iodine. One of the most potent synthetic thyroid hormones has an isopropyl group at position 3'. 3. A hydroxyl group is present at position 4'. This group forms a hydrogen bond with the receptor protein. 4. A substituent is absent at position 5'. This permits full expression of the activity of the hormone. A substituent at this position would cause steric hindrance in binding with receptor and interfere with hydrogen bond formation. Thus, T4 lacks significant activity whereas T3 has about six times the activity of T4.

769

770

CHAPTER33 Endocrine Metabolism IV: Thyroid Gland

Diphenylether I

i

NH=

,/ I

\

I

Alanine

I

Diiodotyrosine

F I G U R E 33-1 Structure of 3,3',5,5'-tetraiodo-L-thyronine (T4). T4 is a substituted tyrosine or a diphenylether derivative of alanine. Positions in the outer phenolic ring have prime designations, in contrast to those in the inner ring. T4 is iodinated at positions 3 and 5 in both rings. [Modified and reproduced with permission from V. Cody, Thyroid hormone interactions: molecular conformation, protein binding, and hormone action. Endocr. Rev. 1, 140 (1980). @ 1980 by the Endocrine Society.]

5. The molecule must be the L form. D Isomers have minimal biological activity; for example, D-T3 has about 7% the activity of L-T3.

33.2 Thyroid Hormone Synthesis Two substrates are required in the synthesis of thyroid hormones. The intrinsic substrate is thyroglobulin (Tg),

F I G U R E 33-2 Skewed conformation of T4. The molecule shown at the top is viewed in parallel to the plane of the inner phenolic ring, while that shown at the bottom is viewed perpendicularly to the plane of the inner ring. The ether linkage between the two rings is angled at about 120 ~ [Reproduced, with permission from V. Cody, Thyroid hormone interactions: molecular conformation, protein binding, and hormone action. Endocr. Rev. 1, 140 (1980). @ 1980 by the Endocrine Society.]

a homodimeric glycoprotein (M.W. 669,000) synthesized in the rough endoplasmic reticulum of the thyrocytes and secreted into the follicular lumen by exocytosis. Tg contains 134 tyrosyl residues, only 25-30 of which undergo iodination and only four of which become a hormonogenic segment of the molecule. The extrinsic substrate is elemental iodine, present in food as inorganic iodide. Iodide is readily absorbed via the small intestine; it is almost entirely removed from the general circulation by the thyroid and kidney. Iodide that is taken up by salivary and gastric glands is secreted in salivary and gastric fluids and is returned to the plasma iodide pool by intestinal reabsorption. A small amount of iodide is taken up by the lactating mammary gland and appears in milk. The synthesis and release of thyroid hormone involves a number of steps (Figure 33-3).

1. Uptake oflodide.Iodide (I-) is actively taken up by the follicular cells against electrical and concentration gradients. The active uptake of I- is mediated by the Na+/I - symporter, an intrinsic membrane protein with approximately 13 transmembrane segments. The Na + and I- bound symporter releases both Na + and I- on the cytoplasmic side. The empty symporter then returns to its original conformation exposing binding sites on the external surface of the cell. The internalized Na + is pumped out of the cell by the ATP-dependent Na +, K+-ATPase (oubain sensitive) to maintain an appropriate ion gradient. In the normal gland, the limiting step of thyroid hormone synthesis is uptake of I-. Both I- and thyroid-stimulating hormone (TSH) regulate the Na+/I - symporter function. TSH promotes I- uptake whereas excess Idecreases I- uptake (discussed later). Anions of similar charge and ionic volume (e.g., C10 4, BF-, TcO 4, SCN-) compete with I- for transport in the follicular cell. 2. Activation and Organification of lodide. Iodide that enters thyrocytes is "activated" by oxidation that is catalyzed by thyroperoxidase (TPO). Hydrogen peroxide is required and is supplied by an H202-generating system which may be an NADPH (NADH) oxidase system similar to that of leukocytes (Chapter 15). Thyroperoxidase is a glycosylated heme enzyme that is bound to the apical plasma membrane of the thyrocyte. Thyroperoxidase exists in two molecular forms (M.W. 105,000 and 110,000) and its catalytic domain faces the colloid space. In thyroid autoimmune disorders, one of the major microsomal antigenic components is thyroperoxidase. In vitro, iodide peroxidase catalyzes the reaction 2H+§ 21-+ H202 -+ 12 -4- 2H20. However, in vivo

SECTION 33.2

Thyroid Hormone Synthesis

771

33-3 Synthesis of thyroidhormonein the thyrocyte.Tg, Thyroglobulin.

FIGURE

the enzyme probably forms an enzyme-bound iodinium ion (E-I +) or a free radical of iodine. These activated derivatives iodinate the tyrosyl residues of Tg ("organification of iodide") by the action of thyroperoxidase and H202. Thionamides (e.g., thiourea, thiouracil, propylthiouracil, methimazole) inhibit this organification of iodide without affecting iodide uptake. The first position iodinated is position 3, which forms monoiodotyrosyl-Tg (MIT-Tg). The second is position 5, which forms diiodotyrosyl residues (DIT-Tg). Iodination is accompanied by structural changes (e.g., cystine formation) and includes a conformational change in the Tg molecule (illustrated diagrammatically in Figure 33-3). Activation and organification occur at the cell-colloid interface where thyroperoxidase activity is prevalent. 3. Coupling Reaction and Storage as Colloid. Iodide peroxidase or a "coupling enzyme" catalyzes the "coupling reaction" at the cell-colloid interface by intramolecular coupling of two iodotyrosyl residues with formation of an iodothyronyl residue. Coupling of DIT residues is favored; thus, formation of T4

residues predominates. The coupling reaction requires that both substrates be iodinated and that one of the substrates be DIT-Tg. For this reason, no To, T1, or T2 residues are formed. Under normal conditions less than half of the iodotyrosyl residues in Tg undergo coupling such that of the total iodinated residues in Tg, 49% are MIT, 33% DIT, 16% T4, 1% T3, and a trace amount rT3. The ratio of T4 to T3 is > 10 on a typical American diet, but it rises or falls with dietary iodine content. The coupling reaction appears to be the most sensitive to inhibition by the thionamides (propylthiouracil, methimazole), being inhibited at doses that do not inhibit the organification reactions. As a final note, although proteins other than Tg are iodinated in nonthyroid tissues that take up iodine (e.g., salivary gland, mammary gland, intestinal mucosa), no coupling of the iodinated residues occurs. Thus, the requirements for the coupling reaction may be more stringent than previously believed. 4. Processing of TG and Release of Thyroid Hormone. Thyroglobulin, with its tetraiodothyronyl residues, is a pre-prohormone stored in the follicular lumen.

772

CHAPTER 33

When follicle cells are stimulated by TSH or TSI (see below), their luminal border ingests colloid (and hence Tg) into endocytotic vesicles. These vesicles fuse with lysosomes to form phagolysosomes, which, during transit toward the basolateral surface of the cell, hydrolyze Tg and release T4, DIT, MIT, and a small amount of T3. These substances are released into the cytosol, and T4 and T3 diffuse into the blood. MIT and DIT do not enter the circulation in significant amounts because they are rapidly deiodinated by iodotyrosine deiodinase, an enzyme complex that contains ferredoxin, NADPH: ferredoxin reductase, and a deiodinase containing flavin mononucleotide (FMN). This reaction promotes recycling of iodide within the follicle cell. Some of the iodide, however, diffuses into plasma and constitutes the daily "iodide leak," estimated to be about 16 #g/d. By mechanisms that are not clear, a small amount of intact Tg also leaks out of the thyroid and can be found circulating in plasma in normal individuals. Tg leakage at a rate of 100 #g/d has been reported in euthyroid individuals, and a concentration of 15-25 #g Tg per liter of serum is considered to be normal. The route of Tg leakage is by way of the lympathic drainage and is increased when the gland is excessively stimulated. Increased entry of Tg into the circulation may result in an immune response because of the antigenic nature of this glycoprotein. The release of thyroid hormone following endocytosis of colloid is inhibited by iodide and is known to be reduced with high dietary intake of iodine. This inhibition is due to the inverse relationship between the iodide content of Tg and the digestibility of iodo-Tg by lysosomal peptidase; that is, poorly iodinated Tg is more readily digested than richly iodinated Tg.

Regulation of Thyroid

H o r m o n e Synthesis

Thyroid-Stimulating Hormone (TSH) TSH (also called thyrotropin), which is secreted by the pituitary, plays a central role in the regulation of growth and function of the thyroid gland. TSH receptors are functionally coupled to G-proteins; thus, the extracellular stimulus by TSH is transduced into intracellular signals mediated by a number of G-proteins. Activation of Gs-protein results in the stimulation of the adenylate cyclase-cAMP-protein phosphorylation cascade. Other G-proteins coupled to TSH-receptor activation include GQ-protein, which mediates the phospholipase C-phosphatidylinositol 4,5-bisphosphate-Ca 2+ signaling pathway (see Chapter 30 for a detailed discussion).

Endocrine Metabolism IV: Thyroid Gland

The TSH receptor is a glycoprotein that consists of three major domains: a long amino-terminal extracellular segment that confers binding specificity, a transmembrane segment, and a carboxy-terminal intracytoplasmic segment that mediates G-protein interactions (see Figure 30-6). TSH-receptor activation promotes iodide trapping, organification of iodide, and endocytosis and hydrolysis of colloid by a mechanism that involves cAME Thyroid disorders consisting of either hypofunction or hyperfunction can result form constitutive inactivating or activating mutations, respectively, in the TSH receptor. Similarly, Gs~-inactivating (loss of function) or activating (gain of function) mutations can result in hypothyroidism and hyperthyroidism. An example of the former is Albright hereditary osteodystrophy and the latter is McCune-Albright syndrome. Both syndromes are discussed in Chapter 30. All of the effects of TSH on the thyroid gland are exaggerated in hyperthyroidism due to excessive stimulation of the TSH receptor whether or not the ligand is TSH. For example, in Grave's disease hyperactivity of the thyroid gland is due to a thyroidstimulating antibody that binds the TSH receptor and, in so doing, activates it. This thyroid-stimulating immunoglobulin (TSI) or long-acting thyroid stimulator (LATS) is an expression of an autoimmune disease that may be hereditary, although in some cases it may be the result of a normal immune response. For example, certain strains of bacteria contain a membrane protein which is homologous to the TSH receptor and which elicits an immune response in infected individuals. The antibodies generated resemble TSI. Thus, some cases of hyperthyroidism may be due to cross-recognition across phylogenetic lines (molecular mimicry). Human chorionic gonadotropin (hCG), a placental hormone, has some TSH-like activity, hCG synthesis is initiated during the first week after fertilization and reaches its highest level near the end of the first trimester, after which levels decrease. The action ofhCG on thyrocytes may help meet the increased requirement of thyroxine during pregnancy. The total thyroxine pool is increased in pregnancy due to elevated levels of thyroxine-binding globulin. However, the serum free T4 level, which represents the biologically active form, remains the same compared to nonpregnant status. During the first trimester, the fetal requirement for thyroxine is met by maternal circulation until the fetal pituitary-thyroid axis becomes functional late in the first trimester. The maternal-fetal transport of thyroxine which occurs throughout pregnancy minimizes the fetal brain abnormalities in fetal hypothyroidism (discussed later). Maternal hypothyroidism (e.g., due to women consuming iodine-deficient diets) is associated with defects in the neurological functions of offspring.

SECTION

33.2 Thyroid Hormone Synthesis

773

Cold

Stress

Other] P'~--,O/ stimuli/7 Adrenergicneural network J i Norepinephrine i ,,

(9

: I Hypothalamus

TRH

Som~Starvation

an

Estrogens

(chronic)

1

TSI

TSH

[Thy;oidi r4

Peripheral tissues

; l~

J

Starvationand Non-thyroidal illness

"1"3----.-Targetcells

: Activation of metabolism,required for growth differentiation and mental development

FIGURE 33-4 Schematicrepresentationof the major stepsin the regulationof thyroidhormonesecretionsand metabolismat five levels, namely,brain, hypothalamus,pituitarythyrotropes,thyroid, and peripheraltissues.

Release of TSH from the anterior pituitary is stimulated by hypothalamic TRH and inhibited by thyroid hormone (Chapter 31). These opposing signals to the anterior pituitary determine the magnitude of TSH secretion. Many other factors influence secretion of TSH and thyroid hormone (Figure 33-4). For example, starvation reduces conversion of T4 to T3 in peripheral tissues but not in the anterior pituitary; consequently, TSH secretion is not suppressed but circulating T3 levels fall. Stress, somatostatin, estrogens, and cold exposure (in infants) also affect TSH release, but except for cold stress in infants, they are not regarded as important regulators of TSH and thyroid hormone secretion. Iodine

The thyroid gland is capable of adjusting its synthetic activity to the supply of iodine. When the iodine supply

is low, the thyroid makes maximal use of iodide; when the iodide supply is abundant, the thyroid defends itself against hormone overproduction by reducing iodide uptake. The thyroid gland autoregulates its iodine supply by an internal feedback mechanism that controls the intraglandular handling of iodide and the thyroid response to TSH. The exact nature of this feedback is not known, but the amount of the organified iodide pool is believed to have an inhibitory effect on the rate of iodide uptake, intraglandular T4/T3, the completeness of thyroglobulin hydrolysis, and the responsiveness to TSH. This autoregulatory mechanism is not prominent when iodine intake is within the normal range but becomes physiologically significant when there is a moderate deficiency or excess of iodine. On an ordinary diet in which the iodine intake ranges between 300 #g and 1000 #g/d, T4 production increases linearly with increased consumption

774

CHAPTER 33

of iodine. With lower iodine intake (e.g., 50-150 #g/d), T4 production increases due to increased efficiency in iodide processing and to a greater response to TSH; in fact, although plasma TSH levels are within the normal range, a mild enlargement of the gland is not an uncommon consequence (e.g., nontoxic goiter). On the other hand, with a diet enriched in iodine (e.g., 1-2 mg/d), the increase in T4 production as a function of iodine consumption is subnormal, due to negative feedback of the organified iodide pool on T4 production and the thyroid response to TSH; as a result, plasma TSH levels are maintained within the normal range. Abnormal thyroid function is seen at the extremes of iodine intake: at the lower end, less than 50 #g/d; at the upper end, 5 mg/d or more. At the lower end, frank hypothyroidism occurs because there is an insufficient supply of iodine to meet the T4 production demands despite the efficiency with which the limited iodide is used. In this condition, the iodide deficiency creates an organified iodide pool consisting of MITs and a paucity of DITs, and, consequently, little or no coupling occurs because substrate requirements are not met. Products of Tg digestion are mainly MITs, small amounts of DITs, and possibly some T3 and T4. The decrease in thyroid hormone increases output of TSH which, in its futile attempt to increase T4 production, creates glandular overgrowth due to thyrocyte hypertrophy and colloid accumulation ("hypothyroid goiter," "colloid goiter," or "endemic goiter"). At the upper end, further increase in iodine intake causes a shut-down of thyroid gland function and acutely reduces the thyroid response to TSH; consequently, there is a paradoxical decline in T4 production as a function of iodine consumption accompanied by a dramatic reduction in vascular supply to the gland. This phenomenon is called the Wolff-Chaikoff effect and is observed only in

Endocrine Metabolism IV: Thyroid Gland

thyroid glands that are stimulated (e.g., with TSH or in hyperthyroidism).

33.3 Transport and Metabolism of Thyroid Hormones Thyroid hormones in blood are mainly bound to serum proteins. The major thyroid hormone-binding protein in blood is thyroxine-binding globulin (TBG), a glycoprorein (M.W. 63,000) of electrophoretic mobility between that of ot 1- and ot2-globulins. TBG exhibits a high affinity for T4 (Kd -- 50 pM) and a moderate affinity for T3 (Ko = 500 pM). Approximately three-fourths of circulating T4 is firmly bound to TBG, while about one-half of circulating T3 is TBG-bound (Table 33-1). Thyroxine-binding prealbumin (TBPA), more properly known as transthyretin, which functions in the transport of retinol, has about 1% of the affinity of TBG for T4, but it accounts for about 20% of circulating T4 and does not bind T3. These differences in binding can be partially explained, since T4 is almost completely ionized at physiological pH, whereas T3 is not. The 4'-hydroxyl group of T4 probably exists as a 4'-phenoxide ion, which optimizes binding to serum proteins. However, the 4'-hydroxyl group is required for receptor binding. Binding to albumin accounts for much of the circulating T4 and T3 that is not bound to TBG and TBPA. About 0.03% of T4 and about 0.3% of T3 is unbound, so that only a small fraction is accessible for target cell uptake. The albumin-bound hormone dissociates rapidly in the vicinity of target cells. The cellular uptake of T4 and T3 is mediated by carrier-mediated processes at stereospecific binding sites. At high-affinity sites the uptake is energy, temperature, and often Na +dependent. T3 is metabolized about six times more

TABLE 33-1

Source, Transport, and Metabolism of the Thyroid Hormones

T4 3, 5, 3, 5'-Tetraiodo-L-thyronine Molecular weight Production rate (pg/day) % Produced by thyroid gland Half-life Total plasma concentration % Free % Bound to plasma proteins % Bound to TBG % Bound to albumin % Bound to transthyretin

777 90 100 7 days 51-142 nmol/L (4-11 gg/dl) 0.02% 99.98% 70-75 5-10 15-20

3, 5,

T3 3'-Triiodo-L-thyronine

651 35 20 0.75 day 1.2-3.4 nmol/L (75-220 ng/dl) 0.3% 99.7% 70-75 20-25 0-5

SECTION 33.3 Transport and Metabolism of Thyroid Hormones

775

rapidly than T4 and has a metabolic clearance rate (MCR) 25 times as high as that of T4 (Table 33-1). Mutations in a human serum albumin gene that substitute either histidine or proline for arginine at position 218 increase binding affinity for T4. These mutations are autosomal dominant and occur with relatively high frequency. Carriers of these mutations have high levels of total serum T4 but their free-T4 and TSH concentrations are within the normal range. Individuals are euthyroid (normal thyroid) and their conditions are known as familial dysalbumine-

mic hyperthyroxinemia (FDH). Two reactions account for the metabolic fate of about 80% of the T4 in plasma: about 40% is converted to T3 via 5'-deiodination (activation), and another 40% of the T4 is converted to rT3 by 5'-deiodination (inactivation). These two reactions are catalyzed by three enzymes designated types I, II, and III iodothyronine deiodinases (Figure 33-5 and Table 33-2). Types I and II both catalyze the 5'-deiodination reaction but differ with respect to substrate specificity, tissue distribution, and regulation. Type III is a 5-deiodinase, which catalyzes the removal of iodine from position 5 of the inner ring. Type I is deiodinase selenocysteine-containing microsomal enzyme present in the liver, kidney, and thyroid, with specificity for

HO-- ii>--O __

thyronines bearing iodine at position 5'. It is responsible for catalyzing the formation of most of the circulating T3 and is a high-capacity (Km ~1 #M) processor ofT4. A unique feature of the type I enzyme is that it is sensitive to inhibition by propylthiouracil (PTU), a known inhibitor of thyroid gland function that has little or no effect on the activity of the other deiodinases. It is also inhibited in conditions such as starvation, caloric restriction, and nonthyroid-related illnesses, which may deplete the cofactor supply and/or depress enzyme synthesis. The type II deiodinase is less abundant and has a higher specificity and a higher affinity for T4 than type I. The type II enzyme is present in the pituitary, brain, brown fat, and skin, and is responsible for intracellular T3 formation that leads to a cellular response; whereas the type I enzyme produces T3 for export to other cells, and the type II enzyme is selfserving, producing its own supply of intracellular T3. A unique feature of the type II enzyme is that it is sensitive to inhibition by T4 by an unknown mechanism. The activity of the type II enzyme is increased in hypothyroidism and is depressed in hyperthyroidism, whereas these conditions have the opposite effect on the activity of the other two deiodinases. The type III deiodinase is mainly involved with the degradation of T3 and the inactivation of

H2 CHE--'CH---COOH

If (thyroxine)

5"-Deiodinase/

(Type I and II)

~5-Deiodinase

/

HO' / _I . ~

.... y-c.

c

(Type III)

cooH

i/

i/

r~ (inactive)

T3 (active hormone) 5-Deiodinase (Type III)

COOH 3,3"-T2 (inactive) FIGURE 33-5 Activating and inactivatingpathwaysof T4.

776

CHAPTER 33

Endocrine Metabolism IV: Thyroid Gland

TABLE 33-2

The Iodothyronine Deiodinases Type I Reaction catalyzed Substrate preference Tissue distribution Affinity (Km) for T4 Effect of: Iopanoic acid* Amiodarone** Fetal life Propylthiouracil (PTU) Selenium deficiency Starvation Caloric restriction Glucocorticoids$ Excess glucocorticoids Nonthyroid illnesses High carbohydrate diet Hypothyroidism Hyperthyroidism Propranolol

5'-deiodination (outer ring) rT3 > T4 > T3 Liver, kidney, thyroid, et al. Low (--1 gM) Decrease Decrease Decrease Decrease Decrease Decrease Decrease Increase Decrease Decrease Increase Decrease Increase Decrease

Type II

Type III

5'-deiodination (outer ring) 5-deiodination (inner ring) T4 > rT3 T3 > T4 Brain, pituitary, skin, brown fat Liver, kidney, et al. High (-~1 nM) High (-40 nM) Decrease Decrease Decrease None None None None None None None None Increase Decrease None ?

Decrease Decrease ? None

Decrease Increase ?

*Iopanoic acid (3-amino-a-ethyl-2,4,6-triiodo-hydrocinnamicacid) is a radiopaque iodine compound used as a contrast medium in cholecystography. **Amiodarone (2-butyl-3-benzofuranyl) [4- [2- (diethylamino)ethoxyl]-3,5-diiodo-phenyl] methanone) is a coronary vasodilator used in the control of ventricular arrhythmias, and in the managementof angina pectoris. ;The physiologicrise in cortisol levels at about the time of birth stimulates 5'-deiodinase and causes a rise in T3 and fall in rT3. The effect of physiologic levels of cortisol on 5'-deiodinase in adults is unclear.

T4, both involving the removal of the inner ring 5-iodide. The type III enzyme is widely distributed among tissues and is responsible for the formation of almost all (95%) of the circulating rT3. Its activity is not altered by most of the factors or conditions that inhibit the activity of types I or II deiodinases, and this explains why there is a reciprocal relationship between the plasma levels of T3 and rT3. Not all metabolic processing of iodothyronines involves deiodination, although it is the major metabolic route for T4, T3, and rT3, accounting for about 80% of the fate of each compound. The remaining 20% is conjugated in the liver with sulfate or glucuronide, and/or processed by oxidative deamination and decarboxylation to form thyroacetic acid derivatives with minimal biological activity (Figure 33-6). Inorganic iodide liberated from deiodinations enter the extracellular compartment and is excreted in the urine (~488 #g/d) or stool (~12 #g/d via bile), or is reabsorbed by the thyroid gland (~108 #g/d) for resynthesis of thyroid hormones. At the average daily intake of ~500 #g inorganic iodide, this amounts to no net change in the iodide status, although the small amount of iodide that

is lost through the skin and gastrointestinal tract would place the body in a negative iodide balance unless intake is adjusted accordingly.

33.4 Biological Actions of Thyroid Hormones M e c h a n i s m of Action and Latency Period All but a few of the thyroid effects that have been identified occur at the level of gene transcription, mediated by nuclear thyroid hormone receptors (Chapter 30). These effects have a longer latency period than for most steroids; some of the relatively early responses show a latency period of several hours.

Physiological Effects

Cardiovascular System Thyroid hormone enhances cardiac contractility and exerts a positive chronotropic effect on the heart, increasing heart rate by a mechanism that may involve more than a potentiation of the fi-adrenergic effect. In the

SECTION 33.4

Biological Actions of Thyroid Hormones

Hydrolysis of conjugate Intestine*---- Bile :

777

= (Enterophepatic circulation)

Glucumnide/SO~=- ~ conjugation ~ " (liver)

TRIAC o (+ active)

~20%

5-Deiodination

To ~

Deaminationdecarboxylation

3"-T, ~ (inactive)

Liberated I -

L

3,3"-% (inactive)

- Thyroids or urine

Stool

Hydrolysis ----* (Enterophepatic of conjugate circulation) Intestine ~

Bile ~

Glucuromde/SO4 conjugation

(liver)

/

.,.

~

~20%

IT J

~80%

i

Deamination-

~50Olo

decarboxylation

5"-Deiodination

-30o/o 5"-Deiodination

L

TETRAC : - - - ' TRIAC (_+active) "'-., rTRIAC

(inactive)~ Undergoes rapid /

5"deiodinatio~)

To" Stool

3"-T~ (inactive)

3,3"-% " (inactive)

(active) ,~, [T3 P~176

IT, contributes to the % pool]

FIGURE 33-6 Metabolism of T3 and T4. To, Thyronine; 3,3'-T2, 3,3~-diiodothyronine; 3~-T, 3'-iodothyronine; TETRAC, 3,T, 5,5~-tetraiodothyroacetic acid; TRIAC, 3,3~,5-triiodothyroacetic acid.

ventricular myocardium, thyroid hormone stimulates the synthesis of myosin heavy chain o~(MHCot), while inhibiting the synthesis of MHC/3, by regulating the expression of the respective genes. The result is that myocardium with a higher MHCo~ content has higher calcium and actin-activated ATPase activities and increased velocity of muscle fiber shortening. Thus, the thyroid hormoneconditioned ventricle is capable of increased contractile performance. Intermediary Metabolism

Thyroid hormone increases both lipolysis and lipogenesis, although lipogenesis is stimulated before lipolysis, due to early induction of malic enzyme (malate dehydrogenase), glucose-6-phosphate dehydrogenase, and fatty acid synthase. Thyroid hormone lowers serum cholesterol levels by maintaining low-density lipoprotein (LDL) receptor density, by maintaining hepatic lipase activity which catalyzes conversion of intermediate density lipoproteins

(IDL) to LDL, and probably also by maintaining lipoprotein lipase activity which promotes triglyceride clearance. In liver, kidney, skeletal muscle, cardiac muscle, and adipose tissue, thyroid hormone stimulates Na+,K+-ATPase gene expression and promotes thermogenesis. Growth and Maturation

Thyroid hormone stimulates production of IGF-I directly (liver) and indirectly (via increased growth hormone, GH). In adults with hypothyroidism, basal serum GH levels are normal, but the GH responses to provocative stimuli, nocturnal GH secretion, and serum IGF-I are all subnormal. In cretinism (infantile hypothyroidism), linear growth is severely retarded and the resultant dwarfism is characterized by a retention of the high upper to lower body ratio of infancy. The growth retardation in persons with cretinism is primarily due to delayed appearance of ossification centers in long bone, and secondarily to a deficiency in growth factors.

CHAPTER33 Endocrine Metabolism IV: Thyroid Gland

~8

A deficiency of thyroid hormone during fetal development due to untreated or undertreated maternal hypothyroidism results in a neurological deficit in the offspring. In congenital hypothyroidism, a normal maternal thyroid can meet the fetal requirements for thyroid hormone. However, in the postnatal period these infants require a prompt thyroid hormone replacement therapy throughout their life, beginning in the first few weeks of life. If untreated, they inevitably develop growth and mental retardation. Thyroid hormone is essential for maturational development of the CNS and is required for the development of axonal projections and myelination. One of the important effects of thyroid hormone is to promote the synthesis of myelin basic protein. Congenital hypothyroidism in Western populations occurs in about 1 in 4000 births. Congenital hypothyroidism is caused by embryogenic defects leading to thyroid agenesis (50-55%), dysgenesis (30-35%), errors in the synthesis of thyroid hormone ( 10-15 %), and defects in the pituitary-hypothalamic axis (4%). Neonatal screening for congenital hypothyroidism is performed using blood collected on filter paper from newborns older than 12 hours. The laboratory test consists of measuring T4, TSH, or both. Abnormal values must be reconfirmed and additional tests include other parameters (e.g., TBG).

Reproductive System Thyroid hormone increases total plasma androgen levels by increasing the production of testosterone-binding globulin (TeBG) by the liver. Hyperthyroidism is associated with increased plasma TeBG levels, higher total testosterone levels, but normal free testosterone levels in adult men. There is a high incidence of gynecomastia in hyperthyroid men (40-83%) due to a higher circulating level of free estradiol in plasma. The elevated free-estradiol level may be explained by the lower affinity of estrogen for TeBG and also by increased conversion of androgen to estrogen in hyperthyroidism. In prepubertal boys, but not in older boys or adult men, hypothyroidism is associated with a high incidence of increased testicular growth (macroorchidism) that is not accompanied by any change in testosterone levels. Elevated follicle-stimulating hormone levels are found in most cases, but there is no testicular maturation. The mechanism of this disorder is not known.

Pharmacological Effects The effects of thyroid hormone may persist for up to a week because of its long half-life in the circulation, its long-acting effect on the genome of the target cell, and the slow rate of recovery of some target tissues (e.g., brain).

Thyroid hormone excess depletes body energy stores and imparts hypersensitivity of tissues to catecholamines. Protein synthesis is inhibited, protein catabolism is accelerated, and the antianabolic actions of cortisol are enhanced. During the period of growth, these factors would lead to growth suppression, muscle weakness, weight loss, and depletion of liver and muscle glycogen stores. Gluconeogenesis is fueled by elevated substrate levels (amino acids, glycerol, lactate, and pyruvate), and this contributes to the blood glucose pool; however, since peripheral glucose utilization is also increased, hyperglycemia is not severe. Thyroid excess also leads to depletion of triacylglycerol stores, mainly due to its action on catecholamine-induced lipolysis. Tissue consumption of free fatty acids and ketones is also increased. Thus, serum lipid levels fall, including the level of serum cholesterol. In essence, the storage forms of glucose, amino acids, and fatty acids are depleted, and these substrates are rapidly metabolized, resulting in the need for increased food intake. Because thyroid hormone increases tissue responsiveness to catecholamines, many symptoms of thyroid hormone excess are those that characterize catecholamine excess. Effects of T3 on cardiovascular hemodynamics consist of increased tissue thermogenesis, decreased systemic vascular resistance, decreased effective arterial filling volume, increased renal sodium reabsorption, increased blood volume, increased cardiac inotropy and chronotropy leading to increased cardiac output. Behavioral changes (such as nervousness, restlessness, short attention span, and emotional lability) are common, some of which require a longer period for decay than do the metabolic changes following normalization of thyroid hormone levels. Clearly, the central actions of thyroid hormone involve more than potentiation of adrenergic neurotransmission. Thyroid disorders. Disturbances in thyroid metabolism can occur at any level of the hypothalams-pituitarythyroid-peripheral tissue axis. Several of these disorders have been discussed previously. Hyperthyroidism is more prevalent in women than men. The three most common causes of hyperthyroidism are Graves' hyperthyroidism, toxic multinodular goiter, and toxic adenoma. The clinical features of hyperthyroidism include hyperkinesis, weight loss, cardiac anomalies (e.g., atrial fibrillation), fatigue, weakness, sweating, palpitations, and nervousness. The typical biochemical laboratory parameters are increased serum free T4 and decreased serum TSH. The most common cause of hypothyroidism is failure of the thyroid gland; this is known as primary hypothyroidism. In adults, the cause of primary hypothyroidism is often spontaneous autoimmune disease (e.g., Hashimoto's thyroiditis) or destructive therapy for hyperthyroid states

Supplemental Readings and References

(e.g., Graves' disease). In adults, hypothyroidism has an insidious onset with a broad range of symptoms. The typical biochemical laboratory parameters include the measurement of serum free T4 and TSH, which are decreased and increased, respectively. In addition, measurement of serum autoantibodies for thyroglobulin and thyroperoxidase is also utilized.

Supplemental Readings and References D. L. Bodenner and R. W. Lash: Thyroid disease mediated by molecular defects in cell surface and nuclear receptors. American Journal of Medicine 105, 524 (1998). S. C. Boyages: Clinical review 49: Iodine deficiency disorders. Journal of Clinical Endocrinology and Metabolism 77, 587 (1993). G. A. Brent: The molecular basis of thyroid hormone action. New England Journal of Medicine 331, 847 (1994). H. B. Burch and L. Wartofsky: Graves' ophthalmopathy: current concepts regarding pathogenesis and management. Endocrine Review 14, 747 (1993). G. N. Burrow: Thyroid function and hyperfunction during gestation. Endocrine Review 14, 194 (1993). G. N. Burrow, D. A. Fisher, and P. R. Larsen: Mechanisms of disease: maternal and fetal thyroid function. New England Journal of Medicine 331, 1072 (1994). I. J. Chopra: Clinical review 86: Euthyroid sick syndrome: is it a misnomer? Journal of Clinical Endocrinology and Metabolism 82, 329 (1997). D. Glinoer: The regulation of thyroid function in pregnancy: pathways of endocrine adaptation from physiology to pathology. Endocrine Review 18, 404 (1997). M. A. Lazar: Thyroid hormone receptors: multiple forms, multiple possibilities. Endocrine Review 14, 184 (1993). J. H. Lazarus: Hyperthyroidism. Lancet 349, 339 (1997). R. S. Lindsay and A. D. Toft: Hypothyroidism. Lancet 349, 413 (1997). S. M. McLachlan and B. Rapoport: The molecular biology of thyroid peroxidase: cloning, expression and role as autoantigen in autoimmune thyroid disease. Endocrine Review 13, 192 (1992).

779 G. Medeiros-Neto, H. M. Targovnik, and G. Vassart: Defective thyroglobulin synthesis and secretion causing goiter and hypothyroidism. Endocrine Review 14, 165 (1993). A. J. Mixson, R. Parrilla, S. C. Ransom, et al.: Correlations of language abnormalities with localization of mutations in the B-thyroid hormone receptor in 13 kindreds with generalized resistance to thyroid hormone: identification of four new mutations. Journal of Clinical Endocrinology and Metabolism 75, 1039 (1992). J. C. Morris: The clinical expression of thyrotropin receptor mutations. Endocrinologist 8, 195 (1998). R. Paschke and M. Ludgate: Mechanism of disease: the thyrotropin receptor in thyroid disease. New England Journal of Medicine 337, 1675 (1997). S. P. Porterfield and C. E. Hendrich: The role of thyroid hormones in prenatal and neonatal neurological development--current perspectives. Endocrine Review 14, 94 (1993). S. Refetoff, R. E. Weiss, and S. J. Usala: The syndromes of resistance to thyroid hormone. Endocrine Review 14, 348 (1993). E. Roti, R. Minelli, E. Gardini, and L. E. Braverman: The use and misuse of thyroid hormones. Endocrine Review 14, 401 (1993). V. Singh and J. P. Catlett: Hematologic manifestations of thyroid disease. Endocrinologist 8, 87 (1998). M. I. Surks and R. Sievert: Drug therapy: drugs and thyroid function. New England Journal of Medicine 333, 1688 (1995). A. D. Toft: Thyroxine therapy. New England Journal of Medicine 331, 174 (1994). Y. Tomer and T. E Davies: Infection, thyroid disease, and autoimmunity. Endocrine Review 14, 107 (1993). J. I. Totrtns and H. B. Burch: Serum thyroglobulin measurement: utility in clinical practice. Endocrinologist 6, 125 (1996). G. Vassart and J. E. Dumont: The thyrotropin receptor and the regulation of thyrocyte function and growth. Endocrine Review 13, 596 (1992). A. De La Vieja, O. Dohan, O. Levy, et al.: Molecular analysis of the sodium/iodide symporter: impact on thyroid and extrathyroid pathophysiology. Physiological Reviews 80, 1083 (2000). A. P. Weetman and A. M. McGregor: Autoimmune thyroid disease: further developments in our understanding. Endocrine Review 15, 788 (1994). I. Klein and K. Ojamaa: Thyroid Hormone and the Cardiovascular System. New England Journal of Medicine 344, 501 (2001).

CHAPTER

34

Endocrine Metabolism V: Reproductive System

The sex of an individual is the outcome of genetic determinants (genotypic sex) and of hormonal effects that confer structural features characteristic of a given sex (phenotypic sex). Both genetic and hormonal determinants normally operate only at two phases of life: during fetal development and at puberty. Genotypic sex is either homogametic (XX) or heterogametic (XY). In humans and other mammals, the homogametic (XX) pairing programs ovarian development and oocyte formation (oogenesis), while the heterogametic (XY) pairing leads to testicular development and spermatogenesis. In both genotypes, the embryonal gonads develop from the epithelium and stroma of the urogenital ridge, a thickening of the coelomic (ventromedial) aspect of the mesonephros that emerges at about the second or third week of pregnancy. Both the epithelium and stroma of the urogenital ridge are derived from the intermediate mesoderm of the embryo; however, invading this structure at about week 4 are primordial germ cells from the yolk sac, which take residence in association with the epithelial cells of the developing gonad and replicate. During the germ cell invasion, the epithelial cells of the gonads undergo proliferation and begin entering the stromal spaces as cord-like projections, called "primary sex cords." Until about week 6, the gonads are undifferentiated and uncommitted; that is, the structure has the potential to develop into either ovaries or testes. A list of expanded acronyms appears as Appendix VIII.

The fetal gonads develop in parallel with the Wolffian (mesonephric) duct and the Mtillerian (paramesonephric) duct, both of which are positioned bilaterally to the gonads within the urogenital ridge. The Wolffian duct begins forming from about week 4, starting at the mesonephros and terminating caudally at the cloaca. The Mtillerian duct begins forming from about week 6 and merges caudally with the urogenital sinus. The Wolffian duct is the primordium of the male reproductive tract, whereas the Mtillerian duct is the primordium of the female reproductive tract. Until about week 8, both ducts are present and undifferentiated. In the absence of a Y chromosome, a gene on the X chromosome directs the development of fetal gonads into ovaries. This critical sex-determining gene located on the X chromosome is known as the DAX-1 gene. Subsequent to the action of this gene, the Mtillerian ducts develop into a female genital tract and the Wolffian duct regresses. In the presence of a Y-chromosome, the SRY gene (sexdetermining region of the Y-chromosome) antagonizes the action of the DAX-1 gene (the SRY gene was previously known as testis-determining factor, TDF). Mutations in the SRY gene can result in an XY female with gonadal dysgenesis. Testis differentiation may also require the participation of a gene located on chromosome 17 (SOX9 gene). The precise interaction between SOX9 and SRY in gonadal development is not yet understood; however, both genes code for DNA-binding proteins. Mutations in the SOX9 gene can result in sex reversal in XY individuals

781

782

with camptomelic dysplasia. The latter is a severe form of skeletal dysplasia associated with dysmorphic features and cardiac defects. After the fetal gonads are converted into testes by SRY, testosterone and Mtillerian-inhibiting substance (MIS) control subsequent sexual differentiation. MIS causes the regression of the Mtillerian duct, which would otherwise develop into female genital tracts. Testosterone promotes the development of the Wolffian duct into male internal genitalia, including epididymis, vas deferens, and seminal vesicles. Testosterone also causes differentiation of the urogenital sinus into the prostate gland and masculinizes the external genitalia; however, these tissues must first convert testosterone into dihydrotestosterone (DHT), a reaction catalyzed by cytoplasmic 5ot-reductase type 2. The effects of testosterone on the Wolffian duct and urogenital sinus are dependent on the presence of androgen receptors and 5ot-reductase, which are expressed by genes on the X chromosome and chromosome 2, respectively, and are present in both genotypic sexes. This explains why exposure of the fetus to androgens during the critical period may lead to masculinization of the internal and external genitalia of genotypic female fetuses. It also explains why lack of expression of either gene can lead to feminization in genotypic male fetuses. The gonad has two interrelated functions: gametogenesis and hormone production. Gametogenesis depends on gonadal hormone production, which is influenced by gametogenesis. Both processes are controlled by luteinizing hormone (LH) and follical-stimulating hormone (FSH), which are released in response to hypothalmic gonadotropin-releasing hormone (GnRH) and to the levels of circulating gonadal hormones. In addition, gametogenesis is regulated by paracrine actions of the gonadal hormones. The obvious difference between the sexes in gametogenesis is formation of ova (ootids) in the female and of sperm (spermatozoa) in the male. Spermatogenesis becomes operational from about the time of puberty and continues throughout life. The process takes about 74 days. Oogenesis occurs in three phases. The initial phase involves proliferation of the stem cells (oogonia) and occurs only in fetal life. The second phase involves the first maturational division (formation of the secondary oocyte) and occurs about the time of ovulation (see below). The third phase involves the second and final maturational division (formation of ova) and occurs at fertilization. Because the number of oogonia is determined before birth, the number of fertilizable ova that can be produced in a woman's lifetime is limited and is greatly reduced by normal degenerative processes (atresia), so that of the estimated seven million oogonia in the fetal ovaries,

CHAPTER 34

Endocrine Metabolism V: Reproductive System

only about 300-500 ova ultimately develop. Unlike the continuous generation of sperm in the testes, the ovaries generally produce only one secondary oocyte every 22-28 days. The fertile lifetime in the average woman is about 35-40 years. Ovaries and testes produce identical steroid hormones, but the amounts and their patterns of secretion are different (Table 34-1). Although testosterone is present in both males and females, its level in the male is about 18 times that in the female; conversely, circulating levels of estradiol in the female are about 3-15 times those in the male. In either sex, the major sex steroid originates in the gonads, whereas in the opposite sex, this steroid is generated in sustantial amounts by the adrenal cortex or by peripheral conversion of another steroid. For example, almost all of the circulating estradiol in the female comes from the ovaries, whereas in the male, only about one-third comes from the tests, the rest being generated by peripheral conversion of androgenic precursors. Finally, the pattern of gonadal steroid secretion is different. In the male, the secretion of gonadal steroids is fairly constant, although minor fluctuations occur as a result of circadian rhythms. In the adult female, gonadal steroid secretion undergoes dramatic, cyclic changes at about monthly intervals. These cyclic changes, which are dictated by processes regulating oogenesis, are referred to as the menstrual cycle.

34.1 Testes Regulation of Spermatogenesis: Sertoli-Neuroendocrine Axis Sertoli cells are epithelial cells that line the seminiferous tubules of the testes. At their basal aspects, these cells form the basement membrane and tight junctions that make up the highly selective "blood-testes barrier," which normally prevents entry of immune cells into the lumen. Their function is to provide nutritional and hormonal support for cells undergoing spermatogenic transformation (i.e., spermatocytes and spermatids). For this reason, they are often referred to as "nurse cells." Sertoli cells require FSH stimulation for their maintenance and specifically for production of androgen-binding protein (ABP). ABE which has a high affinity for testosterone, maintains high levels of testosterone in the seminiferous tubules and thereby helps maintain spermatogenesis. FSH also stimulates aromatase activity in the Sertoli cell, thus stimulating conversion of testosterone into estradiol. This FSH-sensitive Sertoli cell activity accounts for most of the testicular output of estradiol. FSH secretion by the anterior pituitary is held in check by negative feedback from the testes. This feedback is

|

~q

=1

oil

oll

oil

illil

=I oil

I=

c~

oil

lil

m

oml

ill

I=

.,e

E~

oil

.
.,

~ ~

~

~~

[-...-

.

~

-~

~

E

t'.-

-

[-~

~'~

0~

_ ~

~1:) ~ ,.~

..:o

m

oo~od

. -

~

~

-

"~GZ

.~

o

~

~ ""

o~

,-

~,

-=,-

~

9

o

r

=

.,-..~

".,~,,~

r

0

r~

o~,,i

9

c~

c~

~;o,d

r~ 0

9

9~,.-i

o~.,~

"7 .1

9

~2z


p-.

0 0

9

E E

9

E =

.,i.~1

Z

C"-

0 0

E

9.~

oil

9~.-.i

.,.~

~

0

U:

< Z

Z

m

:::

9

"~

~

.~

Z


r,.,)

LT..,

o~~

0

9

9

i

0

>, ~

..>

Z


-

"o .,.

;> ;>

Z




,e,--(

0 I

.,-

0

-.

I

Z




o

I

og

X

843

844

,..,~

..~

e,l

~

r~

o~

~

z


o

Z

.~ .~

9

Z


0

r~

om

=~.

..~

F

9

o~

Z


~ ~ ~ ~.= 0~.._,~

9

~'~ ~

o~..~

c~

@

r~

Z

~:.=-

=t

r,~

m

r

~o

9

9

9

r.~

0

=

=

~,~

,~.-

m

..,_, > ~ = ~_~

9~ ~'~

~

o~,.i > 0

~'~

"~~

9

-

r~ r~

9 0

0

~:~

"'-'

",.._~

8

",,.~

0

>

~9

~

0

~~-.=

~

~

~ ~~~.~

~.~ ,~

,...~ 9

~ ~.~ c~

0




-=~

o

do

.,--,

2~

9

0

~~ ~.-

~

0

"~

~.~ 9-= E~

o~.~

0

0 0

9

,~ 9 .~

"t::

~

9

~

~.~ "~

r.~

2~

9

0

"~

~

I'~,.,~

~',~

9 >

.~,-q

< 9

~~ o

. .....~

,.c:

.,=

o

~ ~,-

~,,_,

r~

=.~

i

g-~

o ~'~

~'~ 0


60 #g/dL). Lead inhibits heme biosynthesis (Chapter 29). Severe lead poisoning can cause lead encephalopathy. Aluminum Aluminum accumulates in the bones of patients undergoing long-term hemodialysis. It is strongly implicated as the cause of dialysis encephalopathy ("dialysis dementia"), and it produces an osteodystrophy that responds poorly to vitamin D. Use of deionized water to prepare the dialysates has largely eradicated these problems. Deferoxamine, used to treat hemochromatosis (Chapter 29), increases aluminum excretion in these patients by forming a water-soluble complex with aluminum that can be removed by dialysis. Chronic use of aluminumcontaining antacids to reduce intestinal phosphate absorption in uremic patients contributes to aluminum overload. An increase in serum aluminum concentration and in bone aluminum content, accompanied by severe osteomalacia, occurs in infants with azotemia who are given large doses of aluminum hydroxide to reduce phosphate absorption. Intestinal absorption of aluminum seems to be increased in hyperparathyroidism. The metal inhibits hexokinase,

CHAPTER37 Mineral Metabolism

ALA-dehydratase, and isocitrate dehydrogenase and decreases Na +, K+-ATPase activity, MgZ+-ATPase activity, and choline uptake into synaptosomes. In vitro, aluminum displaces magnesium from MgZ+-ATP complexes, and it could thus antagonize virtually any phosphatetransferring reaction that uses MgZ+-nucleotide triphosphate complexes. O t h e r Trace Elements Chronic exposure to silver produces a bluish skin discoloration known as argyria. Gold poisoning has resulted from the use of gold salts for treatment of arthritis. Poisoning from antimony, arsenic, bismuth, mercury, and thallium is well known.

37.4 Metallothioneins The metallothioneins are small proteins (M.W. ~7000), rich in sulfhydryl groups, that bind cadmium, zinc, and small amounts of copper, iron, mercury, and perhaps other heavy metals. Within a species, multiple metallothioneins occur that differ in amino acid composition. These proteins typically contain 20 cysteine residues but no disulfide bonds and no aromatic amino acids. They bind one atom of cadmium or zinc for each three sulfhydryl groups. Binding of copper to metallothionein is somewhat different, since a greater number of metal atoms are bound per protein molecule. Metallothioneins occur particularly in liver and kidney but also in the intestine and other tissues. Synthesis of the metallothioneins is increased by the metals that they bind, by dexamethasone, and by many other agents. Cadmium and zinc are potent inducers in the intestine and in cultured HeLa cells, while copper induces intestinal metallothionein only weakly. The metals and dexamethasone act by increasing transcription of metallothionein genes. Metal binding also increases the stability of the apoproteins; metals also cause greater accumulation of metallothionein than dexamethasone. The physiological role of metallothioneins remains unknown, but it may be, in part, to protect cells from metal toxicity by binding metal ions. Metallothioneins may also be important for intestinal and renal absorption of metals and for metal storage and excretion.

37.5 Essential Trace Elements Table 37-4 summarizes current knowledge of the essential trace elements. Bromine, cadmium, lead, and tin may also

SECTION 37.5

EssentialTrace Elements

be essential, but the evidence is not convincing. Many of these elements are toxic when present in excess, and cause changes in membrane permeability or inhibition of vital enzyme processes such as protein synthesis, oxidative phosphorylation, and DNA replication. Chronic exposure to these elements may cause other, poorly understood, disorders. Copper and zinc are discussed below because they have been most thoroughly studied and their involvement in human disease is well documented. Iron and iodine are discussed in Chapters 29 and 33, respectively; cobalt is discussed in Chapter 38.

Copper Copper is necessary, together with iron, for hematopoiesis, probably partly because it is needed for the synthesis of ferroxidase (ceruloplasmin). Many enzymes require copper for activity. Examples of some of the copper-enzymes and their functions are given in Table 37-5. Mitochondrial iron uptake may be blocked by deficiency of a cuproprotein, perhaps cytochrome oxidase. Several inherited diseases involving abnormalities in copper metabolism (Wilson's disease, Menkes' syndrome) or copper enzymes (X-linked cutis laxa, albinism) occur in human and in several animal species. The oxidation state of copper in biological systems is + 1 or +2. Copper(III) is found in inorganic systems and may occur as a reaction intermediate in galactose oxidase, laccase (a plant enzyme), and perhaps other enzymes. The coordination number of copper in these enzymes ranges from two to six and occasionally higher. The average adult human contains 70-100 mg of copper. The highest concentrations (in decreasing order) are in liver, brain, heart, and kidney. Muscle contains about 50%

895

of total body copper. Of the remainder, about one-fifth is in the liver (3-11 #g/g wet weight). Human erythrocytes contain 1.0-1.4 #g of copper per milliliter, of which more than 60% is in superoxide dismutase. Normal serum contains of copper and 200-400 mg/L of ceruloplasmin. Whether copper is released from ceruloplasmin by endocytosis or by conformational change following binding of ceruloplasmin to a membrane receptor remains unknown. Copper is absorbed from food in the upper small intestine. The absorption is primarily dependent on the quantity of the copper present in the diet. High intake of zinc diminishes copper absorption by inducing metallothionein formation in the mucosal cells. Metallothioneins, due to their high affinity for copper, bind it preferentially and the bound copper is lost during the sloughing of cells from the villi. Copper accumulation in patients with Wilson's disease can be reduced by giving oral zinc acetate, which decreases absorption (discussed later). Absorbed copper is transported to the portal blood where it is bound to albumin (and probably transcuprein), amino acids, and small peptides. Copper binds to albumin at the N-terminal tripeptide (Asp-Ala-His) site. The recently absorbed copper is taken up by the liver, which plays a central role in copper homeostasis. Copper occurs in many foods; particularly good sources are liver, kidney, shellfish, nuts, raisins, and dried legumes. Copper deficiency due to diet is rare except in malnutrition and in children with chronic diarrhea. It occurs in total parenteral nutrition with fluids low in copper, particularly following intestinal resection and in patients who receive large amounts of zinc to improve wound healing or for management of sickle cell anemia. Copper is often removed from prepared foods to increase their shelf life.

TABLE 37-5

Examples of Copper-Containing Enzymes and Their Functions

Enzymes 1. Cytochrome c oxidase 2. 3. 4. 5. 6.

Superoxide dismutase Ceruloplasmin Tyrosinase Dopamine/3-monooxygenase Lysyl oxidase (also called protein-lysine 6-oxidase)

7. Peptidylglycine mono-oxygenase

8. Amine oxidases

Functional Significance Terminal enzyme of mitrochondrial electron transport; oxidative phosphorylation. Inactivation of reactive oxygen species; antioxidant defense. Ferroxidase; Iron metabolism. Synthesis of melanin. Synthesis of norepinephrine and epinephrine. Required for cross-linking of collagen and elastin; maturation of collagen. Required for removal for carboxy terminal residue and a-amidation; Processing and maturation of neuroendocrine and gastro-intestinal peptide hormones. Deamination of primary amines.

896 Ascorbic acid, increased intestinal pH, sulfide ion, phytate (inositol hexaphosphate in plants), and some dietary proteins interfere with its intestinal absorption. Copper sulfate causes ulceration of the oral and esophageal mucosa, and an acute dose of 7-8 g is usually fatal. Copper intoxication has occurred when copper salts were used to treat extensive skin burns or when coppercontaining tubing or dialysis membranes were used for hemodialysis. Pulmonary fibrosis has been described in vineyard workers exposed for many years to fungicidal sprays (e.g., "Bordeaux mixture") containing copper sulfate. About 33-50 mg of copper per year (---100 #g/d) dissolves from copper-containing intrauterine contraceptive devices. Part of this copper is lost in menstrual flow, but part is rapidly absorbed. Whether this is harmful is not known but it seems unlikely. Penicillamine is the drug of choice for treatment of copper excess. The RDA for copper increases with age, ranging from 0.5-0.7 mg in infants up to 6 months of age to 2-3 mg in adults. The average adult diet contains 2.5-5 mg/d. Healthy, full-term infants have a relatively high hepatic copper content. The lower copper content of premature and malnourished infants is exacerbated by feeding with cow's milk, which contains less copper than human milk. About 40-70% of ingested copper is absorbed. Appearance in plasma of ingested copper with a peak occurring 1.5-2.5 hours after eating suggests that absorption begins in the stomach and proximal small intestine. Absorption may be inversely related to the metallothionein content of the mucosa. Although pinocytosis occurs in the intestines of human infants, their inability to absorb copper in Menkes' syndrome suggests that physiologically important absorption is by an active process, even at an early age. Absorption is enhanced by complex formation with L-amino acids and small peptides. Newly absorbed copper is taken up by the liver in a saturable transport process and secreted into plasma as ceruloplasmin (0.5-1.0 mg/d). Copper is excreted in bile in an amount roughly equal to daily absorption (--, 1.7 mg/d). Toxic levels in liver occur in primary biliary cirrhosis, Indian childhood cirrhosis (a familial, probably genetic, disease, limited to Asia), and other liver diseases in which bile flow is disrupted. Toxicity is rare unless there is preexisting liver disease or ingestion of large amounts of copper salts. B iliary copper is in a form that cannot be readily reabsorbed. Some loss also occurs in urine and sweat. Renal copper reabsorption may be important for copper homeostasis and may be regulated by insulin or glucagon. Approximately 0.5 mg of copper is lost during each menstrual period. During lactation, copper loss averages 0.4 mg/d. The biological functions of copper are mostly reflected in the proteins listed in Table 37-5. Like iron, copper is

CHAPTER37 Mineral Metabolism

intimately involved in adaptation to an aerobic environment. The proteins contain copper bound directly to specific side chains. Copper is also essential in plants, lower eukaryotic animals, and prokaryotes. Wilson's disease is an autosomal recessive disease of copper metabolism. It has a prevalence of 1 in 30,000 live births in most populations. The disease has a highly variable clinical presentation. It is characterized by impairment of biliary copper excretion, decreased incorporation of copper into ceruloplasmin, and accumulation of copper in the liver and, eventually, in the brain and other tissues. The biochemical findings include low serum ceruloplasmin, high urinary copper excretion, and high hepatic copper content. Some patients have normal serum ceruloplasmia levels, and heterozygous individuals do not consistently show reduced levels of this protein. The genetic defect in Wilson's disease resides in the long arm of chromosome 13 and the gene codes for a copper-transporting ATPase. Thus far, more than 40 different mutations have been found. The defect in the ATPase results in reduced biliary excretion of copper from the hepatocytes. The end result may lead to hepatic failure. The Cu-ATPase belongs to a family of ubiquitous proteins that are involved in the translocation or movement of cations such as H +, Na +, K +, Ca 2+, and other metal ions (discussed in the appropriate areas of the text). This family of ATPases is designated as P-type ATPase. The P designation stems from the fact that, during the catalytic cycle, an invariant aspartic acid residue undergoes phosphorylation by ATP and dephosphorylation by the phosphatase domain that results in a conformational change promoting cation transfer. Another common feature of P-type ATPase is that all possess an ATP binding domain at the carboxy terminus. Because intestinal copper absorption is unaffected, there is a net positive copper balance. Normal hepatic copper uptake ensures that copper accumulation occurs first in the liver. As the disease progresses, nonceruloplasmin serum copper increases, and copper deposits occur in various tissues, e.g., Descemet's membrane in the cornea (Kayser-Fleischer rings), basal ganglia (leading to lenticular degeneration), kidney, muscle, bone, and joints. Cultured fibroblasts from patients exhibit increased copper uptake. A disorder in Bedlington terriers that causes hepatic copper accumulation may serve as an animal model for Wilson's disease, although it differs from the human disorder in several ways. Penicillamine, a chelating agent, solubilizes copper and other heavy metals and promotes their excretion in urine, analogous to the use of deferoxamine in the treatment of iron overload (Chapter 29). Long-term treatment with penicillamine increases the requirement for pyridoxine. Iron supplements should also be given, on a schedule

SECTION 37.5

EssentialTrace Elements

that prevents their interference with penicillamine. Triethylenetetramine, another copper-chelating agent, may be of particular value in patients who are or who are becoming sensitive to penicillamine. As discussed earlier, since excessive zinc prevents copper absorption, oral intake of zinc acetate is used in the management of Wilson's disease. Menkes' syndrome (Menkes' steely-hair syndrome) is a rare, X-linked recessive disorder in which infants have low levels of copper in serum and in most tissues except kidney and intestine, where the concentration is very high. They also have greatly reduced plasma ceruloplasmin levels. Hair of the affected infants has a characteristic color and texture (pili torti, "twisted hair"). It appears tangled and dull, has an ivory or grayish color, and is friable. Weakness and depigmentation of hair and defects in arterial walls (leading to aneurysms) are explained by loss of activity of copper-dependent enzymes (Table 37-5). Cerebral dysfunction may be due to a disturbance in energy metabolism or neurotransmitter synthesis secondary to decreased activity of cytochrome oxidase and dopamine fl-hydroxylase. The defect in copper metabolism is generalized, and the degenerative changes cannot be reversed by parenteral copper administration. However, parenteral copper administration corrects the serum ceruloplasmin and hepatic copper deficiencies. Other tissues take up copper administered parenterally, but activities of their copper enzymes are not normalized. The failure of postnatal treatment is due to the fact that many of the deleterious effects occur in utero. The molecular defect in Menke's syndrome, like that in Wilson's disease, resides in a P-type ATPase. The gene for the enzyme is located on the X chromosome. Wilson's disease is characterized by defective biliary excretion; in Menkes' syndrome, the defect is a failure to transport copper to the fetus during development as well as failure to absorb copper from the gastrointestinal tract after birth. Aceruloplasminemia is an autosomal recessive disorder characterized by progressive neurodegeneration and accumulation of iron in the affected parenchymal tissues. Iron accumulation in this disorder is consistent with ceruloplasmin's role as a ferroxidase in iron metabolism (discussed in Chapter 29).

Zinc Zinc is a necessary nutrient in animals. The discovery that zinc deficiency is the cause of parakeratosis in pigs was the first demonstration of the practical importance of zinc in animal nutrition. More than 100 zinc enzymes are known, many of which are in the liver and include examples from all six classes of enzymes. In these enzymes, zinc may

897

have a regulatory role or be required for structure or for catalytic activity. Zinc is not readily oxidized or reduced from its usual oxidation state of -t-2 and is not involved in redox reactions. Table 37-6 lists some zinc-containing enzymes important for mammalian metabolism. Zinc has an important regulatory function in fructose-l,6-bisphosphatase and has structural catalytic and regulatory roles in DNA and RNA polymerases. Many of the transcription factors, namely, DNA-binding proteins, contain zinc finger motifs consisting of Zn 2+ bound to four cysteine or two cysteine and two histidine residues in a coordinate covalent linkage (Chapter 26). Thymidine kinase is also a zinc enzyme. Zinc is present in gustin, a salivary protein secreted by the parotid glands that may be necessary for proper development of taste buds. A common sign of zinc deficiency is hypogeusesthesia. Gustin is structurally similar to nerve growth factor (NGF) isolated from male mouse submaxillary glands and other sources, which contains 1 mol of zinc per mole of protein. The metal prevents autocatalytic activation of NGE Along with copper, zinc is required for activity of superoxide dismutase (Table 37-5). In pregnant rats, zinc deficiency adversely affects fetal development and parturition and produces fetal abnormalities perhaps because of a requirement of zinc for gene expression. It is involved in synthesis, storage, and secretion of insulin by pancreatic islet fl cells. Physiological stresses, such as trauma, infections, and low protein intake, cause a decrease in the plasma zinc level. Zinc can substitute for iron in protoporphyrin IX during heme synthesis, and quantitation of zinc protoporphyrin in erythrocytes is useful for evaluating iron deficiency anemia. Zinc interacts with plasma membranes of many cells, including erythrocytes (see below), and it may be vital for their structural integrity. The metal also has roles in wound healing, male fertility, bone formation, and cell-mediated immunity. A healthy 70-kg man contains 1.4-2.3 g (~ 30 mmol) of zinc. The prostate gland is rich in zinc (about 100 # g Zn 2+ per gram wet weight), as are prostatic secretions and semen. In rats, prostatic zinc concentration is decreased by castration and increased by administration of testosterone or gonadotropin. Testicular atrophy and failure of spermatogenesis occur in zinc-deficient rats. The skin contains approximately 20% of total body zinc. Dermatitis and poor wound healing are the most common symptoms of zinc deficiency in patients receiving total parenteral nutrition with zinc-deficient fluids. Zinc supplements do not promote wound healing when zinc stores are adequate. Bone has a high zinc content (> 100 #g/g); however, bone zinc is not readily mobilized. Hair and nails also have high concentrations of the metal.

898

CHAPTER37 Mineral Metabolism

TABLE 37-6

Some M a m m a l i a n Zinc Proteins

Source

Molecular Weight

Alcohol dehydrogenase

Human liver

87,000

Alkaline phosphatase

Human placenta

125,000

Leucine aminopeptidase

Porcine kidney

300,000

5'-AMP aminohydrolase Carbonic anhydrase B

Rat muscle Human erythrocyte

290,000 26,600

Carboxypeptidase A

Bovine pancreas

Glutamate dehydrogenase a2-Macroglobulin Malate dehydrogenase Metallothionein

Bovine Human Bovine Human

Rhodanese (sulfur transferase)

Bovine liver

Enzyme

liver serum heart renal cortex

Zn Atoms/ Protein Molecule

--3

Requires 2NAD+; two zinc atoms used for catalysis; function of other two not known but may stabilize enzyme. Unknown function; role of Z n 2+ unknown but needed for activity. N-terminal exopeptidase; Zn binds at two sites, one for catalysis, the other for regulation. Catalyzes reversible hydration of CO2; needed for CO 2 transport, buffeting of ECE gastric and renal acid secretion; Zn is catalytic. C-terminal exopeptidase; involved in digestion of proteins; Zn functions in catalysis as a Lewis acid.

34,500

1,000,000 840,000 40,000 10,500

2-4 3-8 1 3

37,000

2

The zinc content of human blood is 8.8 #g/mL, of which 80-90% is within erythrocytes, mostly in carbonic anhydrase. Zinc binds to hemoglobin, increasing its oxygen affinity. By binding to erythrocyte membranes, it increases the flexibility of irreversibly sickled cells in vitro but plasma levels high enough to be therapeutic cannot be achieved. Leukocytes and platelets also contain an appreciable amount of zinc. Plasma zinc levels vary with sex, age, time of day, geographic location, and time elapsed since the last meal prior to phlebotomy. The normal range for an adult is probably 70-95 #g/dL of plasma. Approximately 60% of zinc in plasma is bound to albumin, 30-40% to an ot2macroglobulin of unknown function, and a small amount to transferrin (Chapter 29) and amino acids, particularly cysteine and histidine. Copper does not compete with zinc for binding sites on albumin. Zinc newly absorbed from

Functions/Comments

i

Usually also contains 4-5 Cd; may serve as source of Zn. Metabolizes SCN-.

the intestine is albumin-bound. This zinc is the fraction that changes most in disease states and that correlates best with total serum zinc. The zinc bound to amino acids accounts for urinary excretion of zinc, and it may be utilized by some tissues. The RDA for zinc depends on age. For infants younger than 6 months, it is 3 mg; from 6 months to 1 year, 5 mg; for ages 1-10 years, 10 mg; and more than 10 years, 15 mg. For women, it increases during pregnancy (20 mg) and lactation (25 mg). A typical daily diet in the United States supplies 12-15 mg (about 200 #mob, of which 30-40% is absorbed. Dietary fiber binds and decreases the absorption of zinc. Human zinc deficiency was first reported in boys from Iran and Egypt who were small in stature, hypogonadal, and mildly anemic. The zinc content of their hair, plasma, and erythrocytes was below normal. Etiological factors included high dietary phytate content, loss

899

Supplemental Readings and References

of zinc through sweating, and geophagia (the practice of eating earth and clay, which bind zinc and other metals). All showed clinical improvement following oral supplementation with zinc. The jejunum is the principal absorptive region, although zinc absorption occurs through out the small intestine. Uptake occurs both by an active transport process, the activity of which varies inversely with zinc status, and by a passive process. The increase in serum zinc following a single oral or parenteral bolus of zinc decreases absorption within about 6 hours. In contrast, with slow intravenous infusion, significant decrease in zinc absorption may not occur for 4 days. Intestinal metallothionein is induced by and binds zinc. It increases by about 6 hours following a bolus of zinc and may block absorption by sequestering the metal in the mucosal cells. The evidence is largely circumstantial. Uptake by the liver is by an active process. Fecal zinc is derived from unabsorbed dietary zinc and zinc in desquamated epithelial cells and gastrointestinal secretions. Active intestinal secretion of zinc decreases in zinc-depleted animals and may be a regulated step in zinc homeostasis. Excretion of zinc in urine (0.3-0.6 mg/d) is independent of dietary intake. It is increased in renal disease, in alcoholic cirrhosis, in hepatic porphyria, following major operations, in severely burned patients, and in response to treatment with ethylenediaminetetraacetic acid. The normal concentration of zinc in sweat is 1.15 #g/mL, decreasing to about 0.6 #g/mL in zinc deficiency. In tropical climates, zinc loss from this source may reach 4 mg/d. Menstrual zinc loss is about 15/~g/d. The zinc content of human milk is highest in colostrum. The average zinc concentration in human milk is approximately 1.5 #g/mL. Fresh cow's milk averages 4 #g/mL, but human milk is a better source of zinc for infant nutrition because of lower bioavailability of zinc in cow's milk. Zinc is relatively nontoxic but, if present in the diet at high concentration and in readily available form, it can interfere with the absorption of calcium, copper, iron, and cadmium and can produce anemia. These elements, in turn, can decrease zinc absorption if present in sufficient quantities. Use of zinc salt lozenges has been claimed to reduce the duration of the common cold. However, many randomized, double-bind, placebo-controlled trials have failed to show any beneficial effects of zinc lozenges in reducing the duration of upper respiratory tract viral infections. Acrodermatitis enteropathica (AE) is an autosomal recessive disorder characterized by defective absorption of zinc, thickened, ulcerated skin on the extremities and around body orifices, alopecia, diarrhea, growth failure, and abnormalities in immune function. The concentration

of zinc in serum and hair is reduced. AE, and a similar disease of cattle known as adema disease or lethal trait A-46, respond to large amounts of oral zinc. Malabsorption of zinc occurs in both the human and animal diseases. Human milk seems to alleviate AE, presumably because it provides zinc in a form that can be readily absorbed.

Supplemental Readings and References J. S. Adams and G. Lee: Gains in bone mineral density with resolution of vitamin D intoxication. Annals of Internal Medicine 127, 203 (1997). D. G. Barceloux: Manganese. Clinical Toxicology 37, 293 91999). D. G. Barceloux: Zinc. Clinical Toxicology 37, 279 (1999). D. Beshgetoor and M. Hambidge: Clinical conditions altering copper metabolism in humans. Clinical Nutrition 67, 1017 S (1998). M. J. Bingham, T-J. Ong, K. H. Summer, et al.: Physiologic function of the Wilson disease gene product, ATP7B. Clinical Nutrition 67, 982S (1998). A. L. Boskey: Biomineralization: conflicts, challenges, and opportunities. Journal of Cellular Biochemistry Supplements 30, 83 (1998). E Bronner: Calcium absorption--a paradigm for mineral absorption. Journal of Nutrition 128, 917 (1998). F. Bronner and D. Pansu: Nutritional aspects of calcium absorption. Journal of Nutrition 129, 9 (1999). D. A. Bushinsky and R. D. Monk: Calcium. Lancet 352, 306 (1998). S. Chan, B. Gerson, and S. Subramaniam: The role of copper molybdenum, selenium, and zinc in nutrition and health. Toxicology 18, 673 (1998). T. W. Clarkson: Mercury--an element of mystery. New England Journal of Medicine 323, 1137 (1990). A. Cordano: Clinical manifestations of nutritional copper deficiency in infants and children. Clinical Nutrition 67, 1012S (1998). H. E DeLuca and C. Zierold: Mechanisms and functions of vitamin D. Nutrition Reviews 56, $4 (1998). J. S. Duthie, H. E Solanki, M. Knshnamurthy, et al.: Milk-alkali syndrome with metastatic calcification. American Journal of Medicine 99, 102 (1995). R. Eastell: Treatment of postmenopausal osteoporosis. New England Journal of Medicine 338, 736 (1998). K. E. Ensrud, D. M. Black, L. Palermo, et al.: Treatment with alendronate prevents fractures in women at highest risk. Archives of Internal Medicine 157, 2617 (1997). E. Evron, S. Goland, J. von der Walde, et al.: Idiopathic calcitriol-induced hypercalcemia. Archives of Internal Medicine 157, 2142 (1997). D. Feldman: Vitamin D, parathyroid hormone, and calcium: a complex regulatory network. American Journal of Medicine 107, 637 (1999). E Garnero, W. J. Shih, E. Gineyts, et al.: Comparison of new biochemical markers of bone turnover late postmenopausal osteoporotic women in response to alendronate treatment. Journal of Clinical Endocrinology and Metabolism 79, 1693 (1994). E H. Gloneux, N. J. Bishop, H. Plotkin, et al.: Cyclic administration of pamidronate in children with severe osteogenesis imperfecta. New England Journal of Medicine 339, 986 (1998). G. A. Greendale, N. E Lee, and E. R. Arriola: The Menopause. Lancet 353, 571 (1999). J. Guardiola, X. Xiol, R. Sallie, et al.: Influence of the vitamin D receptor gene polymorphism on bone loss in men after liver transplantation. Annals oflnternal Medicine 131, 752 (1999). J. G. Hall: A bone is not a bone is not a bone. Journal of Pediatrics 133, 5 (1998). J. R. Hansford and L. M. Mulligan: Multiple endocrine neoplasia type 2 and RET: from neoplasia to neurogenesis. Journal of Medical Genetics 37, 817 (2000).

900 Z. L. Harris, L. W. J. Klomp, and J. D. Gitlin: Aceruloplasminemia: an inherited neurodegenerative disease with impairment of iron homeostasis. Clinical Nutrition 67, 972S (1998). R. E Heaney: Bone mass, bone loss and osteoporosis prophylaxis. Annals oflnternal Medicine 128, 313 (1998). J. L. Jackson, C. Peterson, and E. Lesho: A meta-analysis of zinc salts lozenges and the common cold. Archives of Internal Medicine 157, 2373 (1997). H. M. Kronenberg, B. Lanske, C. S. Kovacs, et al.: Functional analysis of the PTH/PTHrP network of ligands and receptors. Recent Progress in Hormone Research 53, 283 (1998). L. Kuritzky, G. P. N. Samraj, and D. M. Quillen: Improving management of type 2 diabetes mellitus: 6. Chromium. Hospital Practice 35, 113 (2000). M. C. Linder, L. Wooten, E Cerveza, et al.: Copper transport. Clinical Nutrition 67, 9655 (1998). E J. Malloy and D. Feldman: Vitamin D resistance. American Journal of Medicine 106, 355 (1999). R. Marcus: Exercise and the regulation of bone mass. Archives oflnternal Medicine 149, 2170 (1989). E. Mariani, G. Ravaglia, A. Meneghetti, et al.: Natural immunity and bone remodelling hormones in the elderly. Mechanisms of Ageing and Development 102, 279 (1998). M. M. Maricic: Early prevention vs. late treatment for osteoporosis. Archives oflnternal Medicine 157, 2545 (1997). J. C. Marini: Osteogenesis imperfecta--managing brittle bones. New England Journal of Medicine 339, 986 (1998). L. A. Martini: Magnesium supplementation and bone turnover. Nutrition Reviews 57, 227. M. McClung, B. Clemmesen, and A. Daifotis: Alendronate prevents postmenopausal bone loss in women without osteoporosis. Annals oflnternal Medicine 128, 253 (1998). C. R. Meier, R. G. Schlienger, M. E. Kraenzlin, et al.: HMG-CoA reductase inhibitors and the risk of fractures. Journal of the American Medical Association 283, 3205 (2000). W. G. Meijer, E. Van Der Veer, and P. H. B. Willemse: Biochemical parameters of bone metabolism in bone metastases of solid tumors. Oncology Reports 5, 5 (1998). J. E B. Mercer: Menkes syndrome and animal models. Clinical Nutrition 67, 1022S (1998). B. A. Michel, D. A. Bloch, and J. E Fries: Weight-bearing exercise, overexercise, and lumbar bone density over age 50 years. Archives of Internal Medicine 149, 2325 (1989). W. L. Miller and A. A. Portale: Genetic disorders of vitamin D biosynthesis. Pediatric Endocrinology 28, 825 (1999). E D. Miller, N. B. Watts, A. A. Licata, et al.: Cyclical etidronate in the treatment of postmenopausal osteoporosis: efficacy and safety after seven years. American Journal of Medicine 103, 469 (1997). G. Mundy, R. Garrett, and S. Harris: Stimulation of bone formation in vitro and in rodents by statins. Science 286, 1946 (1999). G. R. Mundy and T. A. Guise: Hypercalcemia of malignancy. American Journal of Medicine 103, 134 (1997). G. R. Mundy and T. A. Guise: Hormonal control of calcium homeostasis. Clinical Chemistry 45, 1347 (1999). K. M. Prestwood, C. C. Pilbeam, J. A. Burleson, et al.: The short term effects of conjugated estrogen on bone turnover in older women. Journal of Clinical Endocrinology and Metabolism 79, 366 (1994). L. G. Raisz: The osteoporosis revolution. Annals oflnternal Medicine 126, 459 (1997).

CHAPTER37 Mineral Metabolism L. G. Raisz: Physiology and pathophysiology of bone remodeling. Clinical Chemistry 45, 1353 (1999). S. H. Ralston: Osteoporosis. British Medical Journal 315, 469 (1997). J. K. S. Rao, C. D. Katsetos, M. M. Herman, et al.: Experimental aluminum encephalomyelopathy. Toxicology 18, 687 (1998). J. Sainz, J. M. Van Tornout, M. L. Loro, et al.: Vitamin D-receptor gene polymorphisms and bone density in prepubertal American girls of Mexican descent. New England Journal of Medicine 337, 77 (1997). D. L. Schneider and E. L. Barrett-Connor: Urinary N-telopeptide levels discriminate normal osteopenic, and osteoporotic bone mineral density. Archives of Internal Medicine 157, 1241 (1997). E. Shane, D. Mancini, K. Aaronson, et al.: bone mass vitamin D deficiency, and hyperparathyroidism in congestive heart failure. American Journal of Medicine 103, 197 (1997). S. J. Silverberg, E. Shane, D. W. Dempster, and J. E Bilezikian: The effects of vitamin D insufficiency in patients with primary hyperparathyroidism. American Journal of Medicine 107, 561 (1999). M. Sowers, D. Eyre, B. W. Hollis, et al.: Biochemical markers of bone turnover in lactating and nonlactating postpartum women. Journal of Clinical Endocrinology and Metabolism 80, 2210 (1995). E Steindl, E Ferenci, and H. E Dienes: Wilson's disease in patients presenting with liver disease: a diagnostic challenge. Gastroenterology 113, 212 (1997). G. J. Strewler: The physiology of parathyroid hormone-related protein. New England Journal of Medicine 342, 177 (2000). R. Subramanian and R. Khardori: Severe hypophosphatemia: pathological implications, clinical presentations and treatment. Medicine 79, 1 (2000). T. D. Thacher, P. R. Fischer, J. M. Pettifor, et al.: A comparison of calcium, vitamin D, or both for nutritional rickets in Nigerian children. New England Journal of Medicine 341,563 (1999). R. V. Thakker: Editorial: Multiple endocrine neoplasia-syndromes of the twentieth century. Journal of Clinical Endocrinology and Metabolism 83, 2617 (1998). J. R. Turnlund: Human whole-body copper metabolism. Clinical Nutrition 67, 960S (1998). R. Uauy, M. Oiivares, and M. Gonzalez: Essentiality of copper in humans. Clinical Nutrition 67, 952S (1998). P. S. Wang, D. H. Solomon, H. Mogun, and J. Avorn: HMG-CoA reductase inhibitors and the risk of hip fractures in elderly patients. Journal of the American Medical Association 283, 3205 (2000). N. B. Watts: Clinical utility of biochemical markers of bone remodeling. Clinical Chemistry 45, 1359 (1999). R. S. Weinstein and S. C. Manolagas: Apoptosis and osteoporosis. American Journal of Medicine 108, 153 (2000). R. S. Weinstein and S. C. Manolagas: Bone cell apoptosis: implications for the pathogenesis and treatment of osteoporosis. Physiology in Medicine 108, 153 (2000). J. R. Weisinger and E. Beilorin-Font: Magnesium and phosphorous. Lancet 352, 391 (1998). P. Whittaker: Iron and zinc interactions in humans. American Journal of Clinical Nutrition 68, 442S (1998). G. Worley, S. J. Claerhout, and S. P. Combs: Hypophosphatemia in malnourished children during refeeding. Clinical Pediatrics 6, 347 (1998). Y. Kureishi, Z. Luo, I. Shiojima, et al: The HMG-CoA reductase inhibitor simvastatin activates the protein kinase Akt and promotes angiogenesis in normocholesterolemic, animals. Nature Medicine 6, 1004 (2000).

CHAPTER

38

Vitamin Metabolism

The word vitamin is used to describe any of a heterogeneous group of organic molecules that are needed in small quantities for normal growth, reproduction, and homeostasis but that the human body is unable to synthesize in adequate amounts. The group includes the fat-soluble vitamins (A, D, E, and K) and the water-soluble vitamins (B-complex and C). Vitamins are generally needed in catalytic quantities and do not function as structural elements in the cell. Other organic compounds are not synthesized in the body but are required for maintenance of normal metabolism, such as essential fatty acids (sometimes inaccurately called "vitamin F"; Chapter 18) and essential amino acids (Chapter 2). These substances are needed in relatively large quantities, serve as nonregenerated substrates in metabolic reactions, and are used primarily as structural components in lipids and proteins, respectively. A number of other substances are essential food factors in various nonhuman species, e.g., biopterin, inositol, ubiquinone, lipoic acid, phosphatidylcholine, and paraaminobenzoic acid. They are often classified as "vitamin-like." Vitamins discussed in other chapters include vitamin D (Chapter 37) and vitamin K (Chapter 36). All the B vitamins function as cofactors or precursors for cofactors in enzyme-catalyzed reactions and are discussed in appropriate chapters. Less well-defined actions are reviewed here.

General Considerations Classification of vitamins into fat-soluble and watersoluble groups reflects the history of their discovery. This grouping is still useful, despite the lack of chemical relatedness within each class, because it mirrors other underlying similarities. Vitamers are chemically similar substances that have a qualitatively similar vitamin activity. Thus, "vitamin D" refers to ergocalciferol (D2) and cholecalciferol (D3) and sometimes to their 25-hydroxy- and 1,25-dihydroxy derivatives (Chapter 37). Similarly, pyridoxine (pyridoxol), pyridoxal, and pyridoxamine are vitamin B6 vitamers, riboflavin is the active form of vitamin B2 and cobalamin is vitamin B I:. The members of a particular vitamin "family" are functionally interchangeable and protect against deficiency symptoms for that vitamin. A vitamin and its corresponding deficiency disease are related as follows: 1. The putative vitamin is a normal dietary constituent of healthy individuals. 2. The diet consumed by persons exhibiting symptoms of the putative deficiency disease is lacking in the vitamin. 3. Symptoms of such persons can be alleviated by addition of the vitamin to their diet.

901

902

CHAPTER38 Vitamin Metabolism

TABLE 38-1

Genetic Disorders That Respond Favorably to Treatment with Pharmacological Doses of Vitamins* Therapeutic Vitamin

Daily Dose

Vitamin D (adult RDA = 200 IU = 50 ~g of cholecalciferol)

> 100,000 units

Ascorbic acid (vitamin C; adult RDA = 60 mg) Biotin (RDA has not been established)

Cobalamin (vitamin B l 2; adult RDA = 3.0/sg)

Unknown

4g

Ehlers-Danlos syndrome, type VI (ocular type)

Procollagen lysyl hydroxylase

10g

Multiple carboxylase deficiency w

10mg

Propionic acidemia; ketoacidosis

Holocarboxylase synthetase, or defect in intestinal or cellular uptake, or intracellular transport Propionyl-CoA carboxylase

25,000 units

>500/~g

Niacin (adult RDA = 13 mg = 13 niacin equiv.) Pyridoxine [vitamin B6; adult RDA = 2.2 mg (men), 2.0 mg (women)]

Megaloblastic anemia Methylmalonic aciduria; ketoacidosis Methylmalonic aciduria; homocystinuria

Defect in 25-(OH)D- 1-a-hydroxylase

Decreased intestinal absorption due to abnormality of intrinsic factor or ileal mucosal cells, or absence of intrinsic factor Transcobalamin II Adenosyl cobalamin formation Formation of both adenosyl and methyl cobalamins

Megaloblastic anemia and mental retardation w Homocystinuria and hypomethioninemia Formiminoglutamic aciduria and mental retardation

Defect in intestinal and perhaps brain transport of folate NS, N 10_Methylenetetrahydrofolate reductase Probably formiminotransferase

Lactic and pyruvic acidemia ~

Pyruvate carboxylase

Hartnup disease

Intestinal and renal transport of tryptophan (high protein diet needed in addition to niacin to relieve symptoms)

10-50 mg

Infantile convulsions

>10mg >25 mg 5-10 mg > 100mg

B6-responsive anemia~ Cystathioninuria Homocystinuria Primary hyperoxaluria, type I Xanthurenic aciduriaw Gyrate atrophy of choroid and retina; hyperornithinemia

Perhaps glutamate decarboxylase Unknown u Cystathionine synthetase Soluble glyoxalate, a-ketoglutarate carboligase Kynureninase Ornithine: 2-oxoacid aminotransferase

>10mg >10mg >5 mg

Lipoic acid

Biochemical/Biological Defect

Familial X-linked hypophosphatemic rickets~ Vitamin D-dependent rickets, type I

> 100 ~g >250/~g

Folic acid (folacin; adult RDA = 400/~g)

Disorder t

3-10 mg 50-250 mg

5-10 mg 18-30 mg

(continued)

903

CHAPTER38 Vitamin Metabolism

T A B L E 38-1

(continued)

Therapeutic Vitamin

Daily Dose

Disorder t

Thiamine (vitamin B 1; adult RDA = 1 mg/2000 kcal/d)

20 mg 5-20 mg

Megaloblastic anemia Branched-chain ketoaciduria (maple syrup urine disease) Pyruvic acidemia Lactic and pyruvic acidemia; Leigh's encephalomyelopathy ~

5-20 mg > 10 mg

Biochemical/Biological Defect Unknown Branched chain a-ketoacid decarboxylase Pyruvate decarboxylase Pyruvate decarboxylase (low K m form)

*These disorders can be thought of as relative vitamin deficiency states. Not all patients will respond to vitamins owing to the existence of phenocopies. Data from L. E. Rosenberg: Vitamin-responsive inherited metabolic disorders. In Advances in Human Genetics, Vol. 6, H. Harris and K. Hirschhorn, Eds. (Plenum Press, 1976); and S. H. Mudd: Vitamin-responsive genetic abnormalities, In Advances in Nutritional Research, Vol. 4 (Plenum Press, 1982). tAll disorders have an autosomal recessive pattern of inheritance except as follows: X-linked dominant inheritance; w of inheritance unknown; llAutosomal, probably recessive, inheritance; ~X-linked recessive inheritance.

4. The biochemical function of the vitamin should be related directly to the observed deficiency symptoms, although this is not always possible to demonstrate. The classic example is scurvy (scorbutus), which used to be common among sailors who ate no fresh fruit or vegetables during long sea voyages. Scurvy could be prevented by consumption of fresh citrus fruits, now known to be high in vitamin C (ascorbic acid). The deficiency arose because vitamin C was destroyed by the methods used to preserve food while at sea. Other examples of a single vitamin deficiency in human populations are rare, however, except where the dietary supply is adequate but utilization of the vitamin is impaired. Frequently, if one vitamin is lacking, others are as well, and the intake of protein, trace elements, and other nutrients is probably insufficient. This is particularly true of B vitamins. In addition to nutritional inadequacy, vitamin deficiency may result from malabsorption, effects of pharmacological agents, and abnormalities of vitamin metabolism or utilization. Thus, in biliary obstruction or pancreatic disease, the fat-soluble vitamins are poorly absorbed despite adequate dietary intake because of steatorrhea. Absorption, transport, activation, and utilization of vitamins require the participation of enzymes or other proteins whose synthesis is under genetic control. Dysfunction or absence of one of these proteins can produce a disease that is clinically indistinguishable from one caused by dietary deficiency. In vitamin-dependent or vitamin-responsive

disorders, use of pharmacological doses of the vitamin can sufficiently overcome the blockage for normal function to occur (Table 38-1; discussed at the end of the chapter). Vitamin deficiency can result from treatment with certain drugs. Thus, destruction of intestinal microorganisms by antibiotic therapy can produce symptoms of vitamin K deficiency. Isoniazid, used to treat tuberculosis, is a competitive inhibitor of pyridoxal kinase, which is needed to produce pyridoxal phosphate. Isoniazid can produce symptoms of pyridoxine deficiency. To prevent this, pyridoxine is often incorporated into isoniazid tablets. Methotrexate and related folate antagonists act by competitively inhibiting dihydrofolate reductase (Chapter 27). Two standards have been established to plan diets that contain adequate supplies of the vitamins and to aid in diagnosing vitamin deficiency diseases. 1. The minimum daily requirement (MDR) is the smallest amount of a substance needed by a person to prevent a deficiency syndrome. It is considered to represent the body's basic physiological requirement of the material. MDR values, which are established by the U.S. Food and Drug Administration (FDA), are not available for all vitamins. 2. The recommended daily allowance (RDA) is the amount of a compound needed daily to maintain good nutrition in most healthy people. RDA values are intended to serve as nutritional goals, not as dietary requirements. They are defined by the Food and Nutrition Board of the National Academy of Sciences

904

CHAPTER38 Vitamin Metabolism

of the USA. RDA values have been established for mineral elements, energy, protein, electrolytes, and water. Those for the vitamins are given in Appendix IV. Overdosage of vitamins A and D produces hypervitaminosis. In recognition of this, the FDA has ruled that vitamin A in doses greater than 10,000 IU and vitamin D in doses greater than 400 IU should be available on a prescription-only basis and that all products containing quantities of a vitamin in excess of 150% of its RDA be classified as a drug. Hypervitaminosis D was discussed in Chapter 37; vitamin A toxicosis is discussed below. The toxicity of high doses of vitamin B6 is also covered later in the chapter. Certain vitamins can be synthesized by humans in limited quantities. Niacin can be formed from tryptophan (Chapter 17). This pathway is not active enough to satisfy all the body's needs; however, in calculating the RDA for niacin, 60 mg of dietary tryptophan is considered equivalent to 1 mg of dietary niacin. In Hartnup's disease (see Table 38-1 and Chapter 17), a rare hereditary disorder in the transport of monoaminomonocarboxylic acids (e.g., tryptophan), a pellagra-like rash may appear, suggesting that over a long period of time dietary intake of niacin is insufficient for metabolic needs. This pattern also occurs in carcinoid syndrome in which much tryptophan is shunted into the synthesis of 5-hydroxytryptamine. Vitamin D is synthesized in the skin, provided radiant energy is available for the conversion (Chapter 37): 7-Dehydrocholesterol

photons

> cholecalciferol (vitamin D3) (38.1)

This pathway is adequate to supply the body's need for vitamin D, provided exposure to sunlight is adequate. Vitamin D becomes a vitamin when environmental conditions prevent synthesis of an adequate supply by the skin. Physiological age-related changes in the elderly can affect the nutritional status. Decreased active intestinal transport and atrophic gastritis impair the absorption of vitamins and other nutrients. Reduced exposure to sunlight can lead to decreased vitamin D synthesis. Many drugs may impair both appetite and absorption of nutrients. Some examples of unfavorable drug-nutrient interactions are drugs that inhibit stomach acid production (e.g., omeprazole); drugs that reduce vitamin B12 absorption; anticonvulsant drugs (e.g., barbiturates, phenytoin, primidone) that act by inducing hepatic microsomal enzymes which accelerate inactivation of vitamin D metabolites and aggravate osteoporosis(Chapter 37); interference with folate metabolism by antifolate drugs (methotrexate) used in the treatment of some neoplastic diseases; and vitamin B6 metabolism affected by isoniazid, hydralazine, and D-penicillamine. Examples of negative impacts of

vitamins on drug action are vitamin B6-dependent action of peripheral conversion of L-3,4-dihydroxyphenylalanine (L-dopa) to L-dopamine that is mediated by aromatic L-amino acid decarboxylase and prevents L-dopa's transport across the blood-brain barrier; also ingestion of large amounts of vitamin K-rich foods or supplements, and action of warfarin on anticoagulation (Chapter 36). L-Dopa is the metabolic precursor of L-dopamine and is used in the treatment of Parkinson's disease (Chapter 32). Thus, L-dopa is administered along with a peripherally acting inhibitor of aromatic L-amino acid decarboxylase (e.g., carbidopa). Vitamins A (and retinoids), C, and E are antioxidants. Randomized trials of dietary supplementation of antioxidant vitamins to determine their favorable and/or unfavorable physiological effects on cardiovascular diseases, cancer, and other chronic diseases are in progress.

38.1 Fat-Soluble Vitamins The fat-soluble vitamins share many properties despite their limited chemical similarity. They are absorbed into the intestinal lymphatics, along with other dietary lipids, after emulsification by bile salts. Lipid malabsorption accompanied by steatorrhea usually results in poor uptake of all the fat-soluble vitamins. Deficiency disease (except in the case of vitamin K) is difficult to produce in adults because large amounts of most fat-soluble vitamins are stored in the liver and in adipose tissue. The fat-soluble vitamins are assembled from isoprenoid units; this fact is apparent from examination of the structures of vitamins A, E, and K; cholesterol, the precursor of vitamin D, is derived from six isoprenoid units (Chapter 18). Specific biochemical functions for vitamins A, D, and K are known, but a role for vitamin E, other than as a relatively nonspecific antioxidant, remains elusive. Vitamin A

Nutrition and Chemistry The role of vitamin A in vision is fairly well understood, but the other functions of vitamin A are only beginning to be elucidated (e.g., mucus secretion, maintenance of the integrity of differentiated epithelia and of the immune system, growth, and reproduction). Loss of night vision (nyctalopia) is an early sign of vitamin A deficiency, and clinical features of well-developed deficiency include epidermal lesions, ocular changes, growth retardation, glandular degeneration, increased susceptibility to infection, and sterility. Natural and synthetic compounds with vitamin A activity and inactive synthetic analogues of vitamin A are collectively termed retinoids. The most biologically active,

SECTION 38.1

Fat-SolubleVitamins

905 CH,

CH,

H

H=OH

:L.L'

,o

,,

13-1onone (trimethylcyclohexenyl) ring FIGURE 38-1 Chemical structureof all-transretinol (vitaminA1), the most activeform of vitaminA. Oxidationof C15 to an aldehydeor an acid produces, respectively,retinaldehyde(retinal) and retinoic acid. The cis-trans isomerizationof the doublebondbetweenCll and C12 occursduring functioningof retinaldehydein vision. naturally occurring retinoid is all-trans retinol, also called vitamin A (Figure 38-1). The/3-ionone ring is required for biological activity. Vitamin A1 exists free or as retinyl esters of fatty acids (primarily palmitic acid) in foods of animal origin, including eggs, butter, cod liver oil, and the livers of other vertebrates. In most of the dietary retinol, the four double bonds of the side chain are in the trans configuration. They are readily oxidized by atmospheric oxygen, inactivating the vitamin. They can be protected by antioxidants such as vitamin E. Several provitamins are present in yellow and dark green leafy vegetables and fruits, such as carrots, mangoes, apricots, collard greens, and broccoli. They are collectively known as the carotenoid pigments or carotenes. The most widely occurring and biologically active is /~-carotene (Figure 38-2). Other nutritionally important carotenoid pigments are cryptoxanthine, a yellow pigment found in corn, and o~- and v-carotenes. Other carotenoids, such as lycopene the red pigment of tomatoes, and xanthophyll, lack the/~-ionone ring essential for vitamin A activity.

Absorption, Transport, and Metabolism Retinyl esters are hydrolyzed in the intestinal lumen by pancreatic carboxylic ester hydrolase, which also hydrolyzes cholesteryl esters. In mucosal cells, retinol is reesterified, mostly with long-chain fatty acids, by

/

38-2 Structure of all-trans/~-carotene.

FIGURE

acyl-CoA: retinol acyltransferase. The retinyl esters are incorporated into chylomicron particles and secreted into the lacteals. In humans and rats, 50% of the retinol is esterified with palmitic acid, 25% with stearic acid, and smaller amounts with linoleic and oleic acids. These esters are eventually taken up by the liver in chylomicron remnants./~-Carotene is cleaved in the intestinal mucosa to two molecules of retinaldehyde by/3-carotene-15,15'dioxygenase, a soluble enzyme. Other provitamins are also activated upon cleavage by this enzyme. Retinaldehyde is then reduced to retinol by retinaldehyde reductase, using either NADH or NADPH. In the liver, retinyl esters are hydrolyzed and reesterifled. More than 95 % of hepatic retinol is present as esters of long-chain fatty acids, primarily palmitate. In an adult receiving the RDA of vitamin A, a year's supply or more may be stored in the liver. More than 90% of the body's supply of vitamin A is stored in the liver. The hepatic parenchymal cells are involved in its uptake, storage, and metabolism. Retinyl esters are transferred to hepatic fat-storing cells (also called Ito cells or lipocytes) from the parenchymal cells. The capacity of these fat-storing cells may determine when vitamin A toxicosis becomes symptomatic. During the development of hepatic fibrosis (e.g., in alcoholic liver disease), vitamin A stores in Ito cells disappear and the cells differentiate to myofibroblasts. These cells appear to be the ones responsible for the increased collagen synthesis seen in fibrotic and cirrhotic livers. Retinol is released from the liver and transported in plasma bound to retinol-binding protein (RBP), which is synthesized by hepatic parenchymal cells. Less than 5% circulates as retinyl esters. Retinol-binding protein from human plasma is a monomeric polypeptide (M.W. 21,000), which has a single binding site. Transfer of retinol into cells may be mediated by cell surface receptors that specifically recognize RBE After binding and releasing its vitamin A, RBP appears to have decreased affinity for prealbumin (see below) and is rapidly filtered by the kidney and degraded or excreted. The retinol-RBP complex circulates as a 1:1 complex with prealbumin (transthyretin; M.W. 55,000), which

[3"Carotene is cleaved here to produce.two

~

.

A

906 also functions in transport of thyroid hormones. Interaction between transthyretin and RBP is quite specific (dissociation constant 10-6-10 -7 mol/L). Retinol-RBPtransthyretin complex (RTC) (M.W. 76,000) migrates electrophoretically as an o~-globulin. Normal concentrations of RBP and transthyretin in human plasma are 40-50 and 200-300 #g/mL, respectively. Transthyretin, a glycoprotein with a high tryptophan content, is synthesized in the liver and migrates electrophoretically ahead of albumin. It has a half-life of 2 days and has a small pool size. These two properties make it a sensitive indicator of nutritional status (Chapter 17). Its plasma level decreases in protein-calorie malnutrition, liver disease, and acute inflammatory diseases. The serum concentration of retinol is held remarkably constant despite wide variation in dietary intake and hepatic stores of the vitamin by tight control over the concentration of RBE In vitamin A-deficient rats, hepatic secretion of RBP is specifically blocked, and its level in serum falls whereas that in the liver increases. Administration of vitamin A causes a rapid proportional release of hepatic RBE The rapidity indicates the release of pooled protein rather than new synthesis. Patients suffering from parenchymal liver disease or protein-calorie malnutrition have reduced amounts of serum RBP and vitamin A. Provision of an adequate diet to malnourished children increases plasma concentrations of RBP and retinol, even without vitamin A supplementation, suggesting the presence of stores of retinol that could not be mobilized on the inadequate diet. The plasma concentration of RBP increases in renal disease, indicating that the kidney is a major catabolic site for this protein. Complex formation with transthyretin reduces glomerular filtration and, consequently, renal catabolism of RBE In rat hepatocyte cultures, glucocorticoids stimulate net synthesis of RBE Oxidation of retinol produces retinoic acid (tretinoin). The reaction is irreversible. Retinoic acid enters the portal blood, is transported bound to albumin, and is not stored to any great extent. The concentration of retinoic acid in plasma is normally 3-4 ng/mL. A biologically active metabolite, 5,6-epoxyretinoic acid, has been isolated from the intestinal mucosa of vitamin A-deficient rats following administration of 3H-retinoic acid. Several tissues have specific cellular retinoic acid-binding proteins (CRABPs).

Function

Vitamin A and its derivatives retinal and retinoic acid have many essential biological roles in such processes as vision, regulation of cell proliferation and differentiation, and as morphogenetic agents during embryonic

CHAPTER38 Vitamin Metabolism

development and differentiation. Both natural and synthetic analogues (known as retinoids) possess varying degrees of biological activity ascribed to vitamin A. Other than the role of retinal in vision, retinoids exert their biological actions by binding to specific nuclear receptors, such as transcription factors that modulate gene expression. The retinoid receptors belong to two subfamilies: retinoic acid receptors (RARs) and retinoid X receptors (RXRs). Both the RARs and RXRs contain three isotypes, c~, fl, and V encoded by separate genes. Thus, the retinoid receptor family includes RARol, RARfl, RARv, RXRot, RXRfl, and RXRv, which are members of the steroid and thyroid hormone superfamily of receptors (Chapter 30). All of these receptors possess at least two domains: a ligand binding domain and a DNA binding domain. The DNA binding domain of the receptor recognizes retinoic acid response elements within the target gene via the two zinc finger binding motifs. The receptors form dimers of various combinations and permutations within the superfamily, and each dimer may perform a specific biological function by binding to specific DNA response elements. Activation of retinoid receptors can lead to inhibition of cell proliferation, induction of differentiation, and induction of apoptosis during normal cell development. The importance of RARs in cell proliferation and differentiation is illustrated by acute promyeloeyrie leukemia (PML), which is a subtype of acute myeloid leukemia. PML is characterized by abnormal hypergranular promyelocytes, a cytogenetic translocation, and a bleeding disorder secondary to consumptive coagulopathy and fibrinolysis. The bleeding problems are presumably initiated by procoagulant phospholipids present in the leukemic cells (Chapter 36). The cytogenetic hallmark of PML consists of a balanced reciprocal translocation between the long arms of chromosomes 15 and 17, designated as t(15;17) (Figure 38-3). The translocation results in two recombinant chromosomes, one abnormally long 15q+ and one shortened 17q-. This translocation fuses the PML gene located on chromosome 15 with the RARo~ gene located on chromosome 17. The two chimeric genes formed are PML-RAR~ and RARo~PML. Of these, the PML-RAR~ gene is transcriptionally active and produces a PML-RARc~ protein. This protein, which is an oncoprotein, is responsible for the pathogenesis of PML by interfering with the normal functions of PML and RARot genes in differentiation of myeloid precursors. The normal PML gene product has growth suppressor activity. Treatment with all-trans retinoic acid has produced complete remissions in PML by promoting the conversion of leukemic blast cells into mature cells. This is because the PML-RARo~ oncoprotein maintains responsiveness to retinoic acid's biological actions. Retinoic acid

907

SECTION38.1 Fat-Soluble Vitamins

F I G U R E 38-3 Karyotype of a promyelocytic leukemic cell, with balanced reciprocal translocation between chromosomes 15 and 17, t(15; 17). The rearranged chromosomes are identified by arrows. (Courtesy of Dr. Mark H. Bogart.)

therapy may also augment the degradation of the PMLRARo~ fusion protein. Transgenic mice with a PML-RARot fusion gene develop PML, which supports the pathogenic role of the fusion protein in human PML. Retinyl phosphate, acting as a sugar carrier in a manner similar to that of dolichol phosphate (Chapter 16), may be necessary for normal glycosylation of a subgroup of essential glycoproteins. In vitamin A deficiency, synthesis of some glycoproteins is decreased. Retinyl phosphate and mannosyl phosphoryl retinol have been identified in vivo and have been synthesized in vitro using biological preparations. In rat liver, GDP-mannose: retinyl phosphate mannosyltransferase has its highest specific activity in the endoplasmic reticulum. However, retinoic acid can fully support growth in the whole animal even though it is not converted to retinol and does not participate in sugar transfer reactions. More than 1500 retinoids have been synthesized. Two in particular, 13-cis-retinoic acid (isotretinoin) and etretihate (Figure 38-4), have generated considerable interest as agents for the treatment of dermatological disorders. 13-cis retinoic acid inhibits sebum production, is the drug of choice for treatment of severe cystic acne, and is useful in the treatment of other forms of acne. Etretinate is used in Europe for treatment of psoriasis and related disorders.

Hypo- and Hypervitaminosis A Serious vitamin A deficiency is not a major public health problem in North America; however, where poverty and poor nutrition are common many persons suffer from vitamin A deficiency with active corneal involvement.

This condition leads to permanent blindness in about half of cases, and many of those affected are preschool children. Night blindness occurs early in vitamin A deficiency. There is a reduction in rhodopsin concentration, followed by retinal degeneration and loss of photoreceptor cells. Degenerative changes may be due to instability of free opsin or may indicate an additional nutritive function for retinaldehyde (or retinoic acid) in retinal cells. The degenerative changes of retinitis pigmentosa, a relatively common, inherited cause of blindness, closely resemble those of vitamin A deficiency. Ingestion of vitamin A in large excess of the RDA can cause toxicity. Daily intake of more than 7500 retinol equivalents (25,000 IU) is not recommended, and doses in excess of 3000 retinol equivalents (10,000 IU) should only be used with medical supervision. Acute toxicity after

OOH

(a) 0

. 0o2

II

(b)

FIGURE 38-4 Structures of 13-cis-retinoic acid (isotretinoin) (a) and etretinate (b), synthetic retinoids used as pharmacological agents.

908

CHAPTER38 Vitamin Metabolism ROD CELL

CONE CELL

Disks

Outer

segment

Invaginations of plasma membrane (equivalent to disks)

Plasma membrane 3ytoplasm

ConneOn0 structure

Mitochondrion _ _ ugh endoplasmic

reticulum

. , ."

~'. ~

~

i~

.%~..'jI

Light path in vivo

Inner

segment - -

Synaptic body

FIGURE 38-5 Schematic representations of mammalian retinal rod and cone cells. There are about 500-1500 disks in the outer segment of each rod cell. The deep membrane invaginationsin the plasma membrane of the cone cell outer segment are thought to be functionally equivalent to the rod cell disks.

a single large dose may last several days. Chronic toxicity occurs in adults after daily doses of 7500-15,000 retinol equivalents (25,000-50,000 IU) taken for several months. Ingestion of carotenes alone does not cause vitamin A toxicity, probably because of markedly decreased absorption at high doses and feedback inhibition of carotene conversion to retinaldehyde, but can cause harmless carotenemia with yellowing of the skin, particularly on the palmar and plantar surfaces. Carotenemia can cause falsely elevated values for the icterus index or for other direct reading methods for estimating serum bilirubin (Chapter 29). In cases of toxicity, total serum vitamin A concentration is elevated two- to eightfold, mostly in the retinyl esters, which are associated with plasma lipoproteins, while the RBP level is normal or only slightly increased. Unesterifled retinol is generally increased less than twofold. Vitamin A is a surfactant, and its toxicity may be due to labilization and disruption of biological membranes. Excessive amounts of retinol also increase synthesis and release oflysosomal hydrolases. In in vitro systems, retinol bound to RBP is not toxic, and RBP alone protects against

toxicity of previously added vitamin A. Thus, clinical toxicity occurs only when the binding capacity of RBP is exceeded and retinol and retinyl esters are present either free or in lipoproteins in the circulation.

Vision and Vitamin A

Photosensitivity in various organisms is based on photoisomerization of retinaldehyde. In humans, l l - c i s retinaldehyde complexes with opsin to produce rhodopsin (visual purple). Absorption of a photon by the :relectron system of retinaldehyde changes it to the alltrans isomer; this reaction--the only light-catalyzed step in vision--is transduced to an action potential transmitted in the optic nerve. The human visual system functions over a range of light intensity. If properly dark adapted, the eye can detect a single photon. Vitamin A deficiency reduces the amount of rhodopsin in the retina, thereby increasing the minimum amount of illumination that can be detected (the visual threshold) and causing night blindness.

909

SECTION38.1 Fat-Soluble Vitamins

The retina consists of a thin layer of photoreceptor cells connected to the optic nerve via axons, synapses, and the bodies of several intermediate cells. Other structures of the eye provide the optical system for focusing light onto the retina. Visual pigments are present in the approximately 100 million rod and 5 million cone cells (Figure 38-5). The cones are less sensitive than the rods but are required for color vision. Three types of cone cells differ with respect to the wavelengths of light to which they are maximally sensitive. Comparison and integration of inputs from these three cell types by the visual cortex produce color vision. At low light levels, only the rods are active and color vision is lost. A higher pigment content and greater molar absorptivity of the pigment contribute to the higher sensitivity of rods. Rhodopsin has a broad absorption spectrum with a maximum at 500 nm, making the rods most sensitive to green light. In the human retina, cone cell absorption maxima are 445 nm (blue), 535 nm (green), and 570 nm (red). Opsins in the three types of cone cell have different primary structures and are coded by three separate genes. Those for red- and green-sensitive opsins are on the X chromosome, while that for blue-sensitive opsin is on an autosome. Congenital color blindness, which affects about 9% of the male population, results from the absence of one or more cone cell types or from a decrease in amount of one of the pigments. Absence of red or green cones occurs in 2.5% of males. Absence of blue cones occurs in only 0.001% of males. Decrease in red cone pigment (protanomaly) or green cone pigment (deuteranomaly) occurs in 1.3% and 5.0%, respectively, of males. The decrease in pigment type presumably is due to mutation in a structural gene. Absence of two or three types of cone cell is extremely rare. Light reception and energy transduction take place in the rod outer segments, while cellular metabolism occurs in the rod inner segment, which is rich in glycogen, mitochondria, and rough endoplasmic reticulum. The outer segment contains 500-1500 flattened membrane sacs (disks), each about 16 nm thick, which are electrically isolated from the surrounding plasma membrane and which contain rhodopsin as an integral, transmembrane protein. The disks are continually replaced from the outer segment nearest the nucleus. Phagocytosis of old disks by cells of the retinal pigment epithelium occurs at the top of the outer segment. In the rhesus monkey, the lifetime of a disk is 9-13 days. Disks are formed by the sealing off of invaginations of plasma membrane, and the composition of their contents differs from that of cytoplasm. Blindness due to hereditary defects in regeneration of disks has been observed in rats. Cone cells are similar in many ways to rod cells (Figure 38-5). Because cone ceils lack distinct disks, their visual pigments are located on deep invaginations

of the plasma membrane that resemble disks and probably are formed by a similar mechanism. Disk membranes are composed primarily of phospholipid and rhodopsin at a molecular ratio of about 70:1. The phospholipids are 40% phosphatidylcholine, 40% phosphatidylethanolamine, and 13% phosphatidylserine. The membrane has high fluidity because of extensive unsaturation of the lipids, which gives rhodopsin considerable rotational and translational mobility. Average spacing of rhodopsin molecules is 5.6 nm. Bovine rhodopsin (M.W. 39,000) has two asparaginelinked oligosaccharides with the structure (mannose)3 (N-acetylglucosamine). Seven hydrophobic o~-helical segments, each 21-28 amino acids long, are embedded in the disk membrane and are connected by hydrophilic sequences that are exposed to either side of the membrane. The retinaldehyde moiety is within the membrane, halfway down the first helical segment, attached by an aldimine linkage to the e-amino group of lysine 53 residues from the carboxyl terminus by aldimine linkage (Figure 38-6).

2H

Additional hydrophilic and hydrophobic residues

(

)

Disk interior

Oligosaccharide chains FIGURE 38-6

Diagrammatic representation of bovinerhodopsinembeddedin the disk membrane. Onlythree of the seveno~-helicalsegmentsare shown.The only point of attachmentbetween retinaldehydeand opsin is the indicated aldimine bond. [Modifiedand reproduced, with permission, from D. E O'Brien, The chemistryof vision. Science 218, 961 (1982), (~) 1982 by the AmericanAssociationfor the Advancementof Science.]

910

CHAPTER38 Vitamin Metabolism

In the dark, rhodopsin has a red color, which changes to pale yellow upon exposure to light. This bleaching takes place within a few milliseconds after absorption of a photon and occurs because all-trans retinaldehyde bound to opsin is unstable. At low temperatures, bleaching proceeds more slowly, and seven intermediates have been identified (Figure 38-7). The structures and order of appearance of these intermediates have yet to be clarified. The final

(53")-opsin Rhodopsin

H

hv

Hypsoriodopsin Bathorhodopsin

I Lumirhodopsin [ Metarhodopsin I 1 Metarhodopsin II l Pararhodopsin l

N'RetiHn~dene~ s -Retinaldehyde + H,N-Lys (53")-opsin F I G U R E 38-7 Bleaching of rhodopsin in bovine retina. Absorption of light by 11-cis-retinaldehyde initiates the configurational change to all-trans retinaldehyde, culminating in hydrolysis of the bond between retinaldehyde and Lys 53' of opsin. The order of formation of the intermediates is tentative. The symbol h v represents a photon of visible light. [Modified and reproduced, with permission, from C. D. B. Bridges, Retinoids in photosensitive systems. In: The Retinoids, Vol. 2, M. B. Sporn, A. B. Roberts, and D. S. Goodman, eds. Academic Press, San Diego, 1984).]

F I G U R E 38-8 Regeneration of 1 l-cis-retinaldehyde (1 l-cisRal). Following light exposure, rhodopsin (opsin -t- I l-cisRal) breaks down to opsin and all-trans retinaldehyde (atRal) initiating a neural signal. The restoration of I l-cisRal begins with the atRal combining with phosphatidylethanolamine (PE) to form the protonatal Schiff base N-retinylidine-PE complex. This complex is transported across the disk membrane by an ATP-binding cassette protein, exclusively found in the retinal outer segment (ABCR). Defects in ABCR cause early-onset maculopathy (Stargardt's disease). After reduction of atRal to all trans-retinol (atRol) by atRol dehydrogenase in the retinal outer segment, atRol is converted to 1 l-cisRal in a series of reactions occuring in the retinal pigment epithelial cell (RPE). Also shown in the bottom is the phagocytosis of disk by RPE.

compound (perhaps N-retinylideneopsin) is unstable and hydrolyzes to all-trans retinaldehyde plus opsin. The regeneration of rhodopsin following exposure to bright light requires the following steps. The all-trans retinaldehyde combines with phosphotidylethanolamine (PE) to form a protonated Schiff base N-retinylidene-PE. This complex is then exported out of the disk by an ATP-binding cassette (ABC) transporter exclusively located in the retinol disk membrane (ABCR). Outside the disk the all-trans retinaldehyde is reduced to all-trans retinol by a dehydrogenase and transported to retinal

911

SECTION 38.1 Fat-Soluble Vitamins

pigment epithelial (RPE) cells. In the RPE cells involving a series of reactions all-trans retinol is converted to 11-cis retinaldehyde which is transported to rod cells. In the rod cells, 11-cis retinaldehyde reassociates to form rhodopsin. The importance of ABCR becomes evident in early onset macular degeneration, due to mutations in the ABCR gene. This disorder is known as Stargardt's disease, an inherited autosomal recessive disease, which affects one person in 10,000. Age-related macular degeneration in susceptible individuals with risk factors (e.g., high lipid diet, smoking) may also be due in part to defects in ABCR gene. ABC transporters are involved in the transport of a wide variety of molecules. For example, cystic fibrosis is caused by a defective ABC transporter for chloride ion (cystic fibrosis transmembrane regulator, Chapter 12) and in Tangier's disease, the abnormality of cholesterol efflux is due to defects in an ABC protein (Chapter 20). In the dark, the cation (Na +, CaZ+)-specific cGMPgated channels located in the rod outer segment (ROS) are open, thus promoting the influx of Na + and Ca 2+. The steady state of cations is maintained by outward pumping of Ca 2+ by the Na+/Ca 2+ exchanger and Na +, K+-ATPase pumps located in the inner segment. Exposure to light blocks cGMP-gated cation channels, causing the inside of the plasma membrane to become more negative and resulting in hyperpolarization. The signal of hyperpolarization is transmitted to the synaptic body and eventually to the brain. In the dark, because of the influx of Na + and Ca 2+, the rod cells are depolarized and neurotransmitters (possibly aspartate or glutamate) are released from the presynaptic membrane of the rod cell at relatively high rates. In the hyperpolarized state the neurotransmitter release is inhibited. Thus, the postsynaptic bipolar cells are either inhibited (hyperpolarized) or excited (depolarized). Coupling of rhodopsin present in the disk membrane to cation-specific cGMP-gated channels of the plasma membrane involves signal amplification after photon absorption. The coupling mechanism (Figure 38-9) begins with the absorption of light by l l-cis-retinal-rhodopsin and its conversion to all-trans retinal and the photoactivated rhodopsin (R*). In the next step, activated rhodopsin initiates an amplification process that consists of activating transducin, a signal-coupling G-protein. The active form of transducin, T~.GTp, activates cGMP-phosphodiesterase (PDE). In the second amplification process, PDE hydrolyzes cGMP with the closure of cation channels. The opening and closing of these cation channels that is mediated by cGMP is similar to Ca 2+ ion channel gating by cAMP in olfaction. PDE belongs to a superfamily of enzymes that regulate cell function by maintaining cyclic nucleotide levels. There are cAMP-specific PDEs (PDE4, PDE7, and PDE8),

Light

Rhodopsin-11-cis retinal (present in the disc membrane) -~ AII-trans retinal Photoactivated Rhodopsin (R*)

I Amplificationprocess GTP Transducin (T)c~.GDP.13~,

GDP )

~

Inactive Phosphodiesterase (PDE-I)

) T .G-rP+ T~_

9 Active PDE + T.GT P - I ' -"

H20 cGMP

\

), 5" -GMP

Plasma membrane cation channels closure; Decreased Ca2+ and Na § concentration; hyperpolarization

FIGURE 38-9 Mechanism for stimulus-response coupling of photon absorption to the closure of plasma membrane cation channels. I, Inhibitory subunit of phosphodiesterase.

cGMP-specific PDEs (PDE5, PDE6, and PDE9), and ones specific for both cAMP and cGMP PDEs (PDE1, PDE2, and PDE10). Light induced-activated transducin activates a cGMP-specific PDE, namely, PDE6. Another previously discussed cGMP-specific PDE is PDE5, which is abundant in vascular smooth muscle (Chapter 17). PDE5 is involved in the nitric oxide and cGMP signaling pathway. A specific inhibitor of PDE5, sildenafil, causes increased blood flow into the sinusoidal spaces of the corpus cavernosum and corpus spongiosum, resulting in erection of the penis (Chapter 17). Photoactivation of rhodopsin produces a conformational change that allows it to interact with and activate transducin, resulting in the replacement of GDP by GTP at the o~subunit. Metarhodopsin II (Figure 38-7) is the form that interacts with transducin at the disk membrane surface. Depending on the lifetime of the activated rhodopsin molecule, each molecule can activate up to 500 transducin molecules. The o~-subunit GTP complex of transducin disinhibits PDE by removing its inhibitor subunit (I), causing it to hydrolyze cGMP to 5'-GME The decrease in cGMP concentration closes the plasma membrane cation channels, decreasing Na + and Ca 2+ concentrations. Some cases of retinitis pigmentosa are caused by mutations in the transducin gene that prevent it from disinhibiting the PDE, leading to an accumulation of cGMP that is toxic to the retina. The transducin-GTP complex remains active as long as the GTP bound to G~ is not hydrolyzed. The recovery and adaptation of the rod and the cone cells to the dark state begins shortly after illumination

912

CHAPTER 38 Vitamin M e t a b o l i s m

(a)

Photoactivated Rhodopsin (R*) ,ow

;,

ATP

> [ R h o d o p s i n kinase

Recoverin J ~

~

ADP

Phosphorylated - R (P- R)

Arrestin

P,