Class 2 Transferases X: EC 2.7.1.113 - 2.7.5.7 (Springer Handbook of Enzymes) [2nd ed.] 9783540478164, 3540478167

The objective of the Springer Handbook of Enzymes is to provide in concise form data on enzymes sufficiently well charac

239 76 3MB

English Pages 625 [643] Year 2007

Report DMCA / Copyright

DOWNLOAD PDF FILE

Recommend Papers

Class 2 Transferases X: EC 2.7.1.113 - 2.7.5.7 (Springer Handbook of Enzymes) [2nd ed.]
 9783540478164, 3540478167

  • 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

       

Dietmar Schomburg and Ida Schomburg (Eds.)

     Volume 37 Class 2  Transferases X EC 2.7.1.113±2.7.5.7 coedited by Antje Chang

Second Edition

13

Professor Dietmar Schomburg e-mail: [email protected] Dr. Ida Schomburg e-mail: [email protected]

Technical University Braunschweig Bioinformatics & Systems Biology Langer Kamp 19b 38106 Braunschweig Germany

Dr. Antje Chang e-mail: [email protected]

Library of Congress Control Number: 2007922568

ISBN 978-3-540-47816-4

2nd Edition Springer Berlin Heidelberg New York

The first edition was published as Volume 14 of the ªEnzyme Handbookº.

This work is subject to copyright. All rights are reserved, whether the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilm or in any other way, and storage in data banks. Duplication of this publication or parts thereof is permitted only under the provisions of the German Copyright Law of September 9, 1965, in its current version, and permission for use must always be obtained from Springer. Violations are liable to prosecution under the German Copyright Law. Springer is a part of Springer Science+Business Media springer.com # Springer-Verlag Berlin Heidelberg 2007 Printed in Germany The use of general descriptive names, registered names, etc. in this publication does not imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations and free for general use. The publisher cannot assume any legal responsibility for given data, especially as far as directions for the use and the handling of chemicals and biological material are concerned. This information can be obtained from the instructions on safe laboratory practice and from the manufacturers of chemicals and laboratory equipment. Cover design: Erich Kirchner, Heidelberg Typesetting: medionet Prepress Services Ltd., Berlin Printed on acid-free paper 2/3141m-5 4 3 2 1 0

          

Information on this handbook can be found on the internet at http://www.springer.com choosing ªChemistryº and then ªReference Worksº. A complete list of all enzyme entries either as an alphabetical Name Index or as the EC-Number Index is available at the above mentioned URL. You can download and print them free of charge. A complete list of all synonyms (> 25,000 entries) used for the enzymes is available in print form (ISBN 3-540-41830-X).

Save 15 %

We recommend a standing order for the series to ensure you automatically receive all volumes and all supplements and save 15 % on the list price.

 

Today, as the full information about the genome is becoming available for a rapidly increasing number of organisms and transcriptome and proteome analyses are beginning to provide us with a much wider image of protein regulation and function, it is obvious that there are limitations to our ability to access functional data for the gene products ± the proteins and, in particular, for enzymes. Those data are inherently very difficult to collect, interpret and standardize as they are widely distributed among journals from different fields and are often subject to experimental conditions. Nevertheless a systematic collection is essential for our interpretation of genome information and more so for applications of this knowledge in the fields of medicine, agriculture, etc. Progress on enzyme immobilisation, enzyme production, enzyme inhibition, coenzyme regeneration and enzyme engineering has opened up fascinating new fields for the potential application of enzymes in a wide range of different areas. The development of the enzyme data information system BRENDAwas started in 1987 at the German National Research Centre for Biotechnology in Braunschweig (GBF), continued at the University of Cologne from 1996 to 2007, and is now returning to Braunschweig, to the Technical University, Institute of Bioinformatics & Systems Biology. The present book ªSpringer Handbook of Enzymesº represents the printed version of this data bank. The information system has been developed into a full metabolic database. The enzymes in this Handbook are arranged according to the Enzyme Commission list of enzymes. Some 4,000 ªdifferentº enzymes are covered. Frequently enzymes with very different properties are included under the same EC-number. Although we intend to give a representative overview on the characteristics and variability of each enzyme, the Handbook is not a compendium. The reader will have to go to the primary literature for more detailed information. Naturally it is not possible to cover all the numerous literature references for each enzyme (for some enzymes up to 40,000) if the data representation is to be concise as is intended. It should be mentioned here that the data have been extracted from the literature and critically evaluated by qualified scientists. On the other hand, the original authors' nomenclature for enzyme forms and subunits is retained. In order to keep the tables concise, redundant information is avoided as far as possible (e.g. if Km values are measured in the presence of an obvious cosubstrate, only the name of the cosubstrate is given in parentheses as a commentary without reference to its specific role). The authors are grateful to the following biologists and chemists for invaluable help in the compilation of data: Cornelia Munaretto and Dr. Antje Chang. Braunschweig Spring 2007

       

VII

!   "  

A Ac ADP Ala All Alt AMP Ara Arg Asn Asp ATP Bicine C cal CDP CDTA CMP CoA CTP Cys d dDFP DNA DPN DTNB DTT EC E. coli EDTA EGTA ER Et EXAFS FAD FMN Fru Fuc G Gal

adenine acetyl adenosine 5'-diphosphate alanine allose altrose adenosine 5'-monophosphate arabinose arginine asparagine aspartic acid adenosine 5'-triphosphate N,N'-bis(2-hydroxyethyl)glycine cytosine calorie cytidine 5'-diphosphate trans-1,2-diaminocyclohexane-N,N,N,N-tetraacetic acid cytidine 5'-monophosphate coenzyme A cytidine 5'-triphosphate cysteine deoxy(and l-) prefixes indicating configuration diisopropyl fluorophosphate deoxyribonucleic acid diphosphopyridinium nucleotide (now NAD+ ) 5,5'-dithiobis(2-nitrobenzoate) dithiothreitol (i.e. Cleland's reagent) number of enzyme in Enzyme Commission's system Escherichia coli ethylene diaminetetraacetate ethylene glycol bis(-aminoethyl ether) tetraacetate endoplasmic reticulum ethyl extended X-ray absorption fine structure flavin-adenine dinucleotide flavin mononucleotide (riboflavin 5'-monophosphate) fructose fucose guanine galactose

IX

List of Abbreviations

GDP Glc GlcN GlcNAc Gln Glu Gly GMP GSH GSSG GTP Gul h H4 HEPES His HPLC Hyl Hyp IAA IC 50 Ig Ile Ido IDP IMP ITP Km lLeu Lys Lyx M mM  Man MES Met min MOPS Mur MW NAD+ NADH NADP+ NADPH NAD(P)H

X

guanosine 5'-diphosphate glucose glucosamine N-acetylglucosamine glutamine glutamic acid glycine guanosine 5'-monophosphate glutathione oxidized glutathione guanosine 5'-triphosphate gulose hour tetrahydro 4-(2-hydroxyethyl)-1-piperazineethane sulfonic acid histidine high performance liquid chromatography hydroxylysine hydroxyproline iodoacetamide 50% inhibitory concentration immunoglobulin isoleucine idose inosine 5'-diphosphate inosine 5'-monophosphate inosine 5'-triphosphate Michaelis constant (and d-) prefixes indicating configuration leucine lysine lyxose mol/l millimol/l  mannose 2-(N-morpholino)ethane sulfonate methionine minute 3-(N-morpholino)propane sulfonate muramic acid molecular weight nicotinamide-adenine dinucleotide reduced NAD NAD phosphate reduced NADP indicates either NADH or NADPH

List of Abbreviations

NBS NDP NEM Neu NMN NMP NTP  Orn  PBS PCMB PEP pH Ph Phe PHMB PIXE PMSF p-NPP Pro Q10 Rha Rib RNA mRNA rRNA tRNA Sar SDS-PAGE Ser T tH Tal TDP TEA Thr TLCK Tm TMP TosTPN Tris Trp TTP Tyr U

N-bromosuccinimide nucleoside 5'-diphosphate N-ethylmaleimide neuraminic acid nicotinamide mononucleotide nucleoside 5'-monophosphate nucleoside 5'-triphosphate   ornithine  phosphate-buffered saline -chloromercuribenzoate phosphoenolpyruvate -log10[H+ ] phenyl phenylalanine -hydroxymercuribenzoate proton-induced X-ray emission phenylmethane-sulfonylfluoride -nitrophenyl phosphate proline factor for the change in reaction rate for a 10 C temperature increase rhamnose ribose ribonucleic acid messenger RNA ribosomal RNA transfer RNA N-methylglycine (sarcosine) sodium dodecyl sulfate polyacrylamide gel electrophoresis serine thymine time for half-completion of reaction talose thymidine 5'-diphosphate triethanolamine threonine Na--tosyl-l-lysine chloromethyl ketone melting temperature thymidine 5'-monophosphate tosyl- (-toluenesulfonyl-) triphosphopyridinium nucleotide (now NADP+ ) tris(hydroxymethyl)-aminomethane tryptophan thymidine 5'-triphosphate tyrosine uridine

XI

List of Abbreviations

U/mg UDP UMP UTP Val Xaa XAS Xyl

XII

mmol/(mg*min) uridine 5'-diphosphate uridine 5'-monophosphate uridine 5'-triphosphate valine symbol for an amino acid of unknown constitution in peptide formula X-ray absorption spectroscopy xylose

!  #  $  

Since its foundation in 1956 the Nomenclature Committee of the International Union of Biochemistry and Molecular Biology (NC-IUBMB) has continually revised and updated the list of enzymes. Entries for new enzymes have been added, others have been deleted completely, or transferred to another EC number in the original class or to different EC classes, catalyzing other types of chemical reactions. The old numbers have not been allotted to new enzymes; instead the place has been left vacant or cross-references given to the changes in nomenclature. Deleted and Transferred Enzymes For EC class 2.7.1.113±2.7.5.7 these changes are: Recommended name

Old EC number Alteration

inositol-trisphosphate 6-kinase inositol-trisphosphate 5-kinase inositol-hexakisphosphate kinase carbamoyl-phosphate synthase (ammonia) carbamoyl-phosphate synthase (glutamine) deoxycytidylate kinase

2.7.1.133 2.7.1.139 2.7.1.152 2.7.2.5

transferred to EC 2.7.1.134 transferred to EC 2.7.1.134 transferred to EC 2.7.4.21 transferred to EC 6.3.4.16

2.7.2.9

transferred to EC 6.3.5.5

2.7.4.5

phosphoglucomutase acetylglucosamine phosphomutase phosphoglyceromutase bisphosphoglyceromutase phosphoglucomutase (glucose-cofactor) phosphopentomutase phosphomannomutase

2.7.5.1 2.7.5.2

deleted, included in EC 2.7.4.14 transferred to EC 5.4.2.2 transferred to EC 5.4.2.3

2.7.5.3 2.7.5.4 2.7.5.5

transferred to EC 5.4.2.1 transferred to EC 5.4.2.4 transferred to EC 5.4.2.5

2.7.5.6 2.7.5.7

transferred to EC 5.4.2.7 transferred to EC 5.4.2.8

XIII

% &  '    ( 

EC-No.

Recommended Name

2.7.2.1 2.7.2.12 2.7.1.128

acetate kinase . . . . . . . . . . . . . . . . . . . . . . . . . acetate kinase (diphosphate) . . . . . . . . . . . . . . . . . . . [acetyl-CoA carboxylase] kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.4) . . . . . . . . . . . . . . . . . .            !" . . . . . acetylglutamate kinase . . . . . . . . . . . . . . . . . . . . . adenosylcobinamide kinase . . . . . . . . . . . . . . . . . . . (deoxy)adenylate kinase . . . . . . . . . . . . . . . . . . . . . adenylate kinase . . . . . . . . . . . . . . . . . . . . . . . . ADP-specific glucokinase . . . . . . . . . . . . . . . . . . . . ADP-specific phosphofructokinase . . . . . . . . . . . . . . . . ADP-thymidine kinase . . . . . . . . . . . . . . . . . . . . . b-adrenergic-receptor kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.15) . . . . . . . . . . . . . . . . . . . . . . . . . agmatine kinase . . . . . . . . . . . . . . . . . . . . . . . . ammonia kinase . . . . . . . . . . . . . . . . . . . . . . . . AMP-thymidine kinase . . . . . . . . . . . . . . . . . . . . . arginine kinase . . . . . . . . . . . . . . . . . . . . . . . . aspartate kinase . . . . . . . . . . . . . . . . . . . . . . . .            " . . . . . . . . . branched-chain-fatty-acid kinase . . . . . . . . . . . . . . . . . butyrate kinase . . . . . . . . . . . . . . . . . . . . . . . . Ca2+ /calmodulin-dependent protein kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.17) . . . . . . . . . . . . . . . . . . caldesmon kinase . . . . . . . . . . . . . . . . . . . . . . . carbamate kinase. . . . . . . . . . . . . . . . . . . . . . . .         "    #! $#" .         "    #!" . ceramide kinase . . . . . . . . . . . . . . . . . . . . . . . . creatine kinase. . . . . . . . . . . . . . . . . . . . . . . . . 4-(cytidine 5'-diphospho)-2-C-methyl-D-erythritol kinase . . . . . . cytidylate kinase . . . . . . . . . . . . . . . . . . . . . . . .  (  )        $ " . . . . . . . deoxyguanosine kinase . . . . . . . . . . . . . . . . . . . . . deoxynucleoside kinase . . . . . . . . . . . . . . . . . . . . . diphosphate-purine nucleoside kinase. . . . . . . . . . . . . . . diphosphoinositol-pentakisphosphate kinase . . . . . . . . . . . . dolichyl-diphosphate-polyphosphate phosphotransferase . . . . . . dTMP kinase . . . . . . . . . . . . . . . . . . . . . . . . . farnesyl-diphosphate kinase . . . . . . . . . . . . . . . . . . . formate kinase . . . . . . . . . . . . . . . . . . . . . . . . . glutamate 1-kinase . . . . . . . . . . . . . . . . . . . . . . .

 2.7.2.8 2.7.1.156 2.7.4.11 2.7.4.3 2.7.1.147 2.7.1.146 2.7.1.118 2.7.1.126 2.7.3.10 2.7.3.8 2.7.1.114 2.7.3.3 2.7.2.4  2.7.2.14 2.7.2.7 2.7.1.123 2.7.1.120 2.7.2.2  ' 2.7.1.138 2.7.3.2 2.7.1.148 2.7.4.14   2.7.1.113 2.7.1.145 2.7.1.143 2.7.1.155 2.7.4.20 2.7.4.9 2.7.4.18 2.7.2.6 2.7.2.13

Page 259 358 123 #$% 342 255 572 493 226 223 50 90 424 411 15 385 314 #& 362 337 64 56 275 !!! ! % 192 369 229 582 & 1 214 208 252 611 555 606 334 360

XV

Index of Recommended Enzyme Names

2.7.2.11 2.7.1.142 2.7.3.1 2.7.4.8 2.7.1.119 2.7.3.6 2.7.4.21 $$ 2.7.1.151 2.7.1.134 2.7.1.140 2.7.1.127 $$!' $$!! 2.7.1.116 2.7.3.5 2.7.1.131 2.7.1.136 2.7.1.115 2.7.4.19 2.7.1.129 2.7.1.117 2.7.1.B1 2.7.4.6 2.7.4.13 2.7.4.4 2.7.4.10 2.7.3.7 2.7.1.137 2.7.1.150 2.7.1.153 2.7.1.154 2.7.1.149 2.7.1.121 2.7.3.9 $  2.7.2.3 2.7.2.10 ! 2.7.4.17  2.7.4.7

XVI

glutamate 5-kinase . . . . . . . . . . . . . . . . . . . . . glycerol-3-phosphate-glucose phosphotransferase . . . . . . . . guanidinoacetate kinase . . . . . . . . . . . . . . . . . . . guanylate kinase . . . . . . . . . . . . . . . . . . . . . . hygromycin-B kinase . . . . . . . . . . . . . . . . . . . . hypotaurocyamine kinase . . . . . . . . . . . . . . . . . . inositol-hexakisphosphate kinase . . . . . . . . . . . . . . .    ()   )     $" . . . inositol-polyphosphate multikinase . . . . . . . . . . . . . . inositol-tetrakisphosphate 1-kinase . . . . . . . . . . . . . . inositol-tetrakisphosphate 5-kinase . . . . . . . . . . . . . . inositol-trisphosphate 3-kinase . . . . . . . . . . . . . . . .      )    $$! " . . . .      #)    $$! " . . . . [Isocitrate dehydrogenase (NADP+ )] kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.5). . . . . . . . . . . . . . . . . lombricine kinase . . . . . . . . . . . . . . . . . . . . . . low-density-lipoprotein kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.29) . . . . . . . . . . . . . . . . . macrolide 2'-kinase . . . . . . . . . . . . . . . . . . . . . [3-methyl-2-oxobutanoate dehydrogenase (lipoamide)] kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.4) . . . 5-methyldeoxycytidine-5'-phosphate kinase . . . . . . . . . . myosin-heavy-chain kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.7) . . . . . . . . . . . . . . . . . . . . . . . . myosin-light-chain kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.18). . . . . . . . . . . . . . . . . . . . . . . NAD kinase . . . . . . . . . . . . . . . . . . . . . . . . nucleoside-diphosphate kinase . . . . . . . . . . . . . . . . (deoxy)nucleoside-phosphate kinase. . . . . . . . . . . . . . nucleoside-phosphate kinase . . . . . . . . . . . . . . . . . nucleoside-triphosphate-adenylate kinase . . . . . . . . . . . opheline kinase . . . . . . . . . . . . . . . . . . . . . . . phosphatidylinositol 3-kinase. . . . . . . . . . . . . . . . . 1-phosphatidylinositol-3-phosphate 5-kinase . . . . . . . . . . phosphatidylinositol-4,5-bisphosphate 3-kinase . . . . . . . . . phosphatidylinositol-4-phosphate 3-kinase . . . . . . . . . . . 1-phosphatidylinositol-5-phosphate 4-kinase . . . . . . . . . . phosphoenolpyruvate-glycerone phosphotransferase. . . . . . . phosphoenolpyruvate-protein phosphotransferase . . . . . . . .          " . . . . . . . . .          "     " . phosphoglycerate kinase . . . . . . . . . . . . . . . . . . . phosphoglycerate kinase (GTP) . . . . . . . . . . . . . . . .           $" . . . . . . . . 3-phosphoglyceroyl-phosphate-polyphosphate phosphotransferase          %" . . . . . . . . phosphomethylpyrimidine kinase . . . . . . . . . . . . . . .

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

351 206 365 543 52 407 613  & 236 155 197 107 $'# $

. .

28 403

. .

147 166

. .

19 609

.

129

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

34 257 521 578 517 567 409 170 234 241 245 231 60 414 #$ #$ 283 349 #$' 604 #! 539

Index of Recommended Enzyme Names

2.7.4.2 # 2.7.4.1 2.7.1.135

phosphomevalonate kinase . . . . . . . . . . . . . . . . . . .          " . . . . . . . . . . . polyphosphate kinase . . . . . . . . . . . . . . . . . . . . . . tau-protein kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.26) . . . . . . . . . . . . . . . . . . . . . . . . 2.7.3.11-12 protein-histidine kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.13.1) . . . . . . . . . . . . . . . . . . . . . . . . . . 2.7.3.11 protein-histidine pros-kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.13.2. See ec 2.7.3.11-12 for detailed organism-specific information). . . . . . . . . . . . . . . . . . . . . . . . . . 2.7.3.12 protein-histidine tele-kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.13.3. See EC 2.7.3.11-12 for detailed, organism-specific information). . . . . . . . . . . . . . . . . . . . . . . . . . 2.7.1.125 rhodopsin kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to to EC 2.7.11.14) . . . . . . . . . . . . . . . . . . . . . . . . . 2.7.1.141 [RNA-polymerase]-subunit kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.23) . . . . . . . . . . . . . . . . . . . 2.7.4.12 T2-induced deoxynucleotide kinase. . . . . . . . . . . . . . . . 2.7.1.144 tagatose-6-phosphate kinase . . . . . . . . . . . . . . . . . . . 2.7.3.4 taurocyamine kinase . . . . . . . . . . . . . . . . . . . . . . 2.7.1.130 tetraacyldisaccharide 4'-kinase . . . . . . . . . . . . . . . . . . 2.7.4.15 thiamine-diphosphate kinase . . . . . . . . . . . . . . . . . . 2.7.4.16 thiamine-phosphate kinase . . . . . . . . . . . . . . . . . . . 2.7.1.132 tropomyosin kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.28) . . . . . . . . . . . . . . . . . . . . . . . . . 2.7.1.124 [tyrosine 3-monooxygenase] kinase (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.6). . . . . . . . . . . . . . . . . . . . 2.7.1.122 xylitol kinase . . . . . . . . . . . . . . . . . . . . . . . . .

487 # 475 160 432

426

430 72 200 575 210 399 144 598 601 150 70 62

XVII

Description of Data Fields

#    #  ) 

All information except the nomenclature of the enzymes (which is based on the recommendations of the Nomenclature Committee of IUBMB (International Union of Biochemistry and Molecular Biology) and IUPAC (International Union of Pure and Applied Chemistry) is extracted from original literature (or reviews for very well characterized enzymes). The quality and reliability of the data depends on the method of determination, and for older literature on the techniques available at that time. This is especially true for the fields *   +  and  

. The general structure of the fields is: Information ± Organism ± Commentary ± Literature The information can be found in the form of numerical values (temperature, pH, Km etc.) or as text (cofactors, inhibitors etc.). Sometimes data are classified as ,    . Here you may find data that cannot be recalculated to the units required for a field or also general information being valid for all values. For example, for    , ,      may contain a list of compounds that are not inhibitory. The detailed structure and contents of each field is described below. If one of these fields is missing for a particular enzyme, this means that for this field, no data are available.

* (    +   The number is as given by the IUBMB, classes of enzymes and subclasses defined according to the reaction catalyzed.     This is the name as given by the IUBMB/IUPAC Nomenclature Committee '     This is the name as given by the IUBMB/IUPAC Nomenclature Committee   Synonyms which are found in other databases or in the literature, abbreviations, names of commercially available products. If identical names are frequently used for different enzymes, these will be mentioned here, cross references are given. If another EC number has been included in this entry, it is mentioned here.

XIX

Description of Data Fields

+    The majority of enzymes have a single chemical abstract (CAS) number. Some have no number at all, some have two or more numbers. Sometimes two enzymes share a common number. When this occurs, it is mentioned in the commentary.

,    -  For listing organisms their systematic name is preferred. If these are not mentioned in the literature, the names from the respective literature are used. For example if an enzyme from yeast is described without being specified further, yeast will be the entry. This field defines the code numbers for the organisms in which the enzyme with the respective EC number is found. These code numbers (form ) are displayed together with each entry in all fields of BRENDA where organism-specific information is given.

 '       +      The reaction as defined by the IUBMB. The commentary gives information on the mechanism, the stereochemistry, or on thermodynamic data of the reaction. '    According to the enzyme class a type can be attributed. These can be oxidation, reduction, elimination, addition, or a name (e.g. Knorr reaction) (          These are substrates and products which are metabolized in vivo. A natural substrate is only given if it is mentioned in the literature. The commentary gives information on the pathways for which this enzyme is important. If the enzyme is induced by a specific compound or growth conditions, this will be included in the commentary. In ,     you will find comments on the metabolic role, sometimes only assumptions can be found in the references or the natural substrates are unknown. In the listings, each natural substrate (indicated by a bold S) is followed by its respective product (indicated by a bold P). Products are given with organisms and references included only if the respective authors were able to demonstrate the formation of the specific product. If only the disappearance of the substrate was observed, the product is included without organisms of references. In cases with unclear product formation only a ? as a dummy is given.        All natural or synthetic substrates are listed (not in stoichiometric quantities). The commentary gives information on the reversibility of the reaction,

XX

Description of Data Fields

on isomers accepted as substrates and it compares the efficiency of substrates. If a specific substrate is accepted by only one of several isozymes, this will be stated here. The field ,     summarizes compounds that are not accepted as substrates or general comments which are valid for all substrates. In the listings, each substrate (indicated by a bold S) is followed by its respective product (indicated by a bold P). Products are given with organisms and references included if the respective authors demonstrated the formation of the specific product. If only the disappearance of the substrate was observed, the product will be included without organisms or references. In cases with unclear product formation only a ? as a dummy is given. %   Compounds found to be inhibitory are listed. The commentary may explain experimental conditions, the concentration yielding a specific degree of inhibition or the inhibition constant. If a substance is activating at a specific concentration but inhibiting at a higher or lower value, the commentary will explain this. +   .     This field contains cofactors which participate in the reaction but are not bound to the enzyme, and prosthetic groups being tightly bound. The commentary explains the function or, if known, the stereochemistry, or whether the cofactor can be replaced by a similar compound with higher or lower efficiency.  "  +    This field lists compounds with a positive effect on the activity. The enzyme may be inactive in the absence of certain compounds or may require activating molecules like sulfhydryl compounds, chelating agents, or lipids. If a substance is activating at a specific concentration but inhibiting at a higher or lower value, the commentary will explain this. / .   This field lists all metals or ions that have activating effects. The commentary explains the role each of the cited metal has, being either bound e.g. as Fe-S centers or being required in solution. If an ion plays a dual role, activating at a certain concentration but inhibiting at a higher or lower concentration, this will be given in the commentary. $ "   0- *1 The kcat is given in the unit min-1 . The commentary lists the names of the substrates, sometimes with information on the reaction conditions or the type of reaction if the enzyme is capable of catalyzing different reactions with a single substrate. For cases where it is impossible to give the turnover number in the defined unit (e.g., substrates without a defined molecular weight, or an undefined amount of protein) this is summarized in ,    .

XXI

Description of Data Fields

  " 021 The unit is micromol/minute/milligram of protein. The commentary may contain information on specific assay conditions or if another than the natural substrate was used in the assay. Entries in ,     are included if the units of the activity are missing in the literature or are not calculable to the obligatory unit. Information on literature with a detailed description of the assay method may also be found. 34  0/1 The unit is mM. Each value is connected to a substrate name. The commentary gives, if available, information on specific reaction condition, isozymes or presence of activators. The references for values which cannot be expressed in mM (e.g. for macromolecular, not precisely defined substrates) are given in ,    . In this field we also cite literature with detailed kinetic analyses. 34  0/1 The unit of the inhibition constant is mM. Each value is connected to an inhibitor name. The commentary gives, if available, the type of inhibition (e.g. competitive, non-competitive) and the reaction conditions (pH-value and the temperature). Values which cannot be expressed in the requested unit and references for detailed inhibition studies are summerized under ,     . 4- The value is given to one decimal place. The commentary may contain information on specific assay conditions, such as temperature, presence of activators or if this optimum is valid for only one of several isozymes. If the enzyme has a second optimum, this will be mentioned here. 4'  Mostly given as a range e.g. 4.0±7.0 with an added commentary explaining the activity in this range. Sometimes, not a range but a single value indicating the upper or lower limit of enzyme activity is given. In this case, the commentary is obligatory. $   0 +1 Sometimes, if no temperature optimum is found in the literature, the temperature of the assay is given instead. This is always mentioned in the commentary. $    0 +1 This is the range over which the enzyme is active. The commentary may give the percentage of activity at the outer limits. Also commentaries on specific assay conditions, additives etc.

XXII

Description of Data Fields

5    /    6 This field gives the molecular weight of the holoenzyme. For monomeric enzymes it is identical to the value given for subunits. As the accuracy depends on the method of determination this is given in the commentary if provided in the literature. Some enzymes are only active as multienzyme complexes for which the names and/or EC numbers of all participating enzymes are given in the commentary.   The tertiary structure of the active species is described. The enzyme can be active as a monomer a dimer, trimer and so on. The stoichiometry of subunit composition is given. Some enzymes can be active in more than one state of complexation with differing effectivities. The analytical method is included.           The main entries in this field may be proteolytic modification, or side-chain modification, or no modification. The commentary will give details of the modifications e.g.: ± proteolytic modification (, propeptide Name) [1]; ± side-chain modification (, N-glycosylated, 12% mannose) [2]; ± no modification [3]

7 %    2     2 /   2       2  For multicellular organisms, the tissue used for isolation of the enzyme or the tissue in which the enzyme is present is given. Cell-lines may also be a source of enzymes. !    The subcellular localization is described. Typical entries are: cytoplasm, nucleus, extracellular, membrane.    The field consists of an organism and a reference. Only references with a detailed description of the purification procedure are cited. '    Commentary on denaturant or renaturation procedure. +    The literature is cited which describes the procedure of crystallization, or the X-ray structure.

XXIII

Description of Data Fields

+  Lists of organisms and references, sometimes a commentary about expression or gene structure.  The properties of modified proteins are described.    Actual or possible applications in the fields of pharmacology, medicine, synthesis, analysis, agriculture, nutrition are described.

8   4  This field can either give a range in which the enzyme is stable or a single value. In the latter case the commentary is obligatory and explains the conditions and stability at this value. $    This field can either give a range in which the enzyme is stable or a single value. In the latter case the commentary is obligatory and explains the conditions and stability at this value. -&     Stability in the presence of oxidizing agents, e.g. O2, H2 O2, especially important for enzymes which are only active under anaerobic conditions. -   "   The stability in the presence of organic solvents is described. 9        This field summarizes general information on stability, e.g., increased stability of immobilized enzymes, stabilization by SH-reagents, detergents, glycerol or albumins etc.      Storage conditions and reported stability or loss of activity during storage.

'  Authors, Title, Journal, Volume, Pages, Year.

XXIV

    



     2.7.1.113     ATP:deoxyguanosine 5'-phosphotransferase    deoxyguanosine kinase   

(dihydroxypropoxymethyl)guanine kinase 2'-deoxyguanosine kinase NTP-deoxyguanosine 5'-phosphotransferase deoxyadenosine kinase/deoxyguanosine kinase ( abbrevation: dAK/ dGK [16]) [16] kinase, deoxyguanosine (phosphorylating)     39471-28-8

  





Mus musculus (3 days old [1]) [1] Bos taurus (calf [2]) [2, 3, 6, 14] Homo sapiens [5, 7, 8, 9, 11, 12, 14, 15, 20] Sus scrofa (piglet [4]) [4] Mus musculus (mitochondrial deoxyguanosine kinase 1 [10]) [10] Mus musculus (cytosolic deoxyguanosine kinase 2, amino-terminally truncated isoform [10]) [10] Bacillus subtilis [13] Lactobacillus acidophilus (strain R-26, heterodimeric enzyme, deoxyadenosine kinase/deoxguanosine kinase [16]) [16, 17, 18] Lactobacillus acidophilus (strain R-26, heterodimeric enzyme, deoxyadenosine kinase/deoxguanosine kinase, i.e. dAK/dGK [19] SwissProt-ID: U01881) [19]

1

Deoxyguanosine kinase

2.7.1.113

       !   "   ATP + deoxyguanosine = ADP + dGMP ( sequential Bi Bi mechanism [13])     phospho group transfer      

 ATP + deoxyguanosine ( first reaction in reutilization of deoxyguanosine for dGTP biosynthesis [1]; enzyme of purine deoxynucleoside salvage pathway [2]) (Reversibility: ? [1, 2]) [1, 2] # ADP + dGMP [1, 2]  ATP + deoxyinosine ( recombinant mitochondrial deoxyguanosine kinase, may be the preferred substrate in vivo [7]) (Reversibility: ? [7]) [7] # ADP + dIMP [7]     

 ATP + 2',2'-difluorodeoxyguanosine ( recombinant mitochondrial deoxyguanosine kinase, 147% of activity with deoxyguanosine [10]) (Reversibility: ? [7]) [10] # ADP + 2',2'-difluorodeoxyguanosine 5'-monophosphate [10]  ATP + 2',3'-dideoxguanosine ( recombinant deoxyguanosine kinase, 14% of activity with deoxyguanosine [14]) (Reversibility: ? [14]) [14] # ADP + 2',3'-dideoxyguanosine 5'-monophosphate [14]  ATP + 2-chloro-2'-arabino-fluoro-2'-deoxyadenosine ( recombinant mitochondrial deoxyguanosine kinase [7]) (Reversibility: ? [7]) [7] # ADP + 2-chloro-2'-arabino-fluoro-2'-deoxyadenosine 5'-monophosphate [7]  ATP + 2-chloro-2'-deoxyadenosine ( recombinant mitochondrial deoxyguanosine kinase [7]; recombinant mitochondrial deoxyguanosine kinase, 200% of activity with deoxyguanosine at 0.005 mM [10]) (Reversibility: ? [7,10]) [7, 10, 14] # ADP + 2-chloro-2'-deoxyadenosine monophosphate [7, 10, 14]  ATP + 2-fluoro-arabinosyl-adenine ( recombinant mitochondrial deoxyguanosine kinase [7]) (Reversibility: ? [7]) [7] # ADP + 2-fluoro-arabinosyl-adenine 5'-monophosphate [7]  ATP + 3'-fluoro-2',3'-dideoxguanosine ( recombinant deoxyguanosine kinase, 14% of activity with deoxyguanosine [14]) (Reversibility: ? [14]) [14] # ADP + 3'-fluoro-2',3'-dideoxguanosine 5'-monophosphate [14]  ATP + 9-(1,3-dihydroxy-2-propoxymethyl)guanine ( antiherpesvirus drug ganciclovir, recombinant mitochondrial deoxyguanosine ki-

2

2.7.1.113

# 

# 

#  #  # 

# 

# 

# 

# 

Deoxyguanosine kinase

nase, 6% of activity with deoxyguanosine [14]) (Reversibility: ? [14]) [14] ADP + 9-(1,3-dihydroxy-2-propoxymethyl)guanine 5'-monophosphate [14] ATP + 9-(4-hydroxy-3-hydroxymethylbutyl-1-yl)guanine ( antiherpesvirus drug penciclovir, recombinant mitochondrial deoxyguanosine kinase, 50% of activity with deoxyguanosine [14]) (Reversibility: ? [14]) [14] ADP + 9-(4-hydroxy-3-hydroxymethylbutyl-1-yl)guanine 5'-monophosphate [14] ATP + 9-b-d-arabinofuranosylguanine ( guanosine analog with activity in patients with T-cell malignancies [15]) (Reversibility: ? [15]) [15] ADP + 9-b-d-arabinofuranosylguanine 5'-monophosphate [15] ATP + 9-b-d-arabinosylguanine (Reversibility: ? [20]) [20] ADP + 9-b-d-arabinosylguanine 5'-monophosphate [20] ATP + arabinosyl adenine ( recombinant mitochondrial deoxyguanosine kinase, very low activity [7]) (Reversibility: ? [7]) [7] ADP + arabinosyl adenine 5'-monophosphate [7] ATP + arabinosyl guanine ( recombinant mitochondrial deoxyguanosine kinase, better substrate than deoxyguanosine kinase [7]) (Reversibility: ? [7]) [7] ADP + arabinosyl guanine 5'-monophosphate [7] ATP + deoxyadenosine ( 4.8%, 25.8% and 93.5% of activity with deoxyguanosine at 0.01 mM, 0.1 mM and 1 mM deoxyguanosine, respectively [5]; 3% of activity with deoxyguanosine [13]) (Reversibility: ? [5,13]) [5, 13] ADP + dAMP [5] ATP + deoxyadenosine ( recombinant mitochondrial deoxyguanosine kinase [7]; recombinant mitochondrial deoxyguanosine kinase, very low activity [10]) (Reversibility: ? [7,10]) [7, 10] ADP + dAMP [7, 10] ATP + deoxycytidine ( recombinant mitochondrial deoxyguanosine kinase [7]; recombinant deoxyguanosine kinase, 3% of activity with deoxyguanosine [13]) (Reversibility: ? [7,13]) [7, 13] ADP + dCMP [7] ATP + deoxyguanosine ( highly specific for phospho group acceptor [3,4]; ATP can be replaced by dTTP, CTP, dCTP and UTP [1,3,5]; 84%, 64%, 35% and 20% of activity with ATP with dTTP, CTP, dCTP and UTP, respectively [1]; ATP can be replaced by dUTP [5]; ATP most efficient at pH 5.5, dTTP or UTP most efficient, i.e. more efficient than ATP, at pH 7.4 [3]; ATP twice as effective as CTP or GTP and 4 times as effective as UTP [4]; poor donor substrates are dATP or GTP [3]; no activity with dATP or GTP [1]; no activity with deoxycytidine [1-3,5]; no activity with adenosine [1-3]; no activity with deoxythymidine, uridine and cytosine [1,2]; no activity with inosine and 9-b-arabinofuranosyladenine [2]; no ac3

Deoxyguanosine kinase

# 

#  # 

# 

#  #  #  # 

# 

#

tivity with dATP and GTP [5]; no activity with dGTP [1, 3, 5]; no activity with dITP [3]; recombinant mitochondrial deoxyguanosine kinase does not discriminate between b-d-2'-deoxyguanosine and bl-2'-deoxyguanosine [9]) (Reversibility: ? [1-5,10]) [1-5, 9, 10] ADP + dGMP [1-5, 9, 10] ATP + deoxyinosine ( recombinant mitochondrial deoxyguanosine kinase [7]; 10% of activity with deoxyguanosine [10]; 63% of activity with deoxyguanosine [13]) (Reversibility: ? [2, 7, 10, 13]) [2, 7, 10, 13] ADP + dIMP [2, 7, 10, 13] ATP + dideoxyinosine ( recombinant mitochondrial deoxyguanosine kinase, very low activity [7]) (Reversibility: ? [7]) [7] ADP + dideoxyinosine 5'-monophosphate [7] ATP + guanosine ( mitochondrial deoxyguanosine kinase [2]; recombinant deoxyguanosine kinase, 7% of activity with deoxyguanosine [13]) (Reversibility: [2,13]) [2, 13] ADP + GMP [2, 13] CTP + deoxyguanosine ( 64% of activity with ATP [1]; 50% of activity with ATP [4]; 62% of activity with ATP at pH 6.0, 1.8fold higher activity than with ATP at pH 7.0 [5]) (Reversibility: ? [1,3,4,5]) [1, 3, 4, 5] CDP + dGMP [1, 3, 4, 5] GTP + deoxyguanosine ( 50% of activity with ATP [4]) (Reversibility: ? [4]) [4] GDP + dGMP [4] UTP + deoxyguanosine ( 20% of activity with ATP [1]; 25% of activity with ATP [4]) (Reversibility: ? [1, 3, 4, 5]) [1, 3, 4, 5] UDP + dGMP [1, 3, 4, 5] dCTP + deoxyguanosine ( 35% of activity with ATP [1]) (Reversibility: ? [1,3,5]) [1, 3, 5] dCDP + dGMP [1, 3, 5] dTTP + deoxyguanosine ( 84% of activity with ATP [1]; 83% of activity with ATP at pH 6.0, 2.8fold higher activity than with ATP at pH 7.0 [5]) (Reversibility: ? [1,3,5]) [1, 3, 5] dTDP + dGMP [1, 3, 5] dUTP + deoxyguanosine ( 69% of activity with ATP at pH 6.0, 1.7fold higher activity than with ATP at pH 7.0 [5]) (Reversibility: ? [5]) [5] dUDP + dGMP [5]

$ %  

1,2-cyclohexanedione [3] 2,3-butanedione [3] 8-azadeoxyguanosine [3] ADP ( 1 mM, 45% inhibition at pH 6.0 [5]) [5] AMP ( weak inhibition [4]) [4] ATP ( weak inhibition [4]) [4]

4

2.7.1.113

2.7.1.113

Deoxyguanosine kinase

CMP ( weak inhibition [4]) [4] CTP [5] CuSO4 [5] EDTA [5] GDP ( 1 mM, 50% inhibition at pH 6.0 [5]) [5] GMP ( weak inhibition [4]) [4] GTP ( 0.1 mM, 33% inhibition [4]) [4, 5] IDP ( 1 mM, 58% inhibition at pH 6.0 [5]) [5] N-ethylmaleimide [3] Rose Bengal mediated photooxidation ( deoxyguanosine protects [3]) [3] UDP ( strong inhibition [1]) [1] UMP ( 0.1 mM, 24% inhibition [4]) [4] UTP ( 0.1 mM, 30% inhibition [4]) [4, 5] arabinoadenosine [5] arabinocytidine [5] arabinosylguanine [3] carbodiimide [3] dADP ( 1 mM, 49% inhibition at pH 6.0 [5]) [5] dATP ( 0.1 mM, 62% inhibition [4]; 1 mM, 71% inhibition at pH 6.0 [5]) [4, 5] dCDP [5] dCMP ( weak inhibition [4]) [4] dCTP ( 0.1 mM, 48% inhibition [4]) [4, 5] dGDP ( 0.0021 mM, 50% inhibition, reversed by 0.2 mM dTTP [5]; 0.1 mM, 97% inhibition [1]; 0.1 mM, 55% inhibition [2]) [1, 2, 5] dGMP ( very weak inhibition [2]; 0.1 mM, 44% inhibition [4]; 1 mM, 64% inhibition at pH 6.0 [5]) [1, 2, 4, 5] dGTP ( 0.001 mM, 50% inhibition, 1 mM, complete inhibition, reversed by 0.2 mM dTTP [5]; 0.1 mM, 99% inhibition [1]; 1 mM, 96% inhibition [2]; 0.1 mM, complete inhibition [4]; 0.5 mM, 95% of recombinant dGK [19]) [1, 2, 4, 5, 16, 19] dITP ( 0.1 mM, 65% inhibition [2]) [2] dTDP ( 1 mM, 51% inhibition at pH 6.0 [5]) [1, 5] dTMP ( weak inhibition [4]) [4] dTTP ( 0.1 mM, 30% inhibition [4]) [4] dUTP [5] deoxyadenosine ( weak inhibition [4]; 0.07 mM, 9% inhibition [1]) [1, 4, 5] deoxyinosine ( 0.07 mM, 17% inhibition [1]) [1, 3, 5] ethoxyformic anhydride ( deoxyguanosine slightly protects [3]) [3] guanosine [5] iodine [3] p-mercuribenzoate [3] ribavirin [5] Additional information ( not inhibited by dADP [2]; not inhibited by dAMP [4]; not inhibited by deoxycytidine, adenosine [3, 5]; 5

Deoxyguanosine kinase

2.7.1.113

not inhibited by inosine, cytidine, uridine, deoxythymidine, deoxyuridine [5]; not inhibited by acyclovir, 6-thiodeoxyguanosine, methylacetimidate, pyridoxal phosphate [3]; not inhibited by dATP [2]; not inhibited by dCTP [2]; not inhibited by dTTP [5]; not inhibited by GTP [2]; not inhibited by guanosine [3]; not inhibited by deoxyadenosine [1,3]) [1-5] 1-a-d-arabinofuranosylguanine [3]  &    

CTP ( activation, with ATP as substrate at pH 7 [5]) [5] Triton X-100 ( maximal activation at concentrations above critical micelle concentration [3]) [3] UDP ( 1 mM, 310% activation of mitochondrial deoxyguanosine kinase, with ATP as substrate [2]) [2] UTP ( 1 mM, 190% activation of mitochondrial deoxyguanosine kinase [2]; stimulation with ATP as substrate, at pH 7 [5]; slight inhibition at pH 6 [5]) [2, 5] dCDP ( slight stimulation, with ATP as substrate [2]) [2] dTDP ( 1 mM, 620% activation of mitochondrial deoxyguanosine kinase, positive modulation with ATP as substrate [2]) [2] dTTP ( 1 mM, 280% activation of mitochondrial deoxyguanosine kinase [2]; 1 mM, 3-4fold activation of deoxyguanosine kinase at pH 7.0 [5]) [2, 5] dUTP ( activation, with ATP as substrate, at pH 7 [5]) [5] sodium diphosphate ( 1 mM, 120% stimulation of mitochondrial deoxyguanosine kinase [2]) [2] Additional information ( not activated by dCTP or dATP, with ATP as substrate [2]; not activated by dTDP [5]) [2, 5] ' ( 

Ba2+ ( slight activation [5]) [5] Ca2+ ( activation [1,3-5]; 52% of activity with Mg2+ [3]; 60% of activity with Mg2+ [5]; 67% of activity with Mg2+ [4]) [1, 3-5] Cd2+ ( slight activation [1,3]) [1, 3] Co2+ ( 53% of activity with Mg2+ [3]; 59% of activity with Mg2+ [5]) [3, 5] Cr2+ ( slight activation [5]) [5] Cu2+ ( 25% of activity with Mg2+ [3]) [3] Fe2+ ( 67% of activity with Mg2+ [3]; 33% of activity with Mg2+ [5]; slight activation [4]) [3-5] Mg2+ ( required for activity [1-5,13]; MgNTP2- is most probably the true phosphate donor [13]) [1-5, 13] Mn2+ ( activation, as effective as Mg2+ [3-5]; can replace Mg2+ to some extent [1]; 92% of activity with Mg2+ [4]) [1, 3-5] Ni2+ ( slight activation [5]) [5] Zn2+ ( activation, can replace Mg2+ to some extent [1]; 28% of activity with Mg2+ [3]; 45% of activity with Mg2+ [5]) [1, 3, 5] 6

2.7.1.113

Deoxyguanosine kinase

Additional information ( divalent cations required, not activated by Zn2+ [4]; not activated by Cu2+ [5]) [4, 5] ) &  * +, 84.6 (deoxyguanosine, pH 8.5, 37 C, cosubstrate UTP [13]) [13] 91.8 (deoxyguanosine, pH 8.5, 37 C, cosubstrate ATP [13]) [13] 101.4 (deoxyguanosine, pH 8.5, 37 C, cosubstrate GTP [13]) [13] 135 (deoxyguanosine, pH 8.5, 37 C, cosubstrate CTP [13]) [13]   !   &  *-., 0.0000338 [4] 0.0007 [1] 0.0103 [5] 0.04-0.05 ( recombinant mitochondrial deoxyguanosine kinase [7]) [7] 8.1 ( recombinant deoxyguanosine kinase [13]) [13] /01 *', 0.00032 (deoxyguanosine, pH 8.0, 37 C [4]) [4] 0.0006 (deoxguanosine, pH 8.5, 37 C, cosubstrate UTP [13]) [13] 0.0025 (deoxyguanosine, pH 6.0, 37 C [5]) [5] 0.0028 (deoxyguanosine, pH 7.6, 37 C, recombinant deoxguanosine kinase, cosubstrate ATP [14]) [14] 0.0037 (deoxyguanosine, pH 7.6, 37 C, recombinant deoxguanosine kinase, cosubstrate UTP [14]) [14] 0.004 (deoxyguanosine, pH 7.6, recombinant mitochondrial deoxyguanosine kinase [7]) [7] 0.0047 (deoxyguanosine, pH 7.4, 37 C, mitochondrial deoxyguanosine kinase [3]) [3, 6] 0.006 (UTP, pH 8.5, 37 C [13]) [13] 0.006 (deoxyguanosine, pH 7.5, 37 C, mitochondrial deoxyguanosine kinase [2]) [2] 0.0065 (deoxguanosine, pH 8.5, 37 C, cosubstrate ATP [13]) [13] 0.007 (deoxyguanosine, pH 5.2, 37 C [1]) [1] 0.008 (2-chlorodeoxyadenosine, pH 7.6, 37 C, recombinant deoxyguanosine kinase, cosubstrate UTP [14]) [14] 0.01 (deoxguanosine, pH 8.5, 37 C, cosubstrate GTP [13]) [13] 0.012 (deoxyinosine, pH 7.6, recombinant mitochondrial deoxyguanosine kinase [7]) [7] 0.017 (deoxguanosine, pH 8.5, 37 C, cosubstrate CTP [13]) [13] 0.021 (deoxyinosine, pH 7.4, 37 C, mitochondrial deoxyguanosine kinase [3]) [3] 0.023 (MgATP2-, pH 5.2, 37 C, at concentrations below 0.5 mM [1]) [1] 0.033 (arabinosyl guanine, pH 7.6, recombinant mitochondrial deoxyguanosine kinase [7]) [7] 0.035 (CTP, pH 8.5, 37 C [13]) [13] 0.036 (ATP, pH 8.5, 37 C [13]) [13]

7

Deoxyguanosine kinase

2.7.1.113

0.046 (GTP, pH 8.5, 37 C [13]) [13] 0.056 (2-chloro-2'-arabino-fluoro-2'-deoxyadenosine, pH 7.6, recombinant mitochondrial deoxyguanosine kinase [7]) [7] 0.062 (2-chlorodeoxyadenosine, pH 7.6, 37 C, recombinant deoxguanosine kinase, cosubstrate ATP [14]) [14] 0.065 (deoxyguanosine, pH 8.0, 20 C [17]) [17] 0.078 (2-chloro-2'-deoxyadenosine, pH 7.6, recombinant mitochondrial deoxyguanosine kinase [7]) [7] 0.08 (dTTP, pH 7.4, 37 C, mitochondrial deoxyguanosine kinase [3]) [3] 0.087 (deoxyguanosine, pH 8.0, 20 C [16]) [16] 0.125 (UTP, pH 7.4, 37 C, mitochondrial deoxyguanosine kinase [3]) [3] 0.13 (MgATP2-, pH 6.0, 37 C [5]) [5] 0.34 (deoxycytidine, pH 7.6, recombinant mitochondrial deoxyguanosine kinase [7]) [7] 0.34 (deoxyguanosine, pH 8.0, 20 C, D84N mutant enzyme [16]) [16] 0.36 (deoxyguanosine, pH 8.0, 20 C, D84A mutant enzyme [16]) [16] 0.46 (2-fluoro-arabinosyl-adenine, pH 7.6, recombinant mitochondrial deoxyguanosine kinase [7]) [7] 0.47 (deoxyadenosine, pH 7.6, recombinant mitochondrial deoxyguanosine kinase [7]) [7] 0.55 (deoxyguanosine, pH 8.0, 20 C, D84E mutant enzyme [16]) [16] 0.63 (MgATP2-, pH 5.2, 37 C, at concentrations above 0.5 mM [1]) [1] 0.63 (deoxyadenosine, pH 6.0, 37 C [5]) [5] 0.76 (deoxyguanosine, pH 8.0, 20 C, R79K mutant enzyme [17]) [17] 0.78 (ATP, pH 7.4, 37 C, mitochondrial deoxyguanosine kinase [3]) [3, 6] 2.2 (MgdTTP2-, pH 6.0, 37 C [5]) [5] 3.3 (MgATP2-, pH 8.0, 37 C [4]) [4] / 01 *', 0.00003 (dGTP, pH 7.4, 37 C, vs. ATP, mitochondrial deoxyguanosine kinase [6]) [6] 0.00007 (dGTP, pH 5.2, 37 C, competitive vs. MgATP2-, at MgATP2concentrations above 0.5 mM [1]) [1] 0.0001 (dITP, pH 7.4, 37 C, vs. ATP, mitochondrial deoxyguanosine kinase [6]) [6] 0.0004 (dGTP, pH 7.6, 37 C, recombinant deoxguanosine kinase [14]) [14] 0.00052 (dGDP, pH 7.4, 37 C, vs. ATP, mitochondrial deoxyguanosine kinase [6]) [6] 0.0007 (dGDP, pH 5.2, 37 C, competitive vs. MgATP2-, at MgATP2concentrations above 0.5 mM [1]) [1]

8

2.7.1.113

Deoxyguanosine kinase

0.00093 (dGTP, pH 7.4, 37 C, vs. deoxyguanosine, mitochondrial deoxyguanosine kinase [6]) [6] 0.0019 (dGTP, pH 5.2, 37 C, competitive vs. deoxyguanosine [1]) [1] 0.0019 (dGTP, pH 8.0, 20 C, vs. ATP [17]) [17] 0.0026 (dITP, pH 7.4, 37 C, vs. deoxyguanosine, mitochondrial deoxyguanosine kinase [6]) [6] 0.003 (UDP, pH 5.2, 37 C, competitive vs. MgATP2-, at MgATP2concentrations above 0.5 mM [1]) [1] 0.00333 (dGTP, pH 8.0, 20 C, vs. deoxyguanosine [17]) [17] 0.004 (dGMP, pH 7.6, 37 C, recombinant deoxguanosine kinase [14]) [14] 0.0058 (dGMP, pH 7.4, 37 C, vs. ATP mitochondrial deoxyguanosine kinase [6]) [6] 0.0087 (dGDP, pH 7.4, 37 C, vs. deoxyguanosine, mitochondrial deoxyguanosine kinase [6]) [6] 0.009 (dGMP, pH 7.4, 37 C, vs. dTTP, mitochondrial deoxyguanosine kinase [6]) [6] 0.021 (dGTP, pH 8.0, 37 C [4]) [4] 0.028 (dAMP, pH 7.6, 37 C, recombinant deoxyguanosine kinase [14]) [14] 0.031 (dIMP, pH 7.4, 37 C, vs. ATP, mitochondrial deoxyguanosine kinase [6]) [6] 0.041 (dATP, pH 7.6, 37 C, recombinant deoxyguanosine kinase [14]) [14] 0.063 (UDP, pH 8.5, 37 C, cosubstrate UTP [13]) [13] 0.075 (deoxyinosine, pH 7.4, 37 C, competitive vs. deoxyguanosine, mitochondrial deoxyguanosine kinase [3]) [3] 0.076 (dGMP, pH 8.0, 37 C [4]) [4] 0.078 (dIMP, pH 7.6, 37 C, recombinant deoxyguanosine kinase [14]) [14] 0.098 (dIMP, pH 7.4, 37 C, vs. deoxyguanosine, mitochondrial deoxyguanosine kinase [6]) [6] 0.1 (dGMP, pH 7.4, 37 C, vs. deoxyguanosine, mitochondrial deoxyguanosine kinase [6]) [6] 0.117 (1-a-d-arabinofuranosylguanine, pH 7.4, 37 C, competitive vs. deoxyguanosine, mitochondrial deoxyguanosine kinase [3]) [3] 0.18 (dGMP, pH 5.2, 37 C, competitive vs. deoxyguanosine [1]) [1] 0.21 (dTDP, pH 7.4, 37 C, vs. ATP, mitochondrial deoxyguanosine kinase [6]) [6] 0.25 (dGMP, pH 7.4, 37 C, vs.ATP, mitochondrial deoxyguanosine kinase [6]) [6] 0.34 (deoxyadenosine, pH 8.0, 37 C [4]) [4] 0.39 (dTDP, pH 7.4, 37 C, vs. deoxyguanosine, mitochondrial deoxyguanosine kinase [6]) [6] 0.43 (8-aza-2'-deoxyguanosine, pH 7.4, 37 C, competitive vs. deoxyguanosine, mitochondrial deoxyguanosine kinase [3]) [3]

9

Deoxyguanosine kinase

2.7.1.113

0.73 (dTDP, pH 7.4, 37 C, vs. dTTP, mitochondrial deoxyguanosine kinase [6]) [6] 1 (ADP, pH 7.4, 37 C, noncompetitive vs. deoxyguanosine, mitochondrial deoxyguanosine kinase [6]) [6] 20   5.2 ( substrate deoxyguanosine [1]) [1] 5.5 [3] 6 ( cosubstrate ATP [5]) [5] 6.8 ( cosubstrate ATP, addition of dTTP [5]) [5] 7 ( cosubstrate dTTP [5]) [5] 7.4 ( cosubstrates dTTP or UTP [3]) [3] 8.5 [4] 9 ( broad optimum [13]) [13] 20  4.5-9 ( approx. 20% of maximal activity at pH 4.5 and pH 6.5 respectively, approx. 10% of maximal activity at pH 9.0 [1]) [1] 5 ( rapid decrease of activity below [3]) [3] 5-9 ( approx. 55% of maximal activity at pH 5.5 and pH 9, plateau with approx. 85% of maximal activity at pH 6.0-7.5 [4]) [4] 5.6-9 ( more than 80% of maximal activity at pH 7.5 and pH 11.5, respectively, 60% of maximal activity at pH 6.0, complete loss of activity below pH 5.6 [13]) [13] )     * , 37 [4] 37-40 [3] )    * , 30-53 ( 17% loss of activity at 53 C, 50% loss of activity at 30 C and 53 C [4]) [4]

3  "  ' 4 % 44000 ( glycerol density gradient centrifugation [1]) [1] 49000 ( recombinant deoxyguanosine kinase, gel filtration [13]) [13] 56000 ( mitochondrial deoxyguanosine kinase, equilibrium sedimentation centrifugation [3]) [3] 58000 ( gel filtration [5]) [5] 58500 ( gel filtration [4]) [4]  

dimer ( 2 * 28000, mitochondrial deoxyguanosine kinase, SDS-PAGE [3]; 2 * 29000, second mayor band at 35000 Da, SDS-PAGE [5]; 2 * 24147, electrospray mass spectrometry [13]; 2 * 27200, heterodimeric dAK/dGK complex, SDS-PAGE [18]) [3, 5, 13, 18]

10

2.7.1.113

5 $   .#   .'  .

Deoxyguanosine kinase

  

.

 brain ( low expression levels [10]) [10, 14] embryo ( human epithelial kidney 293 cells [11]) [11, 12] heart [14] kidney ( human epithelial kidney 293 cells [11]) [11, 12] liver ( low expression [10]) [3, 6, 10, 14] lymphoblast ( CEM cells [11, 20]; acute T lymphoblastic leukemia molt-4 cells [12]) [11, 12, 15, 20] pancreatic adenocarcinoma cell ( cell lines PanC-1 and MIA PaCa-2 [8]) [8] placenta [5] skin ( neonatal skin tissue [1]) [1, 4] spleen ( high expression levels [10]) [10] thymus ( high expression [10]) [2, 10, 14] 6 "  cytosol ( deoxyguanosine kinase 2 [10]) [10] mitochondrial matrix [11] mitochondrion ( membrane associated [3]; mitochondrial deoxguanosine kinase is redistributed to the cytosol during apoptosis [12]) [2, 3, 6, 7, 8, 10, 11, 12] soluble [1] # !   (pH 5.7, streptomycin sulfate, ammonium sulfate [1]) [1] (mitochondrial deoxyguanosine kinase, Sephacryl S-200, Blue Sepharose CL6B [2]; mitochondrial deoxyguanosine kinase, one-step purification via deoxyguanosine-3'-(4-aminophenylphosphate)-Sepharose affinity chromatography [3]) [2, 3] (ammonium sulfate, affinity chromatography on AMP-Sepharose and Blue-Sepharose, DEAE-cellulose, Sephadex G-75 [5]; recombinant enzyme, metal affinity chromatography [7]) [5, 7] (pH 5.5, ammonium sulfate, DEAE-Sephadex [4]) [4] (recombinant deoxyguanosine kinase, BlueA dye affinity, phenyl-Sepharose [13]) [13]   (expression in Escherichia coli [7]; expresssion of mitochondrial deoxyguanosine kinase-green fluorescent protein fusion in pancreatic cancer cells [8]) [7, 8, 10, 14] (expression in Escherichia coli [13]) [13] (expression of dAK/dGK in Escherichia coli [19]) [19] (expression of His-tagged deoxyguanosine kinase in Escherichia coli [10]) [10]

11

Deoxyguanosine kinase

2.7.1.113

    D78A ( no deoxyguanosine kinase activity [17]) [17] D78E ( no deoxyguanosine kinase activity [17]) [17] D78N ( no deoxyguanosine kinase activity [17]) [17] D84A ( 1% of wild-type activity [16]) [16] D84E ( 41% of wild-type activity, not inhibited by dGTP and dATP [16]) [16] D84N ( 5% of wild-type activity [16]) [16] R79k ( 50% of wild-type deoxyguanosine kinase activity [17]) [17]

7    20   5.5 ( 95% loss of activity after 30 min at 37 C [3]) [3] 7 ( most stable [3]) [3] 11 ( 50% loss of activity after 30 min at 37 C [3]) [3] )     37 ( at least 4 h stable in the presence of Triton X-100, t1=2 : 30 min at pH 11, inactivation within 30 min at pH 5.5 [3]) [3] 40 ( denaturation above 40 C, mitochondrial deoxyguanosine kinase [3]) [3] Additional information ( Triton X-100, 0.02%, stabilizes markedly against thermal inactivation [3]) [3] 8     !  , ATP or MgATP2- stabilize, not Mg2+ alone [1] , Triton X-100, 0.02%, stabilizes markedly against thermal inactivation [3] , complete inactivation after freezing at -20 C or -70 C [5] , highly unstable at 0 C in diluted form, no loss of activity in the presence of bovine serum albumin [13] , loss of activity in dilute solution [1, 5]     , -20 C, ammonium sulfate precipitate in 100 mM M Tris-acetate buffer, pH 8, 0.025 M 2-mercaptoethanol, at least 4 months [1] , 4 C, 1 week, 50% loss of activity [5] , -20 C, more than 1 mg/ml protein, 50% glycerol, 1 year, no loss of activity [13]

! 

[1] Barker, J.; Lewis, R.A.: Deoxyguanosine kinase of neonatal mouse skin tissue. Biochim. Biophys. Acta, 658, 111-123 (1981) [2] Gower, W.R.; Carr, M.C.; Ives, D.H.: Deoxyguanosine kinase. Distinct molecular forms in mitochondria and cytosol. J. Biol. Chem., 254, 2180-2183 (1979) 12

2.7.1.113

Deoxyguanosine kinase

[3] Park, I.; Ives, D.H.: Properties of a highly purified mitochondrial deoxyguanosine kinase. Arch. Biochem. Biophys., 266, 51-60 (1988) [4] Green, F.J.; Lewis, R.A.: Partial purification and characterization of deoxyguanosine kinase from pig skin. Biochem. J., 183, 547-553 (1979) [5] Yamada, Y.; Goto, H.; Ogasawara, N.: Deoxyguanosine kinase from human placenta. Biochim. Biophys. Acta, 709, 265-272 (1982) [6] Park, I.; Ives, D.H.: Kinetic mechanism and end-product regulation of deoxyguanosine kinase from beef liver mitochondria. J. Biochem., 117, 10581061 (1995) [7] Sjoberg, A.H.; Wang, L.; Eriksson, S.: Substrate specificity of human recombinant mitochondrial deoxyguanosine kinase with cytostatic and antiviral purine and pyrimidine analogs. Mol. Pharmacol., 53, 270-273 (1998) [8] Zhu, C.; Johansson, M.; Permert, J.; Karlsson, A.: Enhanced cytotoxicity of nucleoside analogs by overexpression of mitochondrial deoxyguanosine kinase in cancer cell lines. J. Biol. Chem., 273, 14707-14711 (1998) [9] Gaubert, G.; Gosselin, G.; Boudou, V.; Imbach, J.L.; Eriksson, S.; Maury, G.: Low enantioselectivities of human deoxycytidine kinase and human deoxyguanosine kinase with respect to 2'-deoxyadenosine, 2'-deoxyguanosine and their analogs. Biochimie, 81, 1041-1047 (1999) [10] Petrakis, T.G.; Ktistaki, E.; Wang, L.; Eriksson, S.; Talianidis, I.: Cloning and characterization of mouse deoxyguanosine kinase. Evidence for a cytoplasmic isoform. J. Biol. Chem., 274, 24726-24730 (1999) [11] Jullig, M.; Eriksson, S.: Mitochondrial and submitochondrial localization of human deoxyguanosine kinase. Eur. J. Biochem., 267, 5466-5472 (2000) [12] Jullig, M.; Eriksson, S.: Apoptosis induces efflux of the mitochondrial matrix enzyme deoxyguanosine kinase. J. Biol. Chem., 276, 24000-24004 (2001) [13] Andersen, R.B.; Neuhard, J.: Deoxynucleoside kinases encoded by the yaaG and yaaF genes of Bacillus subtilis. Substrate specificity and kinetic analysis of deoxyguanosine kinase with UTP as the preferred phosphate donor. J. Biol. Chem., 276, 5518-5524 (2001) [14] Herrstrom Sjoberg, A.; Wang, L.; Eriksson, S.: Antiviral guanosine analogs as substrates for deoxyguanosine kinase: implications for chemotherapy. Antimicrob. Agents Chemother., 45, 739-742 (2001) [15] Lotfi, K.; Mansson, E.; Peterson, C.; Eriksson, S.; Albertioni, F.: Low level of mitochondrial deoxyguanosine kinase is the dominant factor in acquired resistance to 9-b-d-arabinofuranosylguanine cytotoxicity. Biochem. Biophys. Res. Commun., 293, 1489-1496 (2002) [16] Park, I.; Ives, D.H.: Mutations within the putative active site of heterodimeric deoxyguanosine kinase block the allosteric activation of the deoxyadenosine kinase subunit. J. Biochem. Mol. Biol., 35, 244-247 (2002) [17] Hong, Y.S.; Ma, G.T.; Ives, D.H.: Directed mutagenesis of deoxyguanosine site at arginine 79 up-regulates turnover on deoxyadenosine kinase subunit of heterodimeric enzyme from Lactobacillus acidophilus R26. J. Biol. Chem., 270, 6602-6606 (1995) [18] Ikeda, S.; Ma, G.T.; Ives, D.H.: Heterodimeric deoxynucleoside kinases of Lactobacillus acidophilus R-26: functional assignment of subunits using 13

Deoxyguanosine kinase

2.7.1.113

limited proteolysis controlled by end-product inhibitors. Biochemistry, 33, 5328-5334 (1994) [19] Ma, G.T.; Hong, Y.S.; Ives, D.H.: Cloning and expression of the heterodimeric deoxyguanosine kinase/deoxyadenosine kinase of Lactobacillus acidophilus R-26. J. Biol. Chem., 270, 6595-6601 (1995) [20] Rodriguez, C.O., Jr.; Mitchell, B.S.; Ayres, M.; Eriksson, S.; Gandhi, V.: Arabinosylguanine is phosphorylated by both cytoplasmic deoxycytidine kinase and mitochondrial deoxyguanosine kinase. Cancer Res., 62, 31003105 (2002)

14

'#0)%    

3

     2.7.1.114     AMP:thymidine 5'-phosphotransferase    AMP-thymidine kinase   

AMP:dThd kinase AMP:deoxythymidine 5'-phosphotransferase AMP:deoxythymidine kinase adenylate-nucleoside phosphotransferase adenylic acid:deoxythymidine 5'-phosphotransferase thymidine phosphotransferase     60440-28-0

   Herpes simplex (type 1 [1,3]; hamster BHK cells infected with virus [1]) [1, 3] Asplenium nidus (bird's nest fern [2]) [2] Hordeum vulgare (barley [2]) [2] Helianthus tuberosus (Jerusalem artichoke [2]) [2] Epidendrum hybrid (crucifix orchid [2]) [2] Medicago sativa (lucerne [2]) [2]

       !   "   AMP + thymidine = adenosine + thymidine 5'-phosphate     phospho group transfer

15

AMP-Thymidine kinase

2.7.1.114

     

 AMP + thymidine (Reversibility: ? [1-3]) [1-3] # adenosine + dTMP     

 AMP + 5-bromodeoxyuridine (Reversibility: ? [2]) [2] # adenosine + 5-bromodeoxyuridine 5'-phosphate  AMP + adenosine (Reversibility: ? [2]) [2] # adenosine + AMP  AMP + cytidine (Reversibility: ? [2]) [2] # adenosine + cytidine 5'-phosphate  AMP + deoxyuridine (Reversibility: ? [2]) [2] # adenosine + deoxyuridine 5'-phosphate  AMP + guanosine (Reversibility: ? [2]) [2] # adenosine + guanosine 5'-phosphate  AMP + thymidine ( AMP can be substituted by GMP, dAMP, CMP, UMP, dUMP, dTMP [2]) (Reversibility: ? [1-3]) [1-3] # adenosine + dTMP [1, 2]  AMP + uridine (Reversibility: ? [2]) [2] # adenosine + uridine 5'-phosphate $ %  

1,10-phenanthroline [2] 3',5'-cAMP [2] 9-b-d-arabinofuranosyladenine 5'-monophosphate [3] CoCl2 [2] MnCl2 [2] NiCl2 [2] adenosine [2] uridine [2]   !   &  *-., 0.002 ( leaf [2]) [2] 0.01 ( stem [2]) [2] 0.05 ( roots [2]) [2] 0.05 ( leaf [2]) [2] 0.06 ( tops [2]) [2] 0.07 ( senescent leaf [2]) [2] 0.29 ( mature leaf [2]) [2] 0.47 ( young and expanded leaf [2]) [2] 0.84 ( mature leaf [2]) [2] 0.91 ( young mature leaf [2]) [2] 1.09 ( immature leaf [2]) [2] 1.64 ( young bud [2]) [2] 42.2 [1] /01 *', 0.0021 (thymidine, 37 C, pH 7.8 [1]) [1] 0.0023 (AMP, 37 C, pH 7.8 [1]) [1] 16

2.7.1.114

AMP-Thymidine kinase

0.11 (AMP, 38 C, pH 7.5 [2]) [2] 0.84-0.88 (thymidine, 38 C, pH 7.5 [2]) [2] 1.28 (adenosine, 38 C, pH 7.5 [2]) [2] 4.71 (uridine, 38 C, pH 7.5 [2]) [2] 20   7.6 [1] 20  6.3-8.8 ( 17% of maximal activity at pH 6.3, 32% of maximal activity at pH 8.8 [1]) [1] )     * , 37-38 [1]

3  "  ' 4 % 90600-111000 ( glycerol density gradient centrifugation [1]) [1]

5 $   .#   .'  .

  

.

 leaf [2] pollen [2] root [2] stem [2] # !   (type 1, hamster BHK-cells infected with virus [1]) [1] (partial [2]) [2]

7    )     37 ( 60 min, 95% loss of activity [2]) [2] 63 ( 5 min, complete inactivation [2]) [2] 8     !  , bovine serum albumin stabilizes [2] , not stabilized by AMP or thymidine [2]     , 0 C, 6 months, 20% loss of activity [2]

17

AMP-Thymidine kinase

2.7.1.114

! 

[1] Falke, D.; Nehrbass, W.; Brauer, D.; Müller, W.E.G.: Adenylic acid: deoxythymidine 5-phosphotransferase: evidence for the existence of a novel Herpes simplex virus-induced enzyme. J. Gen. Virol., 53, 247-255 (1981) [2] Grivell, A.R.; Jackson, J.F.: Thymidine phosphotransferase and nucleotide phosphohydrolase of the fern Asplenium nidus. General properties and inhibition by adenosine 3:5-cyclic monophosphate. Biochem. J., 155, 571-581 (1976) [3] Labenz, J.; Müller, W.E.G.; Falke, D.: Inhibition of the Herpes simplex viruscoded thymidine kinase-complex by 9-b-d-arabinofuranosyladenine 5monophosphate (ara-AMP) and 9-(2-hydroxyethoxymethyl)guanine-monophosphate (acyclo-GMP). Arch. Virol., 81, 205-212 (1984)

18

90'%0 0  %   *  ,:  

5

     2.7.1.115 (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.4)     ATP:[3-methyl-2-oxobutanoate dehydrogenase (lipoamide)] phosphotransferase    [3-methyl-2-oxobutanoate dehydrogenase (lipoamide)] kinase   

BCK BCKDH kinase branched-chain 2-oxo acid dehydrogenase kinase branched-chain a-keto acid dehydrogenase kinase branched-chain a-ketoacid dehydrogenase kinase branched-chain keto acid dehydrogenase kinase kinase, branched-chain oxo acid dehydrogenase (phosphorylating) Additional information ( kinase activity is an intrinsic activity of branched-chain oxo acid dehydrogenase complex [3]) [3]     82391-38-6

   Oryctolagus cuniculus [1, 2, 7-9, 11-13] Rattus norvegicus (Sprague-Dawley [14,21]; female Sprague-Dawley [26]; male Wistar [28]; clone 9 cells express higher amounts of the enzyme after insulin treatment [25]) [1, 3, 7, 10-14, 16, 18, 21-28] Bos taurus [1, 4-6, 12, 13, 15, 17] Mus musculus (mouse, C57BL/6J [19, 20]) [19, 20]

       !   "   ATP + [3-methyl-2-oxobutanoate dehydrogenase (lipoamide)] = ADP + [3methyl-2-oxobutanoate dehydrogenase (lipoamide)] phosphate 19

[3-Methyl-2-oxobutanoate dehydrogenase (lipoamide)] kinase

2.7.1.115

    phospho group transfer      

 ATP + [3-methyl-2-oxobutanoate dehydrogenase (lipoamide)] ( phosphorylation inactivates EC 1.2.4.4 [1-15, 17, 18, 24, 26]; branched-chain amino acid metabolism [11, 24]; regulatory enzyme of branched-chain 2-oxoacid dehydrogenase complex [15, 24]) (Reversibility: ? [1-15, 17, 18, 24, 26]) [1-15, 17, 18, 24, 26] # ADP + [3-methyl-2-oxobutanoate dehydrogenase (lipoamide)] phosphate     

 ATP + [3-methyl-2-oxobutanoate dehydrogenase (lipoamide)] ( phosphorylates exclusively MW 47000 subunit of substrate [2]; phosphorylates a-subunit of multienzyme complex component E1 [4, 5]; phosphorylates a-subunit of multienzyme complex component E1 and additional sites not associated with inactivation of the enzyme [6]; Ser-residues of MW 46000-subunit [3, 4]; 2 Ser-residues in E1-a-subunit [1, 11-13]; incorporates 0.8 mol phosphate/mol a-subunit [5]; incorporates 0.75 mol phosphate per mol phosphorylation site and 1.5 mol/mol a-subunit [13]; GTP cannot replace ATP [2]; tight binding to multienzyme complex is required for phosphorylation, free enzyme is inactive [26]) (Reversibility: ? [1-18, 20, 21, 22, 24, 26, 27, 28]) [1-18, 20, 21, 22, 24, 26, 27, 28] # ADP + [3-methyl-2-oxobutanoate dehydrogenase (lipoamide)] phosphate [1, 3-6, 11-13]  ATP + histone II-S (Reversibility: ? [14, 27]) [14, 27] # ?  Additional information ( R288A mutant of E1 is not phosphorylated by the enzyme [22]; enzyme has also ATPase activity in absence of E1 [23]) [22, 23] # ? $ %  

2-(N-morpholino)propane sulfonate buffer [12] 2-chloroisohexanoate ( i.e. 2-chloro-4-methylpentanoate, strong [9]; (R)(+)-isomer is twice as effective as (S)(-)-isomer [11]; site-specific inhibitor, [11]; enhanced by monovalent cations and further enhanced by phosphate [10]; potassium phosphate increases sensitivity to this inhibitor [11]; ATP does not protect [9]; no inhibition by (R)(-)-2-chloroisopentanoate [11]; 50% inhibition at 0.014 mM, no inhibition with histone II-S as substrate [14]; 50% inhibition at 0.014 mM [27]) [8-11, 13, 14, 27] 2-oxo-3-methylpentanoate ( more effective than 2-oxoisopentanoate [11]) [8, 11, 13] 2-oxobutanoate [13] 2-oxohexanedioate [12, 13] 2-oxohexanoate [8, 13]

20

2.7.1.115

[3-Methyl-2-oxobutanoate dehydrogenase (lipoamide)] kinase

2-oxoisocaproate ( kinetics, 40% inhibition at 0.065 mM [8]; more effective than 2-oxo-3-methylpentanoate and 2-oxoisopentanoate [11]) [4, 8, 11, 13] 2-oxoisopentanoate ( less effective than 2-oxoisohexanoate and 2-oxo-3-methylpentanoate [11]) [8, 11, 13] 2-oxopentanoate ( kinetics [8]) [8, 12, 13] 3-methyl-2-oxobutanoate [4] 4-(2-thienyl)-2-oxo-3-butenoate ( 2 mM [15]) [15] 4-(3-thienyl)-2-oxo-3-butenoate ( 2 mM [15]) [15] 4-hydroxyphenylacetate [7] 4-hydroxyphenyllactate ( weak [7]) [7] 4-hydroxyphenylpyruvate ( very weak: 3-hydroxyphenylpyruvate [7]) [7, 13] 4-methyl-2-oxopentanoate [4] ADP ( kinetics [2]; 50% inhibition at 0.4 mM, inhibition can be reversed by 2 mM Mg2+ [1]; competitive [13]; product inhibition [23]) [1, 2, 4, 12, 13, 23] ATP ( 50% inhibition at 0.2 mM, inhibition can be reversed by 2 mM Mg2+ [1]) [1, 3] CDP ( 50% inhibition at 0.4 mM, inhibition can be reversed by 2 mM Mg2+ [1]) [1] CTP ( 50% inhibition at 0.25 mM, inhibition can be reversed by 2 mM Mg2+ [1]) [1] Ca2+ (weak [13]) [2, 13] CoA [1] GDP ( 50% inhibition at 0.2 mM, inhibition can be reversed by 2 mM Mg2+ [1]) [1] GTP ( 50% inhibition at 0.06 mM, inhibition can be reversed by 2 mM Mg2+ [1]) [1] Mg2+ ( at concentrations above 1.5 mM, activation below [13]) [13] MgATP2- [4] NADP+ ( 40% inhibition at 1.5 mM [8]) [8, 13] UDP ( 50% inhibition at 0.25 mM, inhibition can be reversed by 2 mM Mg2+ [1]) [1] UTP ( 50% inhibition at 0.1 mM, inhibition can be reversed by 2 mM Mg2+ [1]) [1] acetate ( weak, in vivo and in vitro [7]) [7] acetoacetyl-CoA ( 40% inhibition at 0.01 mM [8]) [8, 12, 13] acyl-CoA [1] branched-chain 2-oxo acids [1, 4] clofibric acid ( in vivo and in vitro [7]) [7, 13] dichloroacetate ( ATP slightly protects [2]; weak [11]; 50% inhibition at 1.8 mM [14, 27]) [2, 7, 9, 11, 13, 14, 27] diphosphate [4] furfurylidenepyruvate ( 1.85 mM [15]) [15] heparin ( 50% inhibition at 0.002 mM [1]; 40% inhibition at 0.012 mg/ml [8]) [1, 8, 13] 21

[3-Methyl-2-oxobutanoate dehydrogenase (lipoamide)] kinase

2.7.1.115

isobutyryl-CoA [8, 13] isovaleryl-CoA [8, 13] malonyl-CoA [8, 13] methylmalonyl-CoA ( 40% inhibition at 0.2 mM [8]) [8, 12, 13] n-octanoate ( 40% inhibition at 0.5 mM [8]) [8, 12, 13] phenylacetate ( strong [7]) [7, 13] phenyllactate ( strong [7]) [7, 13] phenylpyruvate ( in vivo and in vitro [7]) [7] pyruvate ( weak [7]) [7, 9, 13] thiamine [4] thiamine diphosphate ( inhibits phosphorylation of wild-type E1, mutant E1-S303A and mutant E1-D296A/S303A, but not phosphorylation of mutant E1-H292A [22]) [1, 4, 12, 22] Additional information ( no inhibition by lactate [7, 8]; no inhibition by GTP [2]; no inhibition by coenzyme A [1, 4, 8]; no inhibition by acetyl-CoA [4, 8]; no inhibition by NADH, NAD+ 1 mM each [8]; no inhibition by methylcrotonyl-CoA, b-hydroxy-b-methylglutarylCoA, crotonyl-CoA, octanoyl-CoA, succinyl-CoA, propionyl-CoA, 0.1 mM each, propionate, b-hydroxybutyrate, acetoacetate, malonate, a-ketomalonate, succinate, citrate, oxaloacetate, FAD+, NADPH, 2 mM [8]; no inhibition by isovaleryl-CoA [1]; no inhibition by dl-leucine [9]; no inhibition by 2-chloropropionate [11]; no inhibition by acetate [8]) [1, 2, 4, 7-9, 11] ! .  %  

calmodulin ( activation [2]) [2]  &    

histone H3 ( 1.5 to 3fold [1]) [1] poly-l-arginine ( 1.5 to 3fold [1]) [1] poly-l-lysine ( 1.5 to 3fold [1]) [1] protamine ( 1.5 to 3fold [1]) [1] ' ( 

EGTA ( activation, presumably by chelation of Ca2+ [13]; 0.1 mM [2]) [2, 13] K+ ( activation, 0.1 M [10]) [10] Mg2+ ( requirement, actual substrate: MgATP2- [2, 4, 5, 10, 12]; Km -value: 0.025 mM [2]; maximum activity at 1.5 mM, inhibits above 1.5 mM [13]) [2, 4, 5, 10, 12, 13] Rb+ ( activation [10]) [10] Additional information ( no activation by Ca2+ [2]; no activation by Li+ , Na+ [10]) [2, 10] ) &  * +, 3.25 (phosphate, 25 C, recombinant enzyme alone [18]) [18] 28.5 (phosphate, 25 C, reconstituted with lipoylated recombinant E2 [18]) [18]

22

2.7.1.115

[3-Methyl-2-oxobutanoate dehydrogenase (lipoamide)] kinase

  !   &  *-., 0.0247 ( without added salt [10]) [10] 0.0268 ( liver enzyme [14,27]) [14, 27] 0.0357 ( heart enzyme [14]) [14] 0.0357-0.09 ( heart enzyme, depending on purification method [27]) [27] 0.05 ( recombinant enzyme [27]) [27] Additional information ( various assay methods [12]) [12] /01 *', 0.004 (ATP) [10] 0.0126 (MgATP2-, pH 7.5, 30 C [4]) [4] 0.013 (MgATP2-, pH 7.5, 30 C [12]) [12] 0.025 (ATP, pH 7.5, 30 C [2]; pH 7.35, 20 C [13]) [2, 13] / 01 *', 0.00027 (ADP, pH 7.5, 30 C [4]) [4] 0.00048 (4-methyl-2-oxopentanoate, pH 7.5, 30 C [4]) [4] 0.00092 (2-oxoisocaproate, pH 7.5, 30 C [4]) [4] 0.004 (diphosphate, pH 7.5, 30 C [4]) [4] 0.0059 (thiamine, pH 7.5, 30 C [4]) [4] 0.0089 (4-methyl-2-oxopentanoate, pH 7.5, 30 C [4]) [4] 0.13 (ADP, pH 7.35, 20 C [13]) [13] 0.13 (ADP, pH 7.5, 30 C [2]) [2] 0.27 (ADP, pH 7.5, 30 C [12]) [12] 0.5 (2-chloroisohexanoate, 37 C [11]) [11] 4.5 (furfurylidenepyruvate) [15] 20   7.1 [2] 7.4 ( assay at [24]) [24] 7.5 [13] Additional information ( HEPES-potassium buffer promotes higher activity than imidazole-chloride, 4-morpholinopropanesulfonic acid-potassium or potassium phosphate buffer [2]; in decreasing order of activity: HEPES, potassium phosphate, imidazole, 3-(N-morpholino)ethane buffer [13]) [2, 13] 20  6.5-8.3 ( about half-maximal activity at pH 6.5 and 8.3 [2]) [2] )     * , 30 ( assay at [4, 14, 24]) [4, 14, 24] 37 ( assay at [2, 3, 8-10, 15]) [2, 3, 8-10, 15]

23

[3-Methyl-2-oxobutanoate dehydrogenase (lipoamide)] kinase

2.7.1.115

3  "  ' 4 % 43280 ( calculated from amino acid sequence [16]) [16] 44000-45000 ( SDS-PAGE [27]) [27] 460000 ( gel filtration [1]) [1] 2000000 ( above 2000000, gel filtration [2,13]) [2, 13]  

? ( x * 44000, SDS-PAGE [14]) [14] dimer ( dimerizes through direct interaction of two opposing nucleotide-binding domains, crystallographic data [23]) [23] monomer ( 1 * 43000, uncomplexed kinase, SDS-PAGE [17]) [17]

5 $   .#   .'  .

  

.

 adipocyte [12] brain [13, 19, 20] embryo [20] heart [7, 12-14, 16, 19, 20, 23, 27, 28] hepatocyte [11] kidney ( cortex [13]) [1, 3-6, 12, 13, 17, 18, 19, 20, 28] liver ( enzyme activity is 3-5fold higher in female than in male rats [24]; malnutrition results in changed amounts of enzyme level [28]) [2, 7-15, 19, 24, 26, 27, 28] lung [19] muscle [19, 20] skeletal muscle ( enzyme content decreases 0.7fold after running exercise for 5 weeks [21]) [13, 21, 24] testis [19] uterus [19] 6 "  mitochondrial matrix ( 2 forms: first form is bound to E2, second form is free and seems to be inactive [26]) [10, 26] mitochondrion ( part of intramitochondrial branched-chain 2oxoacid dehydrogenase complex [13]) [1, 3-5, 12, 13, 15-17, 19, 20, 25, 28] # !   (a-ketoacid dehydrogenase complex [2,13]) [2, 7, 13] (a-ketoacid dehydrogenase complex [3]; from liver and heart, homogeneity [12]; from heart [15]; from purified branched-chain a-keto acid dehydrogenase complex [14]; liver enzyme, heart enzyme and recombinant enzyme expressed in Escherichia coli [27]) [3, 12-14, 18, 27] (copurifies with EC 1.2.4.4 [5]; 5000fold [17]) [4, 5, 17]

24

2.7.1.115

[3-Methyl-2-oxobutanoate dehydrogenase (lipoamide)] kinase

   (reconstitution with lipoylated recombinant E2 [18]) [18]   "  (vapor diffusion method [23]) [23]   (cloned and expressed in Escherichia coli [16]; fusion protein with maltose-binding protein [18, 23]; fragments of the enzyme cloned into firefly luciferase plasmid [25]) [16, 18, 23, 27] [19, 20]

7    20   7 ( loss of activity during purification at pH-values below 7 [4]) [4] 8     !  , precipitation of branched-chain oxo acid dehydrogenase enzyme complex at acid pH-values, especially below 6.5, results in specific loss of kinase activity [12]     , labile enzyme, best stored at -70 C in the presence of DTT [14]

! 

[1] Reed, L.J.; Damuni, Z.; Merryfield, M.L.: Regulation of mammalian pyruvate and branched-chain a-keto acid dehydrogenase complexes by phosphorylation-dephosphorylation. Curr. Top. Cell. Regul., 27, 41-49 (1985) [2] Paxton, R.; Harris, R.A.: Isolation of rabbit liver branched chain a-ketoacid dehydrogenase and regulation by phosphorylation. J. Biol. Chem., 257, 14433-14439 (1982) [3] Odessey, R.: Purification of rat kidney branched-chain oxo acid dehydrogenase complex with endogenous kinase activity. Biochem. J., 204, 353-356 (1982) [4] Lau, K.S.; Fatania, H.R.; Randle, P.J.: Regulation of the branched chain 2oxoacid dehydrogenase kinase reaction. FEBS Lett., 144, 57-62 (1982) [5] Lawson, R.; Cook, K.G.; Yeaman, S.J.: Rapid purification of bovine kidney branched-chain 2-oxoacid dehydrogenase complex containing endogenous kinase activity. FEBS Lett., 157, 54-58 (1982) [6] Cook, K.G.; Lawson, R.; Yeaman, S.J.: Multi-site phosphorylation of bovine kidney branched-chain 2-oxoacid dehydrogenase complex. FEBS Lett., 157, 59-62 (1982) [7] Paxton, R.; Harris, R.A.: Clofibric acid, phenylpyruvate, and dichloroacetate inhibition of branched-chain a-ketoacid dehydrogenase kinase in vitro and in perfused rat heart. Arch. Biochem. Biophys., 231, 58-66 (1984) 25

[3-Methyl-2-oxobutanoate dehydrogenase (lipoamide)] kinase

2.7.1.115

[8] Paxton, R.; Harris, R.A.: Regulation of branched-chain a-ketoacid dehydrogenase kinase. Arch. Biochem. Biophys., 231, 48-57 (1984) [9] Harris, R.A.; Paxton, R.; DePaoli-Roach, A.: Inhibition of branched chain a-ketoacid dehydrogenase kinase activity by a-chloroisocaproate. J. Biol. Chem., 257, 13915-13918 (1982) [10] Shimomura, Y.; Kuntz, M.J.; Suzuki, M.; Ozawa, T.; Harris, R.A.: Monovalent cations and inorganic phosphate alter branched-chain a-ketoacid dehydrogenase-kinase activity and inhibitor sensitivity. Arch. Biochem. Biophys., 266, 210-218 (1988) [11] Harris, R.A.; Kuntz, M.J.; Simpson, R.: Inhibition of branched-chain a-keto acid dehydrogenase kinase by a-chloroisocaproate. Methods Enzymol., 166, 114-123 (1988) [12] Espinal, J.; Beggs, M.; Randle, P.J.: Assay of branched-chain a-keto acid dehydrogenase kinase in mitochondrial extracts and purified branchedchain a-keto acid dehydrogenase complexes. Methods Enzymol., 166, 166175 (1988) [13] Paxton, R.: Branched-chain a-keto acid dehydrogenase and its kinase from rabbit liver and heart. Methods Enzymol., 166, 313-320 (1988) [14] Shimomura, Y.; Nanaumi, N.; Suzuki, M.; Popov, K.M.; Harris, R.A.: Purification and partial characterization of branched-chain a-ketoacid dehydrogenase kinase from rat liver and rat heart. Arch. Biochem. Biophys., 283, 293-299 (1990) [15] Lau, K.S.; Cooper, A.J.L.; Chuang, D.T.: Inhibition of the bovine branchedchain 2-oxo acid dehydrogenase complex and its kinase by arylidenepyruvates. Biochim. Biophys. Acta, 1038, 360-366 (1990) [16] Popov, K.M.; Zhao, Y.; Shimomura, Y.; Kuntz, M.J.; Harris, R.A.: Branchedchain a-ketoacid dehydrogenase kinase. Molecular cloning, expression, and sequence similarity with histidine protein kinases. J. Biol. Chem., 267, 13127-13130 (1992) [17] Lee, H.Y.; Hall, T.B.; Kee, S.M.; Tung, H.Y.L.; Reed, L.J.: Purification and properties of branched-chain a-keto acid dehydrogenase kinase from bovine kidney. BioFactors, 3, 109-112 (1991) [18] Davie, J.R.; Wynn, R.M.; Meng, M.; Huang, Y.S.; Aalund, G.; Chuang, D.T.; Lau, K.S.: Expression and characterization of branched-chain a-ketoacid dehydrogenase kinase from the rat. Is it a histidine-protein kinase?. J. Biol. Chem., 270, 19861-19867 (1995) [19] Doering, C.B.; Coursey, C.; Spangler, W.; Danner, D.J.: Murine branched chain a-ketoacid dehydrogenase kinase; cDNA cloning, tissue distribution, and temporal expression during embryonic development. Gene, 212, 213219 (1998) [20] Doering, C.B.; Danner, D.J.: Expression of murine branched-chain a-keto acid dehydrogenase kinase. Methods Enzymol., 324, 491-497 (2000) [21] Fujii, H.; Shimomura, Y.; Murakami, T.; Nakai, N.; Sato, T.; Suzuki, M.; Harris, R.A.: Branched-chain a-keto acid dehydrogenase kinase content in rat skeletal muscle is decreased by endurance training. Biochem. Mol. Biol. Int., 44, 1211-1216 (1998)

26

2.7.1.115

[3-Methyl-2-oxobutanoate dehydrogenase (lipoamide)] kinase

[22] Hawes, J.W.; Schnepf, R.J.; Jenkins, A.E.; Shimomura, Y.; Popov, K.M.; Harris, R.A.: Roles of amino acid residues surrounding phosphorylation site 1 of branched-chain a-ketoacid dehydrogenase (BCKDH) in catalysis and phosphorylation site recognition by BCKDH kinase. J. Biol. Chem., 270, 31071-31076 (1995) [23] Machius, M.; Chuang, J.L.; Wynn, R.M.; Tomchick, D.R.; Chuang, D.T.: Structure of rat BCKD kinase: nucleotide-induced domain communication in a mitochondrial protein kinase. Proc. Natl. Acad. Sci. USA, 98, 1121811223 (2001) [24] Nakai, N.; Kobayashi, R.; Popov, K.M.; Harris, R.A.; Shimomura, Y.: Determination of branched-chain a-keto acid dehydrogenase activity state and branched-chain a-keto acid dehydrogenase kinase activity and protein in mammalian tissues. Methods Enzymol., 324, 48-62 (2000) [25] Nellis, M.M.; Doering, C.B.; Kasinski, A.; Danner, D.J.: Insulin increases branched-chain a-ketoacid dehydrogenase kinase expression in Clone 9 rat cells. Am. J. Physiol., 283, E853-E860 (2002) [26] Obayashi, M.; Sato, Y.; Harris, R.A.; Shimomura, Y.: Regulation of the activity of branched-chain 2-oxo acid dehydrogenase (BCODH) complex by binding BCODH kinase. FEBS Lett., 491, 50-54 (2001) [27] Popov, K.M.; Shimomura, Y.; Hawes, J.W.; Harris, R.A.: Branched-chain aketo acid dehydrogenase kinase. Methods Enzymol., 324, 162-178 (2000) [28] Popov, K.M.; Zhao, Y.; Shimomura, Y.; Jaskiewicz, J.; Kedishvili, N.Y.; Irwin, J.; Goodwin, G.W.; Harris, R.A.: Dietary control and tissue specific expression of branched-chain a-ketoacid dehydrogenase kinase. Arch. Biochem. Biophys., 316, 148-154 (1995)

27

9$   %   *#; ,:  

7

     2.7.1.116 (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.5)     ATP:[isocitrate dehydrogenase (NADP+ )] phosphotransferase    [isocitrate dehydrogenase (NADP+ )] kinase   

ICDH kinase/phosphatase [8] IDH kinase [1] IDH kinase/phosphatase [4, 5] IDH-K/P [12] IDHK/P [9, 11] [isocitrate dehydrogenase (NADP+ )] kinase isocitrate dehydrogenase kinase (phosphorylating) isocitrate dehydrogenase kinase/phosphatase [7, 9-11]     83682-93-3

   Escherichia coli (strain W3550 and mutants [5]; ML308, ATCC 15224 [2, 3, 8]; KAT-1/pEM9 [4]; ST2010R [6]; K-12 [7, 8]; JM109 [9,11]) [2-13] Salmonella typhimurium [1]

       !   "   ATP + [isocitrate dehydrogenase (NADP+ )] = ADP + [isocitrate dehydrogenase (NADP+ )] phosphate     phospho group transfer

28

2.7.1.116

[Isocitrate dehydrogenase (NADP+)] kinase

     

 ATP + [isocitrate dehydrogenase (NADP+ )] ( reversible phosphorylation of isocitrate dehydrogenase plays a major role in the control of the Krebs cycle and glyoxylate pathways [1, 5, 6]; phosphorylation of isocitrate dehydrogenase during growth on acetate is to render this enzyme rate-limiting in the citric acid cycle, this should cause an increase in the level of isocitrate and divert the flux of carbon through the glyoxylate bypass [3, 13]; controls the oxidative metabolism, exibits a high intrinsic ATPase activity [9]) (Reversibility: ? [1]; r [2-13]) [1-13] # ADP + [isocitrate dehydrogenase (NADP+ )] phosphate [1-13]     

 ATP + Bacillus subtilis [isocitrate dehydrogenase (NADP+ )] ( BsIDH is a much poorer substrate for the enzyme than EcIDH [12, 13]) (Reversibility: ? [12,13]) [12, 13] # ADP + Bacillus subtilis [isocitrate dehydrogenase (NADP+ )] phosphate  ATP + [isocitrate dehydrogenase (NADP+ )] (Reversibility: ? [1]; r [2-13]) [1-13] # ADP + [isocitrate dehydrogenase (NADP+ )] phosphate [1-13]  ATP + [isocitrate dehydrogenase (NADP+ )]IS (Reversibility: ? [10]) [10] # ADP + [isocitrate dehydrogenase (NADP+ )]IS phosphate  ATP + [isocitrate dehydrogenase (NADP+ )]N115L (Reversibility: ? [10]) [10] # ADP + [isocitrate dehydrogenase (NADP+ )]N115L phosphate  Additional information ( uses only ATP, no other nucleoside triphospates as only very poor phosphate donors for the kinase activity, GTP and UTP can activate the phosphatase activity to some extent [3]) [3] # ? $ %  

2-oxoglutarate ( inhibits kinase activity [3]) [3] 5,5'-dithio-bis(2-nitrobenzoic acid) [9] 8-azido-ATP [4] ADP ( kinase hyperbolically inhibited [3]) [3] AMP ( kinase hyperbolically inhibited [3]) [3, 10] dl-isocitrate ( inhibits only kinase activity [3,10]) [3, 10] NADPH ( inhibits both IDH kinase and IDH phosphatase [10]) [3, 10] [isocitrate dehydrogenase (NADP+ )] phosphate ( wild-type [6]) [6] cupric 1,10 phenanthrolinate [9] glyoxylate ( in combination with oxaloacetate [1]) [1, 3] oxaloacetate ( inhibits kinase activity [3]) [1, 3] phosphoenolpyruvate ( kinase hyperbolically inhibited [3]) [3] pyruvate ( inhibits kinase activity [3]) [1, 3, 10]

29

[Isocitrate dehydrogenase (NADP+)] kinase

2.7.1.116

! .  %  

ADP ( isocitrate dehydrogenase phosphatase requires a nucleotide for activity [3]) [3] ATP ( isocitrate dehydrogenase phosphatase requires a nucleotide for activity [3]) [3]  &    

acetate [1] a-methylglucoside [1] deoxyglucose [1] ethanol [1] ' ( 

Mg2+ ( absolute requirement, isocitrate dehydrogenase phosphatase responds hyperbolically to Mg2+ ions [3]) [3, 11] Additional information ( Mn2+ or Ca2+ cannot replace Mg2+ [3]) [3]   !   &  *-., 0.038 [2] 0.11-0.63 [5] /01 *', 0.00023 ([isocitrate dehydrogenase (NADP+ )], pH 7.5, 37 C, wildtype and mutant AceK4, kinase activity [6]) [6] 0.00025 ([isocitrate dehydrogenase (NADP+ )], pH 7.5, 37 C, mutant AceK3, kinase activity [6]) [6] 0.00035 ([isocitrate dehydrogenase (NADP+ )], pH 7.3, 37 C, kinase activity [3]) [3] 0.00078 ([isocitrate dehydrogenase (NADP+ )], pH 7.5, 37 C, kinase activity at saturating ATP [13]) [13] 0.0009 ([isocitrate dehydrogenase (NADP+ )]N15L, pH 7.5, 37 C [10]) [10] 0.0017 ([isocitrate dehydrogenase (NADP+ )], pH 7.5, 37 C [10]) [10] 0.0049 (Bacillus subtilis [isocitrate dehydrogenase (NADP+ )], pH 7.5, 37 C [13]) [13] 0.0059 ([isocitrate dehydrogenase (NADP+ )], pH 7.5, 37 C [13]) [13] 0.0069 (ATP, pH 7.5, 37 C, wild-type [11]) [11] 0.0087 (ATP, pH 7.5, 37 C, mutant Asp403Ala [11]) [11] 0.0098 (ATP, pH 7.5, 37 C, mutant Glu439Ala [11]) [11] 0.0147 (ATP, pH 7.5, 37 C, mutant Asn377Ala [11]) [11] 0.016 (ATP, pH 7.5, 37 C, wild-type, kinase activity [6]) [6] 0.02 ([isocitrate dehydrogenase (NADP+ )]IS, pH 7.5, 37 C [10]) [10] 0.0582 (Bacillus subtilis [isocitrate dehydrogenase (NADP+ )], pH 7.5, 37 C, kinase activity at saturating ATP [13]) [13] 0.088 (ATP, pH 7.3, 37 C [3]) [3] 0.1 (ATP, pH 7.5, 37 C, mutant AceK3, kinase activity [6]) [6] 0.32 (ATP, pH 7.5, 37 C, mutant AceK4, kinase activity [6]) [6] 30

2.7.1.116

[Isocitrate dehydrogenase (NADP+)] kinase

/ 01 *', 0.008 (AMP, pH 7.5, 37 C [10]) [10] 0.011 (isocitrate, pH 7.5, 37 C, mutant AceK4, kinase activity [10]) [10] 0.015 (isocitrate, pH 7.5, 37 C, mutant AceK3, kinase activity [10]) [10] 0.016 (isocitrate, pH 7.5, 37 C, wild-type, kinase activity [10]) [10] 0.02 (AMP, pH 7.5, 37 C, mutant AceK4, kinase activity [10]) [10] 0.023 (dl-isocitrate, pH 7.3, 37 C [3]) [3] 0.042 (NADPH, pH 7.3, 37 C [3]) [3] 0.056 (AMP, pH 7.3, 37 C [3]) [3] 0.058 (NADPH, pH 7.5, 37 C, mutant AceK4, kinase activity [10]) [10] 0.073 (NADPH, pH 7.5, 37 C, mutant AceK3, kinase activity [10]) [10] 0.082 (NADPH, pH 7.5, 37 C, wild-type, kinase activity [10]) [10] 0.17 (AMP, pH 7.5, 37 C, mutant AceK3, kinase activity [10]) [10] 0.2 (pyruvate, pH 7.5, 37 C, wild-type, kinase activity [10]) [10] 0.45 (ADP, pH 7.3, 37 C [3]) [3] 0.55 (phosphoenolpyruvate, pH 7.3, 37 C [3]) [3] 1 (3-phosphoglycerate, pH 7.5, 37 C, wild-type, kinase activity [10]) [10] 1 (pyruvate, pH 7.5, 37 C, mutant AceK4, kinase activity [10]) [10] 4 (3-phosphoglycerate, pH 7.5, 37 C, mutant AceK4, kinase activity [10]) [10] 4 (pyruvate, pH 7.5, 37 C, mutant AceK3, kinase activity [10]) [10] 20 (3-phosphoglycerate, pH 7.5, 37 C, mutant AceK3, kinase activity [10]) [10] 20   8-8.5 ( kinase activity [3]) [3]

3  "  ' 4 % 130000 ( recombinant enzyme, gel filtration [7]) [7] 135000 ( gel filtration, glycerol density gradient centrifugation [2]) [2]  

dimer ( 2 * 66000, homodimer, SDS-PAGE [2, 9, 11]; 2 * 68800, homodimer, theoretical molecular mass [9]) [2, 7, 9, 11]

31

[Isocitrate dehydrogenase (NADP+)] kinase

5 $   .#   .'  .

2.7.1.116

  

# !   (partial, bifunctional protein [2,4,5]) [2, 4-7, 9]   (bifunctional protein, expressed from the aceK gene [5]; aceK gene [6]; aceK gene of Escherichia coli K-12 cloned in pQE30 expression vector to overproduce the protein in Escherichia coli JM105 [7]; recombinant wild-type IDHK/P on overproducing plasmid pJCD4, expressed in Escherichia coli JM109 [9]) [5-7, 9]     D403A ( site-directed mutagenesis [11]) [11] E439A ( site-directed mutagenesis [11]) [11] N377A ( site-directed mutagenesis [11]) [11]

7        , -20 C, stable for at least 3 months [2] , 4 C, can be stored for several days without significant loss of activity [2]

! 

[1] Wang, J.Y.J.; Koshland, D.E.: The reversible phosphorylation of isocitrate dehydrogenase of Salmonella typhimurium. Arch. Biochem. Biophys., 218, 59-67 (1982) [2] Nimmo, G.A.; Borthwick, A.C.; Holms, W.H.; Nimmo, H.G.: Partial purification and properties of isocitrate dehydrogenase kinase/phosphatase from Escherichia coli ML308. Eur. J. Biochem., 141, 401-408 (1984) [3] Nimmo, G.A.; Nimmo, H.G.: The regulatory properties of isocitrate dehydrogenase kinase and isocitrate dehydrogenase phosphatase from Escherichia coli ML308 and the roles of these activities in the control of isocitrate dehydrogenase. Eur. J. Biochem., 141, 409-414 (1984) [4] Varela, I.; Nimmo, H.G.: Photoaffinity labelling shows that Escherichia coli isocitrate dehydrogenase kinase/phosphatase contains a single ATP-binding site. FEBS Lett., 231, 361-365 (1988) [5] Ikeda, T.P.; Houtz, E.; LaPorte, D.C.: Isocitrate dehydrogenase kinase/phosphatase: identification of mutations which selectively inhibit phosphatase activity. J. Bacteriol., 174, 1414-1416 (1992) [6] Miller, S.P.; Karschnia, E.J.; Ikeda, T.P.; LaPorte, D.C.: Isocitrate dehydrogenase kinase/phosphatase. Kinetic characteristics of the wild-type and two mutant proteins. J. Biol. Chem., 271, 19124-19128 (1996)

32

2.7.1.116

[Isocitrate dehydrogenase (NADP+)] kinase

[7] Rittinger, K.; Negre, D.; Divita, G.; Scarabel, M.; Bonod-Bidaud, C.; Goody, R.S.; Cozzone, A.J.; Cortay, J.C.: Escherichia coli isocitrate dehydrogenase kinase/phosphatase. Overproduction and kinetics of interaction with its substrates by using intrinsic fluorescence and fluorescent nucleotide analogues. Eur. J. Biochem., 237, 247-254 (1996) [8] El-Mansi, E.M.T.: Control of metabolic interconversion of isocitrate dehydrogenase between the catalytically active and inactive forms in Escherichia coli. FEMS Microbiol. Lett., 166, 333-339 (1998) [9] Oudot, C.; Jaquinod, M.; Cortay, J.C.; Cozzone, A.J.; Jault, J.M.: The isocitrate dehydrogenase kinase/phosphatase from Escherichia coli is highly sensitive to in-vitro oxidative conditions role of cysteine67 and cysteine108 in the formation of a disulfide-bonded homodimer. Eur. J. Biochem., 262, 224-229 (1999) [10] Miller, S.P.; Chen, R.; Karschnia, E.J.; Romfo, C.; Dean, A.; LaPorte, D.C.: Locations of the regulatory sites for isocitrate dehydrogenase kinase/phosphatase. J. Biol. Chem., 275, 833-839 (2000) [11] Oudot, C.; Cortay, J.C.; Blanchet, C.; Laporte, D.C.; Di Pietro, A.; Cozzone, A.J.; Jault, J.M.: The ªcatalyticª triad of isocitrate dehydrogenase kinase/ phosphatase from E. coli and its relationship with that found in eukaryotic protein kinases. Biochemistry, 40, 3047-3055 (2001) [12] Singh, S.K.; Matsuno, K.; LaPorte, D.C.; Banaszak, L.J.: Crystal structure of Bacillus subtilis isocitrate dehydrogenase at 1.55 A. Insights into the nature of substrate specificity exhibited by Escherichia coli isocitrate dehydrogenase kinase/phosphatase. J. Biol. Chem., 276, 26154-26163 (2001) [13] Singh, S.K.; Miller, S.P.; Dean, A.; Banaszak, L.J.; LaPorte, D.C.: Bacillus subtilis isocitrate dehydrogenase. A substrate analogue for Escherichia coli isocitrate dehydrogenase kinase/phosphatase. J. Biol. Chem., 277, 7567-7573 (2002)

33

' 0 %0%  



     2.7.1.117 (Protein kinases are in a state of review by the NC-IUBMB. This EC class will presumably be transferred to EC 2.7.11.18)     ATP:myosin-light-chain O-phosphotransferase    myosin-light-chain kinase   

calcium/calmodulin-dependent myosin light chain kinase kinase, myosin light-chain (phosphorylating) myosin kinase myosin light chain protein kinase myosin light-chain kinase smooth-muscle-myosin-light-chain kinase stretchin-MLCK [44] Additional information ( not identical with or immunologically related to protein kinase II from rat [11]) [11]     51845-53-5

  









34

Oryctolagus cuniculus (New Zealand White [2]) [1-5, 20, 21, 23, 46, 48, 49] Gallus gallus [5, 8, 12, 17, 21-23, 27, 28, 31-33, 35-41, 43, 50] Meleagris gallopavo [8, 14, 18-20, 23, 26, 29, 30, 33] Dictyostelium discoideum (strain Ax-3 [24]) [24] Rattus norvegicus [5, 16, 20, 33, 43] Homo sapiens [6, 14, 19, 20, 33, 34, 42, 51] Sus scrofa [15, 20, 47] Bos taurus (steer [33]) [5, 7-13, 20, 23, 33, 34, 42] Ovis aries (pregnant and non-pregnant [14]) [14] Canis familiaris [33] Cavia porcellus [33] Limulus sp. (horseshoe crab [25]) [25] Drosophila melanogaster [44] Oncorhynchus mykiss [45]

2.7.1.117

Myosin-light-chain kinase

       !   "   ATP + myosin light chain = ADP + myosin light chain phosphate     phospho group transfer      

 ATP + myosin light chain ( event in initiation of smoothmuscle contraction [5]; involved in regulation of actin-myosin contractile activity in adrenal medulla [7]; obligatory step in development of active tension in smooth muscle [13]; involved in myosin phosphorylation and enzyme secretion [16]; involved in muscle contractility and motility of non-muscle cells [33]; inhibition of actin-myosin ineraction [36,37]) (Reversibility: ? 1-12 [133]) [1-33] # ADP + myosin light chain phosphate     

 ATP + BpaKKRAARATSNVFA ( Bpa is the photoreactive amino acid p-benzoylphenylalanine [46]) (Reversibility: ? [46]) [46] # ADP + ?  ATP + KKRAARATSNVFA (Reversibility: ? [46]) [46] # ADP + ?  ATP + Lys-Lys-Arg-Ala-Ala-Arg-Ala-Thr-Ser-Asn-Val-Phe-Ala (Reversibility: ? [17]) [17] # ADP + ?  ATP + Lys-Lys-Arg-Pro-Gln-Arg-Ala-Thr-Ser-Asn-Val-Phe-Ser (Reversibility: ? [28]) [28] # ADP + ?  ATP + kemptamide (Reversibility: ? [11]) [11] # ADP + ?  ATP + myosin light chain ( highly specific for regulatory or P-light-chain [1, 2, 9, 11, 15, 20, 21]; highly specific for 18 kDa light chain [1, 2, 9, 11, 15, 16]; specific for 18.5 kDa Dictyostelium or 19 kDa skeletal muscle light chain [20]; specific for 20 kDa light chain [1, 2, 6, 7, 9, 11, 15, 16, 18-21, 25, 30]; not specific for 22 kDa and 15 kDa light chain [2, 33]; not 16 kDa light chain [7, 19, 24]; kinase from skeletal muscle with broader specificity than smooth muscle kinase [33]; acceptor substrates are myosin light chains of cardiac muscle [2, 9, 10, 18-21]; acceptor substrates are myosin light chains of skeletal muscle [1-3, 7, 9, 11, 18-21, 33]; acceptor substrates are myosin light chains of smooth muscle [2, 5, 6, 9, 11, 12, 21, 23, 28, 30, 33]; acceptor substrates are myosin light chains of adrenal medullary myosin [7]; acceptor substrates are myosin light chains of Ml3-myosin rabbit muscle [1]; acceptor

35

Myosin-light-chain kinase

#  #  # 

#

substrates are myosin light chains of non-muscle myosin [33]; acceptor substrates are myosin light chains of smooth muscle myosin [18, 19, 23, 28-30]; 1 mol phosphate per mol light chain in skeletal muscle [7, 21, 25]; transfers the g-phosphate of ATP to a Ser-residue of myosin light chain [19]; phosphorylation sites: Serresidues in smooth and skeletal muscle [20]; phosphorylation sites: Thr-residues in smooth muscle and pancreas myosin light chain [11, 16, 20]; phosphorylation sites: Ser-19 and Thr-18 in smooth muscle myosin light chain [20]; ITP, GTP, CTP or UTP cannot replace ATP [2]; no substrate is phosphorylase b [1, 2, 6, 7, 9, 11, 12, 14, 15, 18, 23, 24, 33]; no substrate is casein [1, 2, 6, 9, 11, 12, 14, 15, 18, 23-25, 33]; no substrate is troponin [1, 2, 33]; no substrates are 15 kDa and 22 kDa light-chain or heavy-chain fractions of myosin from white skeletal muscle [2]; no substrates are myosin heavy chain and phosvitin [6, 9, 15, 24, 33]; no substrates are actin and tropomyosin [25]; no substrate is protamine [6, 15]; no substrate is histone III-S from thymus [2]; no substrate is histone 2-A [6, 9, 12, 14, 16, 18, 24, 25]; no substrate is histone 2 b [11, 25]; no substrate is histone V-S [6, 12, 18]; no substrate is histone H1 [25]; no substrate is phosphorylase kinase [9, 15, 18, 33]; no substrate is molluscan adductor myosin [2, 33]; no substrate is synapsin [11, 16]; no substrates are myelin basic protein, glycogen synthase, tubulin, microtubule-associated protein 2, kemptide and peptide pp60src [11]) (Reversibility: ? [1-33]) [1-33] ADP + myosin light chain phosphate [1-29] ATP + myosin regulatory light chain (Reversibility: ? [49]) [49] ? ATP + telokin ( telokin may modulate enzyme activity in vivo [39]) (Reversibility: ? [39]) [39] ADP + ? Additional information ( the enzyme possesses ATP-ase activity [9]; skeletal, gizzard smooth and cardiac enzymes perform intramolecular autophosphorylation in the absence of acceptor substrate [24, 33]; no intramolecular autophosphorylation in the absence of acceptor substrate [15, 25]) [9, 15, 24, 25, 33] ?

$ %  

(+)-catechin ( IC50: 0.44 mM [27]) [27] (-)-epicatechin ( IC50: 0.32 mM [27]) [27] 1,12-diaminododecane ( IC50: 0.063 mM [32]) [32] 1-hexadecylpyridinium bromide ( IC50: 0.049 mM [32]) [32] 2,2'-dihydroxychalcone [27] 3',4',5'-tri-O-methyltricetin [27] 3',4'-dihydroxyflavone ( IC50: 0.262 mM [27]) [27] 3,3',4'-trihydroxyflavone ( IC50: 0.001 mM [27]) [27]

36

2.7.1.117

2.7.1.117

Myosin-light-chain kinase

5,4'-dihydroxyflavone ( IC50: 0.024 mM [27]) [27] 5,7-dihydroxyflavone ( IC50: 0.043 mM [27]) [27] 7,8,3',4'-tetrahydroxyflavone ( IC50: 0.02 mM [27]) [27] 7-O-methylapigenin [27] AKKLSKDRMAAYMARRK ( synthetic peptide, analog of inhibitory region of myosin light chain kinase [26]) [26] AKKLSKDRMKKYMA ( synthetic peptide, analog of inhibitory region of myosin light chain kinase [26]) [26] AKKLSKDRMKKYMAAAA ( synthetic peptide, analog of inhibitory region of myosin light chain kinase [26]) [26] AKKLSKDRMKKYMARR ( synthetic peptide, analog of inhibitory region of myosin light chain kinase [26]) [26] AKKLSKDRMKKYMARRK ( synthetic peptide, analog of inhibitory region of myosin light chain kinase [26]) [26] AKKLSKDRMKKYMARRKWQKTG ( synthetic peptide, analog of inhibitory region of myosin light chain kinase [26]) [26] ARRKWQKTGHAVRAIGRLSS [47] ATP ( free form, strong, not in the presence of excess Mg2+ [2]) [2] Ca2+ ( at higher free concentrations, 0.4-3 mM, independent of Mg2+ or pH-value [7]) [7] d-sphingosine ( IC50: 0.006 mM [32]) [32] EGTA ( strong [2]) [1, 2, 6-8] KCl [3] KDRMKKYMARR ( synthetic peptide, analog of inhibitory region of myosin light chain kinase [26]) [26] LSKDRMKKYMARRKWQK ( synthetic peptide, analog of inhibitory region of myosin light chain kinase [26]) [26] ML-7 [45] MS-347a ( from Aspergillus sp. KY52178, structurally related to sydowinin B, irreversible, inhibition of calmodulin-dependent and independent activity, IC50: 0.0092 mM [31]) [31] N-alkyl-N,N-dimethyl-3-ammonio-1-propanesulfonates ( zwittergents 3-14 and 16 [32]) [32] N-methyloctadecylamine ( IC50: 0.01 mM [32]) [32] NaCl [3] RRKWQKTGHAVRAIGRL [47] RRKYQKTGHAVRAIGRL [47] acylcarnitin ( weak [32]) [32] alizarin ( IC50: 0.014 mM [17]) [17] alkylamine ( long and straight chain, most effective with chain length C-13 to C-18 [32]) [32] alkyltrimethylammonium halide [32] amiloride [30] anthraflavic acid ( IC50: 0.037 mM [17]) [17] anthrarufin [17] apigenin ( IC50: 0.023 mM [27]) [27] arachidonic acid [29] 37

Myosin-light-chain kinase

2.7.1.117

cAMP-dependent protein kinase ( phosphorylates light chain myosin kinase leading to decreased affinity from calmodulin [8, 11, 15]) [8, 11, 15] calmodulin-binding protein from bovine cardiac muscle [10] chalcone [27] chrysazine ( IC50: 0.02 mM [17]) [17] chrysophanic acid [17] decylamine ( IC50: 0.2 mM [32]) [32] diaminoanthraquinone ( IC50: 0.018 mM [17]) [17] dihydroapigenin ( IC50: 0.17 mM [27]) [27] dihydrofisetin ( IC50: 0.18 mM [27]) [27] dihydroluteolin [27] dihydroquercetin ( IC50: 0.08 mM [27]) [27] dihydrosphingosine ( erythro- and threo-dihydrosphingosine, IC50: 0.008 mM [32]) [32] dimethyldioctadecylammonium bromide ( IC50: 0.008 mM [32]) [32] dioctylamine ( IC50: 0.055 mM [32]) [32] dodecylamine ( IC50: 0.083 mM [32]) [32] dodecyltrimethylammonium bromide ( IC50: 0.078 mM [32]) [32] emodin ( IC50: 0.008 mM [17]) [17] fisetin ( IC50: 0.005 mM [27]) [27] galangin ( IC50: 0.02 mM [27]) [27] gossypol [27] hesperidin [27] hexadecylamine ( IC50: 0.016 mM [32]) [32] hexadecyltrimethylammonium bromide ( IC50: 0.011 mM [32]) [32] histone 2A [24] hydroxyflavone ( IC50: 0.32 mM [27]) [27] increasing ionic strength ( up to 0.4 M NaCl, weak [2]; above 0.1 M KCl [24]) [2, 9, 24] isoliquiritigenin [27] kaempferid ( IC50: 0.008 mM [27]) [27] kaempferol ( i.e. 3,5,7-trihydroxy-2-(4-hydroxyphenyl)-1-benzopyran-4-one, IC50: 0.00045 mM [13]; IC50: 0.004 mM [27]) [13, 27] lauroylcholine iodide ( IC50: 0.12 mM [32]) [32] linoleic acid [29] luteolin ( IC50: 0.026 mM [27]) [27] merocyanine dye (C16 ) ( IC50: 0.040 mM [32]) [32] merocyanine dye (CH3 ) [32] mitoxanthrone ( IC50: 0.002 mM [17]) [17] morin ( IC50: 0.028 mM [27]) [27] myricetin ( IC50: 0.006 mM [27]) [27] myristoylcarnitine chloride [32] myristoylcholine iodide ( IC50: 0.02 mM [32]) [32] naphthalene sulfonamide derivatives [23] octadecylamine ( IC50: 0.011 mM [32]) [32] 38

2.7.1.117

Myosin-light-chain kinase

okanin [27] oleic acid [29] oleylamine ( IC50: 0.006 mM [32]) [32] p21-activated kinase 1 [45] palmitoylcarnitine chloride [32] palmitoylcholine iodide ( IC50: 0.014 mM [32]) [32] phosphate ( up to 0.1 M, weak [2]) [2] phosphorylation ( at 2 sites [23]) [20, 23] pseudobabtisin [27] purpurin ( IC50: 0.025 mM [17]) [17] quercetagetin ( IC50: 0.026 mM [27]) [27] quercetin ( IC50: 0.006 mM [27]) [27] quercetrin ( IC50: 0.137 mM [27]) [27] quinalizarin ( IC50: 0.053 mM [17]) [17] quinizarin ( IC50: 0.026 mM [17]) [17] rutin ( IC50: 0.32 mM [27]) [27] sodium alkylsulfate [32] sodium dodecylsulfate ( IC50: 0.049 mM [32]) [32] sodium octadecylsulfate ( IC50: 0.043 mM [32]) [32] sodium tetradecylsulfate ( IC50: 0.038 mM [32]) [32] stearoylcarnitine chloride [32] stearoylcholine iodide ( IC50: 0.013 mM [32]) [32] tetradecylamine ( IC50: 0.012 mM [32]) [32] tetradecyltrimethylammonium bromide ( IC50: 0.011 mM [32]) [32] tricetin ( IC50: 0.012 mM [27]) [27] tridecylamine ( IC50: 0.019 mM [32]) [32] trifluoperazine [23] unsaturated fatty acids ( irreversible by Ca2+ /calmodulin [29]) [29] wortmannin ( i.e. MS-54, IC50: 0.0019 mM, irreversible, highly selective, kinetics, high concentrations of ATP protect [28]) [28] Additional information ( structural requirements of autoinhibition of myosin light chain kinase [26]; no inhibition by 3',5'-cAMP [1,2]; no inhibition by AMP [2]; no inhibition by epicatechin, pseudobabtisin, 4-dimethylaminobenzaldehyde [27]; inhibition by autophosphorylation [36]; no inhibition by diacylglycerol, phosphatidylserine [29]) [1, 2, 26, 27, 29, 36] ! .  %  

calmodulin ( requirement [3, 5-23, 25-33]; only active as ternary complex of calmodulin, Ca2+ and kinase: activation is initiated by binding of Ca2+ to calmodulin [9]; 1:1 stoichiometric complex in the presence of Ca2+ [23, 33]; lower affinity for Ca2+ /caldesmon after phosphorylation by cAMP-dependent protein kinase, Ka: 0.0000006 mM [15]; trypsin or chymotrypsin digested smooth muscle enzyme is independent of Ca2+ /calmodulin [20, 22, 23, 33]; aged enzyme loses Ca2+ /calmodulin sensitivity by proteolysis [7];

39

Myosin-light-chain kinase

2.7.1.117

not [24]; calmodulin in presence of Ca2+ abolishes the inhibition of actin-myosin interaction [36-38]; optimal ratio of enzyme to calmodulin for telokin phosphorylation is 1:4 [39]) [3, 5-23, 25-33, 36-40, 43, 47, 48]  &    

autophosphorylation ( activation [24]; no activation [15,25]) [24] phosphorylation ( activation, smooth muscle enzyme, not bovine cardiac enzyme [9]; no activation [25]) [9] Additional information ( no activation by mild proteolysis [9]; no activation by phosphatidylserine [15]; no activation by cAMP, [1, 2, 7, 15, 24]; no activation by cGMP [24]; no activation by Ca2+ plus b-lactoglobulin, cytochrome c, troponin C or parvalbumin [7]) [1, 2, 7, 9, 15, 24] ' ( 

Ca2+ ( requirement, only in combination with calmodulin [7]; Km -value: 0.0003 [15]; effect depends on Mg2+ concentration [2]; aged enzyme loses Ca2+ /calmodulin sensitivity by proteolysis [7]; inhibits at higher free concentrations [7]; not [24]; KM : 0.0025 mM [16]; Ca2+ in presence of calmodulin abolishes the inhibition of actin-myosin interaction [36-38]) [1-3, 7, 5-23, 25-33, 36-40, 47, 48] Mg2+ ( requirement, varies with ATP-concentration, MgATP2is the active substrate [2]; Km -value: 2 mM [11]) [2, 7, 9, 11, 24] ) &  * +, 310 (myosin light chain, 23-25 C, pH 7.5 [6]) [6] 960 (myosin light chain, 25 C, pH 7.6, isolated [3]) [3] 1140 (myosin light chain, 25 C, pH 7.6, bound to myosin [3]) [3] 5280 (myosin light chain, 25 C, pH 7.6, isolated, freshly prepared [3]) [3]   !   &  *-., 0.00615 [24] 0.3-0.8 [1] 0.47 [16] 1.8 ( myosin light chain, isolated [11]) [11, 20] 2.51 ( myosin light chain, in native myosin [11]) [11] 3.1 [6] 5.4 ( rabbit uterine myosine [14]) [14] 6 [18-20] 6.1-8.7 [8] 7.7 ( turkey gizzard myosine [14]) [14] 7.9 [12, 15, 20] 8.1-8.9 [8] 13 ( BpaKKRAARATSNVFA as substrate [46]) [46] 18.5 [10] 19.9 ( KKRAARATSNVFA as substrate [46]) [46]

40

2.7.1.117

Myosin-light-chain kinase

24 ( skeletal muscle [3,20]) [3, 20] 25 [2] 33 [21] Additional information ( 1280 pmol/min/pmol [48]) [48] /01 *', 0.000002 (calmodulin, 30 C, pH 6.8 [11]; 30 C, pH 7.5 [16]) [11, 16] 0.004 (Dictyostelium myosin, 22 C, pH 7.5 [24]) [24] 0.005 (myosin light chain, 24 C, pH 7.3 [18]) [18, 19] 0.005-0.0095 (myosin light chain, 30 C, pH 7.2 [12, 13]) [5, 12, 13, 21] 0.0067 (myosin regulatory light chain, pH 7, M968P [49]) [49] 0.0075 (BpaKKRAARATSNVFA, 25 C, pH 7, Bpa is the photoreactive amino acid p-benzoylphenylalanine [46]) [46] 0.0084 (KKRAARATSNVFA, 25 C, pH 7 [46]) [46] 0.011 (myosin regulatory light chain, pH 7, A986P [49]) [49] 0.011-0.02 (myosin light chain, 30 C, pH 7 [10]) [10] 0.014 (myosin regulatory light chain, pH 7, wild type [49]) [49] 0.0156 (Limulus myosin light chain, 25 C, pH 7.5 [25]) [25] 0.018 (myosin light chain, 23-25 C, pH 7.5 [6]; 30 C, pH 7.5 [16]) [6, 16] 0.019 (myosin light chain, 25 C, pH 7.6, bound to myosin [3]) [3] 0.02-0.027 (turkey gizzard myosin light chain, 30 C, pH 7 [15]; 30 C, pH 7.6, ATP [7]; 30 C, pH 6.8 [11]) [7, 11, 15] 0.04 (turkey gizzard myosin light chain, pH 7.5, myometrium enzyme [14]) [14] 0.05 (ATP, 24 C, pH 7.3 [18]) [18, 19] 0.05-0.063 (bovine cardiac muscle myosin light chain) [21] 0.05-0.063 (isolated myosin light chain, 25 C, pH 7.6 [3]) [3] 0.073 (ATP, 30 C, pH 7.5 [16]) [16] 0.075 (ATP) [13] 0.094-0.096 (skeletal muscle myosin light chain) [21] 0.1-0.2 (rabbit white skeletal muscle myosin P-light chain, 27 C, pH 7.6 [2]) [2] 0.11 (kemptamide, 30 C, pH 6.8 [11]) [11] 0.121 (ATP, 23-25 C, pH 7.5 [6]) [6] 0.167 (ATP) [21] 0.175 (ATP, 30 C, pH 8 [9]) [9] 0.22 (ATP, 30 C, pH 7 [10]) [10] 0.224 (ATP) [21] Additional information ( in the presence of wortmannin [28]; kinetic constants for enzymes from various sources with different myosin light chains as substrates [33]) [28, 33] 20   6.5 [2] 7-8 [1] 41

Myosin-light-chain kinase

2.7.1.117

7.8-8 [23] 8.1 [9] 20  5.7-8.2 ( about half-maximal activity at pH 5.7 and 8.2, with a small shoulder of 77% of maximal activity at 7-7.5 [2]) [2] 6.3-9.2 ( about half-maximal activity at pH 6.3 and 9.2 [3]) [3] 6.8-8.8 ( about half-maximal activity at pH 6.8 and about 75% of maximal activity at pH 8.8 [9]) [9] )     * , 22 ( assay at [14, 24]) [14, 24] 23-25 ( assay at [6]) [6] 24 ( assay at [18]) [18] 25 ( assay at [1-3]) [1-3] 28 ( assay at [28, 31]) [28, 31] 30 ( assay at [1, 9-11, 16, 27]) [1, 9-11, 16, 27]

3  "  ' 4 % 34000 ( gel filtration [24]) [24] 37000 ( PAGE, 2 forms of myosin light chain kinase [25]) [25] 39000 ( PAGE, 2 forms of myosin light chain kinase [25]) [25] 77000 ( gel filtration [2]) [2] 85000 ( gel filtration [9]) [9] 103000 ( sedimentation equilibrium method [3]) [3] 124000 ( sedimentation equilibrium centrifugation [18,19,33]) [18, 19, 33] 125700 ( calculated from sequence of DNA [5]) [5] 127000 ( sucrose density gradient centrifugation [11]) [11] 130000 ( gel filtration [18]) [18] 150000 ( gel filtration and sedimentation studies [7]) [7] 211000 ( deduced from nucleotide sequence [42]) [42] 926000 ( deduced from nucleotide sequence, stretchin-MLCK [44]) [44] Additional information ( relative masses of various animal skeletal muscle enzymes [33]; amino acid composition of rabbit [3]; different molecular weights may be due to high sensitivity to proteolysis during purification [33]) [3, 8, 18, 23, 33]  

? ( x * 152000, SDS-PAGE, recombinant enzyme [5]; x * 92000, SDS-PAGE [1]; x * 155000, SDS-PAGE [5]; x * 94000, SDSPAGE [10]; x * 214000, SDS-PAGE, from endothelium [34]; x * 105000, SDS-PAGE [6]; x * 130000, SDS-PAGE [15]; x * 135000,

42

2.7.1.117

Myosin-light-chain kinase

SDS-PAGE [12]; x * 136000, SDS-PAGE, enzyme from smooth muscle [5]; x * 138000, SDS-PAGE [16]; x * 152000, SDS-PAGE [11]; x * 160000, SDS-PAGE, [12]; SDS-PAGE [14]) [1, 5, 6, 10-12, 14-16] monomer ( 1 * 34000, SDS-PAGE [24]; 1 * 37000, SDS-PAGE 2 forms of myosin light chain kinase [25]; 1 * 39000, SDSPAGE, 2 forms of myosin light chain kinase [25]; 1 * 77000, SDS-PAGE [2]; 1 * 85000, SDS-PAGE [9]; 1 * 94000, SDS-PAGE [3]; 1 * 130000, SDS-PAGE [8, 16, 18, 19, 33]; 1 * 214000, SDS-PAGE, endothelial enzyme [42]) [2, 3, 8, 9, 16, 18, 19, 24, 25, 33, 42] Additional information ( gel electrophoresis in various buffers gives different molecular weights [3]; skeletal muscle enzyme structure: overall asymmetric shape, globular head and tail region [23]; skeletal muscle myosin light chain kinases from different species share more identity than skeletal muscle and smooth muscle myosin light chain kinases from the same species [22]; different molecular weights may be due to high sensitivity to proteolysis during purification [33]; multienzyme complex with smooth muscle myosin light chain phosphatase [41]; the high moecular weight endothelial enzyme is stable associated to a complex containing p60src and 80000 cortactin [51]) [3, 22, 23, 33, 41, 51]

5 $   .#   .'  .

  

.

 adrenal medulla ( medulla [7]; not rat [5]) [5, 7] aorta ( thoracic [13]) [5, 13] bladder [14] brain [11, 20, 47] breast [35] cardiac muscle ( myocardium [9]) [1, 9, 10, 33] endothelium [34, 42, 51] gizzard ( pregnant sheep myometrium, turkey and chicken gizzard enzyme are immunologically related [14]; rat pancreatic and turkey gizzard enzyme are immunologically related [17]) [8, 12, 14, 16-18, 20, 23, 2629, 33, 36-41, 43] kidney ( not rat [5]) [5, 45] leukocyte ( polymorphonuclear and alveolar [4]) [4] liver ( not rat [5]) [5] myometrium ( pregnant sheep myometrium, turkey and chicken gizzard enzyme are immunologically related [14]) [14, 15, 20] pancreas ( rat pancreatic and turkey gizzard enzyme are immunologically related [16]) [16, 20] platelet [6, 19, 20, 33] rumen [8]

43

Myosin-light-chain kinase

2.7.1.117

skeletal muscle ( red and white [33]; back and hindlegs [20]; pectoralis muscle [21]) [1-3, 20-22, 33, 35, 46] skin [43] smooth muscle ( arterial [12, 20]; uterus, trachea, aorta, ileum, gizzard [5]) [5, 12, 20, 22, 23, 33, 35-37, 40, 41, 43, 47, 49] stomach [14, 23] tenson ( muscle [25]) [25] thyroid gland [33] trachea [14] uterus ( not rat [5]) [5] Additional information ( myosin light chain kinases in smooth muscle and non-muscle tissues are the same protein [5]) [5] 6 "  actin filament ( F-actin-associated along cellular stress fibres [12]) [12] actomyosin ( associated [6]) [6] myofibril ( associated [8, 18-20]) [8, 18, 20] sarcoplasm [1, 2] soluble [9, 11, 15, 16] # !   (partial [1]; affinity chromatography on calmodulin-Sepharose [3]) [1-4, 20, 21] [21, 22, 35-39, 41] (affinity chromatography on calmodulin-Sepharose [18]) [8, 18-20] [24, 35] (partial [16,20]) [16, 20] (partial, affinity chromatography on calmodulin-Sepharose [6]) [6, 20] [15] [20] [14] [25] (purified to homogeneity from a number of vertebrate muscles and partially purified from non-muscle tissues [33]) [33]   (expression in A7r5 rat thoracic aorta smooth muscle cells as GFP-fusion protein [48]) [48] (expression in COS-7 african green monkey kidney cells [49]) [49] (expression in COS-cells [5]) [5] (expression in Sf9 insect cells [46]) [46] (expression in BL21(DE3) cells as GFP fusion protein [50]) [50] (skeletal muscle enzyme [22]) [22] (transient expression as GFP fusion protein in A7r5, HeLa, NIH3T3 or COS-7 cells [50]) [50] (expression as His-tagged protein [51]) [51]

44

2.7.1.117

Myosin-light-chain kinase

    A983P ( dramatic increase in Ca2+ required for half-maximal activity [49]) [49] A986P ( significantly increase in Ca2+ required for half-maximal activity, slightly decreased KM for regulatory light chain [49]) [49] M968P ( 10% Ca2+ /calmodulin independent activity of total activity, decreased KM for regulatory light chain [49]) [49] Additional information ( deletion of DRFXXL motifs leads to a worse binding to actin filaments especially in the presence of Mg2+ [50]) [50]

7    20   5 ( rapid inactivation below [2, 3]; 30-60 min, about 50% loss of activity [9]) [2, 3, 9] 6.3-8 ( stable in 10% sucrose [2]) [2] 8     !  , EGTA prevents Ca2+ -dependent proteolysis during initial purification [18] , MgCl2 is critical for kinase extraction from myofibrils [18] , glycerol and Tween 40 stabilize [11] , protease inhibitors with broad specificity and glycerol stabilize during initial purification, unstable to further purification [7] , repeated freeze-thawing decreases activity [19, 20] , unstable upon lyophilization [3, 11] , protease inhibitors stabilize during purification [14, 16, 19]     , -20 C, in 5% w/v sucrose, several weeks [20] , -30 C, 30% loss of activity within 3 weeks [18] , -70 C, at least 6 months [18] , -70 C, 1% bovine serum albumin, more than 2 months [16] , -20 C, quite unstable on storage [15] , proteolysis occurs even on storage at -80 C, myometrium enzyme [14]

! 

[1] Pires, E.; Perry, S.V.; Thomas, M.A.W.: Myosin light-chain kinase, a new enzyme from striated muscle. FEBS Lett., 41, 292-296 (1974) [2] Pires, E.M.V.; Perry, S.V.: Purification and properties of myosin light-chain kinase from fast skeletal muscle. Biochem. J., 167, 137-146 (1977) [3] Nagamoto, H.; Yagi, K.: Properties of myosin light chain kinase prepared from rabbit skeletal muscle by an improved method. J. Biochem., 95, 11191130 (1984)

45

Myosin-light-chain kinase

2.7.1.117

[4] Yang, H.H.; Boxer, L.A.: Purification of myosin light chain kinase from rabbit polymorphonuclear leukocytes. Pediatr. Res., 15, 229-234 (1981) [5] Gallagher, P.J.; Herring, B.P.; Griffin, S.A.; Stull, J.T.: Molecular characterization of a mammalian smooth muscle myosin light chain kinase [published erratum appears in J Biol Chem 1992 May 5;267(13):9450]. J. Biol. Chem., 266, 23936-23944 (1991) [6] Hathaway, D.R.; Adelstein, R.S.: Human platelet myosin light chain kinase requires the calcium-binding protein calmodulin for activity. Proc. Natl. Acad. Sci. USA, 76, 1653-1657 (1979) [7] Serventi, I.M.; Coffee, C.J.: Characterization of myosin light-chain kinase from bovine adrenal medulla. Arch. Biochem. Biophys., 245, 379-388 (1986) [8] Walsh, M.P.; Hinkins, S.; Flink, I.L.; Hartshorne, D.J.: Bovine stomach myosin light chain kinase: purification, characterization, and comparison with the turkey gizzard enzyme. Biochemistry, 21, 6890-6896 (1982) [9] Walsh, M.P.; Vallet, B.; Autric, F.; Demaille, J.G.: Purification and characterization of bovine cardiac calmodulin-dependent myosin light chain kinase. J. Biol. Chem., 254, 12136-12144 (1979) [10] Wolf, H.; Hofmann, F.: Purification of myosin light chain kinase from bovine cardiac muscle. Proc. Natl. Acad. Sci. USA, 77, 5852-5855 (1980) [11] Bartelt, D.C.; Moroney, S.; Wolff, D.J.: Purification, characterization and substrate specificity of calmodulin-dependent myosin light-chain kinase from bovine brain. Biochem. J., 247, 747-756 (1987) [12] Yamazaki, K.; Itoh, K.; Sobue, K.; Mori, T.; Shibata, N.: Purification of caldesmon and myosin light chain (MLC) kinase from arterial smooth muscle: comparisons with gizzard caldesmon and MLC kinase. J. Biochem., 101, 1-9 (1987) [13] Rogers, J.C.; Williams, D.L.: Kaempferol inhibits myosin light chain kinase. Biochem. Biophys. Res. Commun., 164, 419-425 (1989) [14] Pato, M.D.; Lye, S.J.; Kerc, E.: Purification and characterization of pregnant sheep myometrium myosin light chain kinase. Arch. Biochem. Biophys., 287, 24-32 (1991) [15] Higashi, K.; Fukunaga, K.; Matsui, K.; Maeyama, M.; Miyamoto, E.: Purification and characterization of myosin light-chain kinase from porcine myometrium and its phosphorylation and modulation by cyclic AMP-dependent protein kinase. Biochim. Biophys. Acta, 747, 232-240 (1983) [16] Bissonnette, M.; Kuhn, D.; de Lanerolle, P.: Purification and characterization of myosin light-chain kinase from the rat pancreas. Biochem. J., 258, 739-747 (1989) [17] Jinsart, W.; Ternai, B.; Polya, G.M.: Inhibition of myosin light chain kinase, cAMP-dependent protein kinase, protein kinase C and of plant Ca2+ -dependent protein kinase by anthraquinones. Biol. Chem. Hoppe-Seyler, 373, 903910 (1992) [18] Adelstein, R.S.; Klee, C.B.: Purification and characterization of smooth muscle myosin light chain kinase. J. Biol. Chem., 256, 7501-7509 (1981) [19] Adelstein, R.S.; Klee, C.B.: Purification of smooth muscle myosin lightchain kinase. Methods Enzymol., 85, 298-308 (1982)

46

2.7.1.117

Myosin-light-chain kinase

[20] Conti, M.A.; Adelstein, R.S.: Purification and properties of myosin light chain kinases. Methods Enzymol., 196, 34-47 (1991) [21] Nunnally, M.H.; Rybicki, S.B.; Stull, J.T.: Characterization of chicken skeletal muscle myosin light chain kinase. Evidence for muscle-specific isozymes. J. Biol. Chem., 260, 1020-1026 (1985) [22] Leachman, S.A.; Gallagher, P.J.; Herring, B.P.; McPhaul, M.J.; Stull, J.T.: Biochemical properties of chimeric skeletal and smooth muscle myosin light chain kinases. J. Biol. Chem., 267, 4930-4938 (1992) [23] Bailin, G.: Structure and function of a calmodulin-dependent smooth muscle myosin light chain kinase. Experientia, 40, 1185-1188 (1984) [24] Tan, J.L.; Spudich, J.A.: Dictyostelium myosin light chain kinase. Purification and characterization. J. Biol. Chem., 265, 13818-13824 (1990) [25] Sellers, J.R.; Harvey, E.V.: Purification of myosin light chain kinase from Limulus muscle. Biochemistry, 23, 5821-5826 (1984) [26] Ikebe, M.; Reardon, S.; Fay, F.S.: Primary structure required for the inhibition of smooth muscle myosin light chain kinase. FEBS Lett., 312, 245-248 (1992) [27] Jinsart, W.; Ternai, B.; Polya, G.M.: Inhibition of wheat embryo calciumdependent protein kinase and avian myosin light chain kinase by flavonoids and related compounds. Biol. Chem. Hoppe-Seyler, 372, 819-827 (1991) [28] Nakanishi, S.; Kakita, S.; Takahashi, I.; Kawahara, K.; Tsukada, E.; Sano, T.; Yamada, K.; Yoshida, M.; Kase, H.; Matsuda, Y.; Hashimoto, Y.; Nonomura, Y.: Wortmannin, a microbial product inhibitor of myosin light chain kinase. J. Biol. Chem., 267, 2157-2163 (1992) [29] Kigoshi, T.; Uchida, K.; Kaneko, M.; Iwasaki, R.; Nakano, S.; Azukizawa, S.; Morimoto, S.: Direct inhibition of smooth muscle myosin light chain kinase by arachidonic acid in a purified system. Biochem. Biophys. Res. Commun., 171, 369-374 (1990) [30] Higashihara, M.: Inhibition of myosin light chain kinase by amiloride. Biochem. Biophys. Res. Commun., 162, 1253-1259 (1989) [31] Nakanishi, S.; Ando, K.; Kawamoto, I.; Matsuda, Y.: MS-347a, a new inhibitor of myosin light chain kinase from Aspergillus sp. KY52178. J. Antibiot., 46, 1775-1781 (1989) [32] Jinsart, W.; Ternai, B.; Polya, G.M.: Inhibition and activation of wheat embryo calcium-dependent protein kinase and inhibition of avian myosin light chain kinase by long chain aliphatic amphiphiles. Plant Sci., 78, 165175 (1991) [33] Stull, J.T.; Nunnally, M.H.; Michnoff, C.H in: Calmodulin-dependent protein kinases. The Enzymes, 3rd. Ed. (Boyer, P.D., Krebs, E.G., eds.), 17, 113-166 (1986) [34] Verin, A.D.; Gilbert-McClain, L.I.; Patterson, C.E.; Garcia, J.G.N.: Biochemical regulation of the nonmuscle myosin light chain kinase isoform in bovine endothelium. Am. J. Respir. Cell Mol. Biol., 19, 767-776 (1998) [35] Fujita, K.; Ye, L.-H.; Sato, M.; Okagaki, T.; Nagamachi, Y.; Kohama, K.: Myosin light chain kinase from skeletal muscle regulates an ATP-dependent in-

47

Myosin-light-chain kinase

2.7.1.117

teraction between actin and myosin by binding to actin. Mol. Cell. Biochem., 190, 85-90 (1999) [36] Okagaki, T.; Ye, L.H.; Samizo, K.; Tanaka, T.; Kohama, K.: Inhibitory effect of the catalytic domain of myosin light chain kinase on actin-myosin interaction: insight into the mode of inhibition. J. Biochem., 125, 1055-1060 (1999) [37] Okagaki, T.; Hayakawa, K.; Samizo, K.; Kohama, K.: Inhibition of the ATPdependent interaction of actin and myosin by the catalytic domain of the myosin light chain kinase of smooth muscle: possible involvement in smooth muscle relaxation. J. Biochem., 125, 619-626 (1999) [38] Hayakawa, K.; Okagaki, T.; Ye, L.H.; Samizo, K.; Higashi-Fujime, S.; Takagi, T.; Kohama, K.: Characterization of the myosin light chain kinase from smooth muscle as an actin-binding protein that assembles actin filaments in vitro. Biochim. Biophys. Acta, 1450, 12-24 (1999) [39] Sobieszek, A.; Andruchov, O.Y.; Nieznanski, K.: Kinase-related protein (telokin) is phosphorylated by smooth-muscle myosin light-chain kinase and modulates the kinase activity. Biochem. J., 328, 425-430 (1997) [40] Edwards, R.A.; Walsh, M.P.; Sutherland, C.; Vogel, H.J.: Activation of calcineurin and smooth muscle myosin light chain kinase by Met-to-Leu mutants of calmodulin. Biochem. J., 331, 149-152 (1998) [41] Sobieszek, A.; Borkowski, J.; Babiychuk, V.S.: Purification and characterization of a smooth muscle myosin light chain kinase-phosphatase complex. J. Biol. Chem., 272, 7034-7041 (1997) [42] Garcia, J.G.N.; Lazar, V.; Gilbert-McClain, L.I.; Gallagher, P.J.; Verin, A.D.: Myosin light chain kinase in endothelium: molecular cloning and regulation. Am. J. Respir. Cell Mol. Biol., 16, 489-494 (1997) [43] Van Lierop, J.E.; Wilson, D.P.; Davis, J.P.; Tikunova, S.; Sutherland, C.; Walsh, M.P.; Johnson, J.D.: Activation of smooth muscle myosin light chain kinase by calmodulin. Role of Lys30 and Gly40. J. Biol. Chem., 277, 65506558 (2002) [44] Champagne, M.B.; Edwards, K.A.; Erickson, H.P.; Kiehart, D.P.: Drosophila stretchin-MLCK is a novel member of the titin/myosin light chain kinase family. J. Mol. Biol., 300, 759-777 (2000) [45] Sanders, L.C.; Matsumura, F.; Bokoch, G.M.; de Lanerolle, P.: Inhibition of myosin light chain kinase by p21-activated kinase. Science, 283, 2083-2085 (1999) [46] Gao, Z.-H.; Zhi, G.; Herring, B.P.; Moomaw, C.; Deogny, L.; Slaughter, C.A.; Stull, J.T.: Photoaffinity labeling of a peptide substrate to myosin light chain kinase. J. Biol. Chem., 270, 10125-10135 (1995) [47] Toeroek, K.; Cowley, D.J.; Brandmeier, B.D.; Howell, S.; Aitken, A.; Trentham, D.R.: Inhibition of calmodulin-activated smooth-muscle myosin light-chain kinase by calmodulin-binding peptides and fluorescent (phosphodiesterase-activating) calmodulin derivatives. Biochemistry, 37, 61886198 (1998) [48] Lin, P.; Luby-Phelps, K.; Stull, J.T.: Properties of filament-bound myosin light chain kinase. J. Biol. Chem., 274, 5987-5994 (1999)

48

2.7.1.117

Myosin-light-chain kinase

[49] Padre, R.C.; Stull, J.T.: Conformational requirements for Ca2+ /calmodulin binding and activation of myosin light chain kinase. FEBS Lett., 472, 148152 (2000) [50] Smith, L.; Parizi-Robinson, M.; Zhu, M.S.; Zhi, G.; Fukui, R.; Kamm, K.E.; Stull, J.T.: Properties of long myosin light chain kinase binding to F-actin in vitro and in vivo. J. Biol. Chem., 277, 35597-35604 (2002) [51] Dudek, S.M.; Birukov, K.G.; Zhan, X.; Garcia, J.G.N.: Novel interaction of cortactin with endothelial cell myosin light chain kinase. Biochem. Biophys. Res. Commun., 298, 511-519 (2002)

49

#0)%    




#% %   *8)#,

 ?

     2.7.2.10     GTP:3-phospho-d-glycerate 1-phosphotransferase    phosphoglycerate kinase (GTP)   

kinase (phosphorylating), phosphoglycerate (guanosine triphosphate) kinase, phosphoglycerate (phosphorylating guanosine triphosphate) Additional information (cf. EC 2.7.2.3)     62213-34-7

   Entamoeba histolytica [1]

       !   "   GTP + 3-phospho-d-glycerate = GDP + 3-phospho-d-glyceroyl phosphate     phospho group transfer      

 GDP + 3-phospho-d-glyceroyl 1-phosphate ( production of GTP [1]) (Reversibility: r [1]) [1] # GTP + 3-phospho-d-glycerate     

 ATP + 3-phospho-d-glycerate ( in glycolytic direction selectivity for GDP over ADP is 150-fold, selectivity for GTP over ATP is about 50fold [1]) (Reversibility: r [1]) [1] # ADP + 3-phospho-d-glyceroyl 1-phosphate

349

Phosphoglycerate kinase (GTP)

2.7.2.10

 GTP + 3-phospho-d-glycerate ( in glycolytic direction selectivity for GDP over ADP is 150-fold, selectivity for GTP over ATP is about 50fold [1]) (Reversibility: r [1]) [1] # GDP + 3-phospho-d-glyceroyl 1-phosphate  ITP + 3-phospho-d-glycerate (, in glycolytic direction 40% of the activity with GDP, 30% of the activity with GTP [1]) (Reversibility: r [1]) [1] # IDP + 3-phospho-d-glyceroyl 1-phosphate   !   &  *-., 52 ( GDP + 3-phospho-d-glyceroyl phosphate [1]) [1] /01 *', 0.25 (GTP, pH 7, 30 C [1]) [1] 0.3 (GDP, pH 7, 30 C [1]) [1] 0.4 (3-phosphoglycerate, pH 7, 30 C [1]) [1] 0.7 (IDP, pH 7, 30 C [1]) [1] 0.8 (ITP, pH 7, 30 C [1]) [1] 1.2 (ATP, pH 7, 30 C [1]) [1] 10 (ADP, pH 7, 30 C [1]) [1]

5 $   .#   .'  .

  

# !   (partial [1]) [1]

7    8     !  , upon concentration by vacuum dialysis against 20 mM imidazole buffer, pH 7, a loss of about 40% of activity occurs [1]     , 4 C, concentrated enzyme solution, 10% loss of activity per week [1]

! 

[1] Reeves, R.E.; South, D.J.: Phosphoglycerate kinase (GTP). An enzyme from Entamoeba histolytica selective for guanine nucleotides. Biochem. Biophys. Res. Commun., 58, 1053-1057 (1974)

350

8 50  

 

     2.7.2.11     ATP:l-glutamate 5-phosphotransferase    glutamate 5-kinase   

ATP-l-glutamate 5-phosphotransferase ATP:g-l-glutamate phosphotransferase GPK g-glutamate kinase g-glutamyl kinase g-glutamylphosphate kinase glutamate kinase kinase (phosphorylating), glutamate kinase, glutamate (phosphorylating)     54596-30-4

  





Streptococcus thermophilus [1] Escherichia coli (strain CM 25 [3]) [2, 3, 4, 12] Pseudomonas aeruginosa (strain PAO 1 [5]) [5] Triticum aestivum (cv. Mironovska 808 [7]) [6, 7, 8] Lycopersicon esculentum (var. Ailsa Craig [9]) [9, 10] Saccharomyces cerevisiae [11] Thermus ruber [13]

       !   "   ATP + l-glutamate = ADP + l-glutamate 5-phosphate

351

Glutamate 5-kinase

2.7.2.11

    phospho group transfer      

 ATP + l-glutamate ( the enzyme catalyzes the first step in the pathway from glutamate to proline [2,3]; enzyme is involved in biosynthesis of proline [4]; enzyme form GK1 is involved in biosynthesis of l-Pro, enzyme form GK 2 is involved in biosynthesis of glutamine and the function of enzyme form GK 3 has not been found [6]; enzyme GK 1 is the first enzyme of the proline biosynthetic pathway [7]) (Reversibility: ? [2, 3, 4, 6, 7, 10]) [2, 3, 4, 6, 7, 10] # ADP + l-glutamate 5-phosphate     

 ATP + 5-ethyl-l-glutamate ( 5% of the activity with l-glutamate [5]) (Reversibility: ? [5]) [5] # ?  ATP + 5-methyl-l-glutamate ( 6% of the activity with l-glutamate [5]) (Reversibility: ? [5]) [5] # ?  ATP + l-glutamate (Reversibility: ? [1-13]) [1-13] # ADP + l-glutamate 5-phosphate  ATP + l-glutamine ( 10% of the activity with l-glutamate [5]) (Reversibility: ? [5]) [5] # ?  ATP + cis-cycloglutamate ( no reaction with trans-cycloglutamate [3]) (Reversibility: ? [3]) [3] # ADP + cis-cycloglutamyl phosphate [3]  GTP + l-glutamate ( 10% of the activity with ATP [5]) (Reversibility: ? [5]) [5] # GDP + l-glutamate 5-phosphate $ %  

5-oxo-l-Pro ( 12 mM, 10% inhibition [5]) [5] ADP [2, 5] Cd2+ ( 0.1 mM, complete inhibition [5]) [5] dl-3,4-didehydroproline ( 9 mM, 50% inhibition [5]) [5] Hg2+ ( 0.1 mM, complete inhibition [5]) [5] l-Orn ( 12 mM, 10% inhibition [5]) [5] l-Pro ( enzyme from strain PAO1: 5 mM, 50% inhibition, complete inhibition at 30 mM, noncompetitive. Strain PAO 879, a proline-auxotroph mutant lacks a proline-inhibitable g-glutamyl kinase [5]; I50 = 0.08 mM, at room temperature. At low temperatures the inhibition switches over into allosteric activation and the biosynthesis of proline is started [7]; feedback-inhibition [8]) [5, 7, 8, 10] l-azetidine-2-carboxylic acid ( 3 mM, 50% inhibition [5]) [5] l-methionine-dl-sulphoximine ( competitive with l-glutamate [5]) [5]

352

2.7.2.11

Glutamate 5-kinase

l-thioproline ( 12 mM, 10% inhibition [5]) [5] Mg2+ ( above 20 mM [5]) [5] Mn2+ ( above 20 mM [5]) [5] NEM ( 0.125 mM, complete inhibition. Preincubation with 0.25 mM dithiothreitol for 5 min partially protects [5]) [5] PCMB ( 0.125 mM, complete inhibition. Preincubation with 0.25 mM dithiothreitol for 5 min partially protects [5]) [5] iodoacetamide ( 0.125 mM, 60% inhibition. Preincubation with 0.25 mM dithiothreitol for 5 min partially protects [5]) [5] phosphate [5]  &    

l-Pro ( at low temperatures the inhibition switches over into allosteric activation and the biosynthesis of proline is started [7]) [7] ' ( 

K+ ( the enzyme is most active at 30 C at a relative high K+ + Na+ concentration and a K+ /Na+ ratio of 1.8 to 10.2 and at 0 C at both a lower K+ + Na+ concentration and a K+ /Na+ ratio [8]) [8] Mg2+ ( required [4,5]; enzyme GK 1 is strongly activated by Mg2+ , maximum at 60 mM Mg2+ [6]) [4, 5, 6] Mn2+ ( can partially replace Mg2+ [5]) [5] Na+ ( the enzyme is most active at 30 C at a relative high K+ + Na+ concentration and a K+ /Na+ ratio of 1.8 to 10.2 and at 0 C at both a lower K+ + Na+ concentration and a K+ /Na+ ratio [8]) [8]   !   &  *-., 12.68 [5] Additional information [2] /01 *', 0.4 (ATP, pH 7.0, 37 C, g-glutamyl kinase DHPr [2]) [2] 0.5 (ATP, pH 7.0, 37 C, g-glutamyl kinase w+ [2]) [2] 12 (l-glutamate) [5] Additional information ( the concentration of glutamate which yields half-maximal activity is 33 mM for g-glutamyl kinase DHPr, and 37 mM for g-glutamyl kinase w+, no typical Michealis-menten kinetics [2]; plots of the enzyme activity as a function of ATP concentration are non-hyperbolic [5]) [2, 5] / 01 *', 0.06 (ADP, pH 7.0, 37 C [2]) [2] 0.09 (l-Pro, wild-type enzyme [10]) [10] 1.9 (l-Pro, mutant enzyme I79T [10]) [10] 17 (l-Pro, mutant enzyme A62V [10]) [10] 19 (l-Pro, mutant enzyme S159P [10]) [10] 20 (l-Pro, mutant enzyme I149F [10]) [10] 23 (l-Pro, mutant enzyme M94T [10]) [10] 50 (l-Pro, mutant enzyme E153A or E153G [10]) [10]

353

Glutamate 5-kinase

2.7.2.11

55 (l-Pro, mutant enzyme D162G [10]) [10] 58 (l-Pro, mutant enzyme D162N [10]) [10] 82 (l-Pro, mutant enzyme A62T [10]) [10] 90 (l-Pro, mutant enzyme L154S [10]) [10] 180 (l-Pro, mutant enzyme D147G [10]) [10] 310 (l-Pro, mutant enzyme E153K [10]) [10] 20   6-6.3 [5] 6.5-7 [2] 20  5.5-8.5 ( pH 5.5: about 80% of maximal activity, pH 8.5: about 40% of maximal activity [5]) [5] 6-7.5 ( 50% of maximal activity at pH 6.0 and at pH 7.5 [2]) [2] )     * , 55-65 [13]

3  "  ' 4 % 84000 ( gel filtration [5]) [5] 236000 ( g-glutamyl kinase DHPr, gel filtration [2]) [2] 254000 ( gel filtration [6]) [6] Additional information ( two glutamyl kinases of MW 125000 Da and of 38000 Da are detected by gel filtration on Sephadex G-150, a single glutamyl kinase of 250000 Da is detected by Bio-gel A1.5M chromatpgraphy [4]) [4]  

? ( x * 41984, calculation from nucleotide sequence [13]; x * 42000 + x * 84000, gel filtration after dissociation into subunits [6]) [6, 13] hexamer ( 6 * 40000, g-glutamyl kinase DHPr, SDS-PAGE [2]) [2]

5 $   .#   .'  .

  

.

 leaf ( three glutamate kinases: GK 1, GK 2, and GK 3 [6]) [6, 7, 8] # !   (from strain BRL806, designated as g-glutamyl kinase w+ and from reductase-overproducing strain BRL1945, designated as g-glutamyl kinase DHPr [2]) [2, 3] (partial [5]) [5] (enzyme form GK 1 [6]) [6]

354

2.7.2.11

Glutamate 5-kinase

  (an artificial bifunctional enzyme, g-glutamyl kinase/g-glutamyl phosphate reductase obtained by fusing the Escherichia coli genes proA and proB improves NaCl tolerance when expressed in Escherichia coli. The proB gene is fused to the 5'-end of the proA gene with a linker encoding five amino acids [12]) [12] (expression in Escherichia coli [9]) [9] [13]     A62T ( drastic reduction in specific activity. 911fold increase in Ki value for l-Pro compared to wild-type enzyme [10]) [10] A62V ( drastic reduction in specific activity. 188fold increase in Ki value for l-Pro compared to wild-type enzyme [10]) [10] D147G ( reduction in catalytic activity. 2000fold increase in Ki -value for l-Pro compared to wild-type enzyme [10]) [10] D154N ( the mutation results in a prominant increase in cell viability after freezing at -20 C compared to the viability of the cells harboring the wild-type PRO1 gene. The altered g-glutamyl kinase results in stabilization of the complex with g-glutamyl phosphate reductase or has an indirect effect on g-glutamyl phosphate reductase activity which leads to an increase in l-proline production in Saccharomyces cerevisiae [11]) [11] D162G ( drastic reduction in specific activity. 611.1fold increase in Ki -value for l-Pro compared to wild-type enzyme [10]) [10] D162N ( drastic reduction in specific activity. 644fold increase in Ki value for l-Pro compared to wild-type enzyme [10]) [10] D192G ( the mutation causes an enhanced feedback-resistant g-glutamyl kinase activity and conferrs an analogue-resistant phenotype to an Escherichia coli transformant containing the mutated gene [1]) [1] E153A ( reduction in catalytic activity. 555.5fold increase in Ki -value for l-Pro compared to wild-type enzyme [10]) [10] E153G ( reduction in catalytic activity. 555.5fold increase in Ki -value for l-Pro compared to wild-type enzyme [10]) [10] E153K ( reduction in catalytic activity. 3444.4fold increase in Ki -value for l-Pro compared to wild-type enzyme [10]) [10] I149F ( 222.2fold increase in Ki -value for l-Pro compared to wildtype enzyme [10]) [10] I149F ( drastic reduction in specific activity. 222.2fold increase in Ki -value for l-Pro compared to wild-type enzyme [10]) [10] I79T ( reduction in catalytic activity. 21.1fold increase in Ki -value for l-Pro compared to wild-type enzyme [10]) [10] L154S ( reduction in catalytic activity. 1000fold increase in Ki -value for l-Pro compared to wild-type enzyme [10]) [10] M94T ( reduction in catalytic activity. 255.6fold increase in Ki -value for l-Pro compared to wild-type enzyme [10]) [10] S159P ( reduction in catalytic activity. 211.1fold increase in Ki -value for l-Pro compared to wild-type enzyme [10]) [10]

355

Glutamate 5-kinase



2.7.2.11

   biotechnology ( the D154N mutation results in a prominant increase in cell viability after freezing at -20 C compared to the viability of the cells harboring the wild-type PRO1 gene, method for breeding novel freeze-tolerant yeast strains [11]; an artificial bifunctional enzyme, g-glutamyl kinase/g-glutamyl phosphate reductase, improves NaCl tolerance when expressed in Escherichia coli [12]) [11, 12]

7        , -70 C, stable for several months [2] , -40 C, 40% loss of activity within 2 months, then remains stable [5]

! 

[1] Massarelli, I.; Forlani, G.; Ricca, E.; De Felice, M.: Enhanced and feedbackresistant g-glutamyl kinase activity of an Escherichia coli transformant carrying a mutated proB gene of Streptococcus thermophilus. FEMS Microbiol. Lett., 182, 143-147 (2000) [2] Smith, C.J.; Deutch, A.H.; Rushlow, K.E.: Purification and characteristics of a g-glutamyl kinase involved in Escherichia coli proline biosynthesis. J. Bacteriol., 157, 545-551 (1984) [3] Seddon, A.P.; Zhao, K.Y.; Meister, A.: Activation of glutamate by g-glutamate kinase: formation of g-cis-cycloglutamyl phosphate, an analog of gglutamyl phosphate. J. Biol. Chem., 264, 11326-11335 (1989) [4] Hayzer, D.J.; Moses, V.: The enzymes of proline biosynthesis in Escherichia coli. Their molecular weights and the problem of enzyme aggregation. Biochem. J., 173, 219-228 (1978) [5] Krishna, R.V.; Leisinger, T.: Biosynthesis of proline in Pseudomonas aeruginosa. Partial purification and characterization of g-glutamyl kinase. Biochem. J., 181, 215-222 (1979) [6] Vasakova, L.; Stefl, M.: Glutamate kinases from winter-wheat leaves and some properties of the proline-inhibitable glutamate kinase. Collect. Czech. Chem. Commun., 47, 349-359 (1982) [7] Stefl, M.; Vasakova, L.: Allosteric regulation of proline-inhibitable glutamate kinase from winter-wheat leaves by l-proline, adenosine diphosphate and low temperature. Collect. Czech. Chem. Commun., 47, 360-369 (1982) [8] Stefl, M.; Vasakova, L.: Regulation of proline-inhibitable glutamate kinase (EC 2.7.2.11, ATP: g-l-glutamate phosphotransferase) of winter wheat leaves by monovalent cations and l-proline. Collect. Czech. Chem. Commun., 49, 2698-2708 (1984) [9] Garcia-Rios, M.; Fujita, T.; LaRosa, P.C.; Locy, R.D.; Clithero, J.M.; Bressan, R.A.; Csonka, L.N.: Cloning of a polycistronic cDNA from tomato encoding

356

2.7.2.11

Glutamate 5-kinase

g-glutamyl kinase and g-glutamyl phosphate reductase. Proc. Natl. Acad. Sci. USA, 94, 8249-8254 (1997) [10] Fujita, T.; Maggio, A.; Garcia-Rios, M.; Stauffacher, C.; Bressan, R.A.; Csonka, L.N.: Identification of regions of the tomato g-glutamyl kinase that are involved in allosteric regulation by proline. J. Biol. Chem., 278, 14203-14210 (2003) [11] Morita, Y.; Nakamori, S.; Takagi, H.: l-proline accumulation and freeze tolerance of Saccharomyces cerevisiae are caused by a mutation in the PRO1 gene encoding g-glutamyl kinase. Appl. Environ. Microbiol., 69, 212-219 (2003) [12] Meijer, P.-J.; Lilius, G.; Holmberg, N.; Bulow, L.: An artificial bifunctional enzyme, g-glutamyl kinase/g-glutamyl phosphate reductase, improves NaCl tolerance when expressed in Escherichia coli. Biotechnol. Lett., 18, 11331138 (1996) [13] Yaklichkin, S.Y.; Zimina, M.S.; Neumyvakin, L.V.: Proline biosynthesis gene proB of thermophilic bacterium Thermus ruber: cloning, sequencing, and properties of encoded g-glutamylphosphate kinase. Mol. Biol., 33, 628-635 (1999)

357

   * % %,

 

     2.7.2.12     diphosphate:acetate phosphotransferase    acetate kinase (diphosphate)   

acetate kinase (PPi) phosphotransferase, pyrophosphate-acetate pyrophosphate-acetate phosphotransferase pyrophosphate:acetate phosphotransferase     57657-58-6

   Entamoeba histolytica (strain H200 [1]) [1]

       !   "   diphosphate + acetate = phosphate + acetyl phosphate     phospho group transfer     

 phosphate + acetyl phosphate ( much greater activity in the direction of acetate formation [1]) (Reversibility: r [1]) [1] # diphosphate + acetate [1]  Additional information ( no activity with ADP, GDP, UDP, IDP or CDP [1]) [1] # ? ' ( 

MgCl2 ( employed in assay mixture [1]) [1]

358

2.7.2.12

Acetate kinase (diphosphate)

/01 *', 0.06 (acetyl phosphate, pH 7, 30 C [1]) [1] 2.2 (phosphate, pH 7, 30 C [1]) [1]

5 $   .#   .'  .

  

# !   [1]

7        , 4 C, under N2 , stable for a week [1]

! 

[1] Reeves, R.; Guthrie, J. D.: Acetate kinase (pyrophosphate). A fourth pyrophosphate-dependent kinase from Entamoeba histolytica. Biochem. Biophys. Res. Commun., 66, 1389-1395 (1975)

359

8 0  

 

     2.7.2.13     ATP:l-glutamate 1-phosphotransferase    glutamate 1-kinase   

kinase (phosphorylating), glutamate 1    80700-24-9

   Hordeum vulgare (L. cv. Svalöfs Bonus [1]) [1]

       !   "   ATP + l-glutamate = ADP + a-l-glutamyl phosphate     phospho group transfer      

 ATP + l-glutamate (Reversibility: ? [1]) [1] # ADP + a-l-glutamyl phosphate     

 ATP + l-glutamate (Reversibility: ? [1]) [1] # ADP + a-l-glutamyl phosphate

5 $   .#   .'  . .

 seed [1]

360

  

2.7.2.13

Glutamate 1-kinase

6 "  plastid stroma [1]

! 

[1] Wang, W.-Y.; Gough, S.P.; Kannangara, C.G.: Biosynthesis of D-aminolevulinate in greening barley leaves IV. Isolation of three soluble enzymes required for the conversion of glutamate to D-aminolevulinate. Carlsberg Res. Commun., 46, 243-257 (1981)

361

= %0% 0!0   

 3

     2.7.2.14     ATP:branched-chain-fatty-acid 1-phosphotransferase    branched-chain-fatty-acid kinase   

branched-chain fatty acid kinase isobutyrate kinase kinase (phosphorylating), branched-chain fatty acid kinase, branched-chain fatty acid (phosphorylating) Additional information (cf. EC 2.7.2.7)     84177-54-8

   Spirochaeta sp. (MA-2 [1]) [1]

       !   "   ATP + 2-methylpropanoate = ADP + 2-methylpropanoyl phosphate     phospho group transfer     

 ATP + 2-methylbutanoate (Reversibility: ? [1]) [1] # ADP + 2-methylbutanoyl phosphate [1]  ATP + butanoate (Reversibility: ? [1]) [1] # ADP + butanoyl phosphate  ATP + isobutanoate (Reversibility: ? [1]) [1] # ADP + isobutanoyl phosphate  ATP + isopentanoate (Reversibility: ? [1]) [1]

362

2.7.2.14

#  #  #  #  #  #  #

Branched-chain-fatty-acid kinase

ADP + isopentanoyl phosphate ATP + pentanoate (Reversibility: ? [1]) [1] ADP + pentanoyl phosphate ATP + propionate (Reversibility: ? [1]) [1] ADP + propanoyl phosphate CTP + isobutanoate ( 103% of the activity with ATP [1]) [1] CDP + isobutanoyl phosphate GTP + isobutanoate ( 70% of the activity with ATP [1]) (Reversibility: ? [1]) [1] GDP + isobutanoyl phosphate ITP + isobutanoate ( 68% of the activity with ATP [1]) [1] IDP + isobutanoyl phosphate Additional information ( no actrivity with acetate [1]) [1] ?

' ( 

Co2+ ( 65% of the activity with Mn2+ [1]) [1] Cu2+ ( 58% of the activity with Mn2+ [1]) [1] Mg2+ ( 96% of the activity with Mn2+ [1]) [1] Mn2+ ( divalent cation required, highest activity with Mn2+ [1]) [1] Zn2+ ( 42% of the activity with Mn2+ [1]) [1] /01 *', 1.8 (ATP, pH 7.5, 30 C [1]) [1] 4.3 (isobutyrate, pH 7.5, 30 C [1]) [1] 9.5 (isovalerate, pH 7.5, 30 C [1]) [1] 10.8 (2-methylbutyrate, pH 7.5, 30 C [1]) [1] 12.5 (valerate, pH 7.5, 30 C [1]) [1] 14.3 (propionate, pH 7.5, 30 C [1]) [1] 16.9 (butyrate, pH 7.5, 30 C [1]) [1] 20   7.2 [1] 20  6-8.5 ( more than 80% of maximal activity at pH 6.0 and 8.5 [1]) [1]

3  "  ' 4 % 76000 ( gel filtration [1]) [1]

7    )     45 ( 30 min, 25% loss of activity [1]) [1] 60 ( 30 min, complete loss of activity [1]) [1] 363

Branched-chain-fatty-acid kinase

2.7.2.14

8     !  , complete loss of activity if purified through columns of Sephacryl S-300 at room temperature [1] , dialysis, 6 h at 5 C, significant loss of activity [1]     , -50 C, crude cell extract stable for 2 months [1]

! 

[1] Harwood, C.S.; Canale-Parola, E.: Properties of acetate kinase isozymes and a branched-chain fatty acid kinase from a spirochete. J. Bacteriol., 152, 246254 (1982)

364

8    



     2.7.3.1     ATP:guanidinoacetate N-phosphotransferase    guanidinoacetate kinase   

glycocyamine kinase kinase, guanidinoacetate (phosphorylating)     9026-60-2

  





Perinereis sp. (polychaete [1]) [1] Nephtys cocea [2, 3] Polycelis cornuta [4] Myxicola infundibulum [4] Perinereis brevicirris (three isoforms [5]) [5] Neanthes diversicolor [6]

       !   "   ATP + guanidinoacetate = ADP + phosphoguanidinoacetate     phospho group transfer      

 ATP + guanidinoacetate (Reversibility: ? [1]) [1] # ADP + phosphoguanidinoacetate [1]     

 ADP + phosphocreatine ( poor substrate [3]) (Reversibility: ? [3]) [3] # ATP + ? 365

Guanidinoacetate kinase

2.7.3.1

ADP + phosphoguanidinoacetate (Reversibility: ? [3]) [3] ATP + guanidinoacetate [3] ATP + guanidine (Reversibility: r [6]; ? [4]) [4, 6] ADP + phosphoguanidine [6] ATP + guanidinoacetate ( specific for guanidinoacetate, ITP 2%, GTP 0.5% of ATP activity [1]; not: arginine, creatine, lombricine [3]) (Reversibility: r [3]; ? [1,2,4]) [1-4] # ADP + phosphoguanidinoacetate [1]  #  # 

$ %  

3-guanidinopropionate ( slight [1]) [1] 4-guanidinobutyrate ( slight [1]) [1] 5,5'-dithiobis(2-nitrobenzoate) [1] AMP [1] Ca2+ [1] Hg2+ [1] l-arginine ( slight [1]) [1] N-bromosuccinimide [1] N-ethylmaleimide [3] NH2 OH [3] agmatine [1] chloroacetophenone [3] creatine ( slight [1]) [1] guanidine ( slight [1]) [1] iodoacetamide [1] iodoacetic acid [3] methylguanine ( slight [1]) [1] p-chloromercuribenzoate [1, 3] phenylhydrazine ( inhibits phosphorylation of ADP [3]) [3] tauroguanine ( slight [1]) [1] ' ( 

Ca2+ ( 29% of Mg2+ -activation [1]; slightly [3]) [1, 3] Mg2+ ( 5 mM, activation [1]) [1, 3] Mn2+ ( 5 mM, activation [1]) [1, 3]   !   &  *-., 45.3 ( pH 8.1, 25 C [1]) [1] 65 [2] /01 *', 0.33 (guanidinoacetate) [3] 0.8 (ATP, pH 8.1, 25 C [1]) [1] 3.3 (ATP) [3] 4.1 (guanidinoacetate, pH 8.1, 25 C [1]) [1] 6.8 (phosphoguanidinoacetate) [3] 10 (ADP) [3]

366

2.7.3.1

Guanidinoacetate kinase

20   6.8 ( phosphorylation of ADP [3]) [3] 8.1 ( phosphorylation of guanidinoacetate [1]) [1] 8.9 ( phosphorylation of guanidinoacetate [3]) [3] 20  6.5-9 ( less than 50% of maximal activity above and below [1]) [1] )     * , 22 ( phosphorylation of guanidinoacetate [3]) [3] 35 ( phosphorylation of guanidinoacetate [1]) [1] 40 ( optimum above, phosphorylation of ADP [3]) [3] )    * , 16-32 ( less than 50% of maximal activity above and below, phosphorylation of guanidinoacetate [3]) [3]

3  "  ' 4 % 79000 ( sucrose density gradient centrifugation [2]) [2] 80000 ( gel filtration [6]) [6] 82000 ( gel filtration [2]) [2] 87500 ( amino acid composition [2]) [2] 89150 ( equilibrium sedimentation centrifugation [2]) [2] 90000 ( gel filtration [1]) [1]  

dimer ( 1 * 47000 + 1 * 45000, SDS-PAGE [1]; 1 * 42200 + 1 * 43800, SDS-PAGE [6]) [1, 6]

5 $   .#   .'  .

  

.

 muscle [2-4] # !   [1] [2] [5] [6]   [6]

367

Guanidinoacetate kinase

2.7.3.1

7    20   5.5-9.5 [1] )     35 ( stable up to [1]) [1] 40 ( 20 min, 4% loss of activity [1]) [1] 45 ( 20 min, 28% loss of activity [1]) [1] 50 ( inactivation [1]) [1]     , -20 C, 20 mM Tris-acetate buffer, pH 8.0, 1 mM dithiothreitol, 5% glycerol, several months [1] , 4 C, ammonium sulfate precipitate [2]

! 

[1] Shirokane, Y.; Nakajima, M.; Mizusawa, K.: Purification and properties of guanidinoacetate kinase from a polychaete, Perineis sp.. Agric. Biol. Chem., 55, 2235-2242 (1991) [2] Pradel, L.-A.; Kassab, R.; Conlay, C.; van Thoai, N.: Properties and amino acid composition of purified ATP: guanidinoacetate phosphotransferase. Biochim. Biophys. Acta, 154, 305-314 (1968) [3] Pradel, L.-A.; Kassab, R.; van Thoai, N.: On ATP:guanidinoacetate N-phosphotransferase. Biochim. Biophys. Acta, 81, 86-95 (1964) [4] Virden, R.; Watts, D.C.: Distribution of guanidine-adenosine triphosphate phosphotransferases and adenosintriphosphatase (ATPase) in animals from several phyla. Comp. Biochem. Physiol., 13, 161-77 (1964) [5] Furukohri, T.; Suzuki, T.: Preparation of glycocyamine kinase from polychaete, Perinereis brevicirris. Rep. USA Mar. Biol. Inst. Kochi Univ., 9, 215ff (1987) [6] Suzuki, T.; Nishimura, Y.; Umekawa, M.; Yamamoto, Y.; Kawamichi, H.; Furukohri, T.: Evolution of phosphagen kinase VII. Isolation of glycocyamine kinase from the polychaete Neanthes diversicolor and the cDNA-derived amino acid sequences of a and b chains. J. Protein Chem., 18, 13-19 (1999)

368

   



     2.7.3.2     ATP:creatine N-phosphotransferase    creatine kinase   

ATP:creatine phosphotransferase BB-CK CK CK-BB CK-MB CK-MM CKMiMi MB-CK MM-CK Mi-CK MiMi-CK adenosine triphosphate-creatine transphosphorylase creatine phosphokinase creatine phosphotransferase kinase, creatine (phosphorylating) phosphocreatine kinase     9001-15-4

   Bos taurus (calf [37]; two isoforms, interconversion by reversible oxidation of protein sulfhydryl groups [31]) [1, 14, 15, 25, 27, 31, 37, 68] Gallus gallus [1-7, 15, 40, 57, 74] Homo sapiens (three isoforms [35]) [1, 10, 13, 15, 18, 21, 35, 37, 44, 52, 60, 66, 69, 72, 73] Sus scrofa [1, 15-17] Rattus norvegicus [1, 15, 19, 30, 33, 50, 51, 53, 57, 65, 70]

369

Creatine kinase

2.7.3.2

Oryctolagus cuniculus [1, 8, 12, 15, 24, 27-29, 32, 34, 36, 37, 55, 57, 58, 64, 75, 76] Columba livia (pigeon [1]) [1, 57, 68] Strongylocentrotus purpuratus (sea urchin, two isoforms [62]) [1, 62] Discopyge tschudii [9] Torpedo marmorata [9] Canis familiaris [11, 21, 23, 59] trout [15] Papio annubis (monkey [15]) [15] Mus musculus [20, 57, 63] Equus caballus [22] Lepomis cyanellus (green sunfish [26]) [26] Scyliorhinus cani (dogfish [38]) [38] Cyprinus carpio (mirror carp, three isoforms [41]) [41, 42] mammalia [39, 49] Pagrus major (red sea bream [42]) [42] Scomber japonicus (pacific mackerel [42]) [42] Xenopus laevis [43] Eidolon helvum (tropical fruit bat [45]) [45] Ginglymostoma cirratum (nurse shark [46]) [46] Pseudemys scripta (turtle [47]) [47] Strongylocentrotus purpuratus [48] Chaenocephalus aceratus (Antarctic icefish [54]) [54] Clupea harengus (herring [56]) [56] frog [57] echinodermata [77] Danio rerio (zebrafish [61]) [61] Torpedo californica [67, 71]

       !   "   ATP + creatine = ADP + phosphocreatine (N-ethylglycocyamine can also act as acceptor; mitochondrial enzymes, mechanism, overview [1]; enzyme is functionally coupled to ouabain-inhibited (Na+ ,K+ )-ATPase [30]; kinetic model of reaction [33]; mechanism [55,76])     phospho group transfer      

 ATP + creatine ( physiological roles: 1. buffering of ADP/ATP ratio, 2. transport of high-energy phosphates from sites of ATP production to sites of ATP consumption [1]; regeneration of ATP as primary energy source [49]; coupled to (Na+ ,K+ )ATPase system [30]; mitochondrial model of CK in energy transport [6]; role in anaerobic metabolism [47]; 370

2.7.3.2

Creatine kinase

overview on physiological roles [57]; evolution of enzyme, phylogenetics [77]) (Reversibility: r [1, 6, 30]; ? [47, 49, 77]) [1, 6, 30, 47, 49, 57, 77] # ADP + phosphocreatine     

 ATP + creatine ( creatine cannot be replaced by creatinine [34]; Mg-complexes of ATP and ADP are the true substrates for the mitochondrial enzymes [33]; ATP required as MgATP2- [9, 15, 25, 33, 34, 46]) (Reversibility: r [1, 7, 8, 15, 18, 21, 25, 33, 34]; ? [9-14, 16, 17, 19, 20, 22-24, 26-32, 35-42, 46, 48, 54, 56]) [1-42, 46, 48, 54, 56] # ADP + creatine phosphate ( in the reverse direction ADP can be replaced by IDP with 18% efficiency, ADP cannot be replaced by GDP, CDP, UDP, dTDP [15]) [1, 15]  Additional information ( ATP undergoes substrate channelling between enzyme and myosin ATPase [65]) [65] # ? $ %  

4,4'-dithiodipyridine [22] 5,5'-dithiobis(2-nitrobenzoate) [22, 41, 45] Bis-Tris [18] Br- [18] Ca2+ [18] Cl- ( inactivation at -17 C [32]) [18, 32, 46] Co2+ [42] Cu2+ [18] F- [18] Fe3+ [18] I- [18] LiCl ( inactivation due to subunit dissociation, mechanism [64]) [64] MOPS buffer ( i.e. 3-(N-morpholino)propane sulfonate [18]) [18] N-ethylmaleimide [19] NO2- [18] NO-3 ( inactivation at -17 C [32]) [32, 46] NaCl ( inactivation due to subunit dissociation, mechanism [64]) [64] Pipes buffer ( i.e. 1,4-piperazine diethanesulfonic acid [18]) [18] SO23- [18] SO24- [18] Tris [18] Zn2+ [42] catechin [50] chromium ADP ( competitive to MgADP- [36]) [36] chromium ATP ( competitive to MgATP2- [36]) [36]

371

Creatine kinase

2.7.3.2

creatinine phosphate ( competitive to phosphocreatine [34]; competitive to MgATP2- [36]) [34, 36] imidazole [18] iodoacetamide ( protection by MgATP2-, MgADP-, urea [38]) [24, 29, 38] iodoacetic acid [19, 24, 29] iodoethane [24] iodomethane [24, 29] luteolin [50] p-hydroxymercuribenzoate [19] quercetin ( mechanism, role of radicals [50]) [50] taxifolin [50] ! .  %  

NADH ( activation of cytosolic enzyme in the direction of ATPformation [47]) [47]  &    

NADH ( lowers Km for phosphocreatine 3-fold [47]) [47] ' ( 

Co2+ ( can substitute for Mg2+ [45]) [45] Mg2+ ( required, regulatory effect of Mg2+ -concentration [33]; required as MgATP [9, 15, 25, 33, 46]) [9, 15, 18, 25, 33, 42, 45, 46] Mn2+ ( required [18]) [18] Zn2+ ( can substitute for Mg2+ [45]) [45]   !   &  *-., 15 ( synthesis of phosphocreatine [15]) [15] 49.5 ( synthesis of ATP [15]) [15] 250 ( 30 C, pH 7.0 [7]) [7] 410 ( 30 C [13]) [13] 620 [11] Additional information ( assay method [21, 39, 62]; activity in whole muscle fibers and myofibrillar activity [53]) [4, 5, 9-11, 16, 19-23, 25, 26, 30, 31, 35, 37-39, 41, 48, 53, 62] /01 *', 0.015 (MgADP-, 30 C [25]) [25] 0.017 (MgADP-, pH 7.4, dimeric form [68]) [68] 0.027 (ADP, 30 C [67]) [67] 0.042 (MgATP2-, pH 7.4, dimeric form [68]) [68] 0.043 (MgADP-, pH 7.4, octameric form [68]) [68] 0.047 (ADP) [27] 0.051-0.052 (MgADP-, 30 C, pH 7.4 [33]) [33] 0.056 (MgATP2-, 30 C [25]) [25] 0.06 (ADP, 0.5 C, pH 7.6 [54]) [54] 0.065 (ADP) [27]

372

2.7.3.2

Creatine kinase

0.082 (MgATP2-, pH 7.4, octameric form [68]) [68] 0.11 (ATP, 25 C, pH 8.0, ubiquitous mitochondrial isoform [60]) [60] 0.15 (MgADP- ) [15] 0.2-0.33 (ADP) [42] 0.22 (MgADP-, acetylcholine receptor membrane-asscociated enzyme [9]) [9] 0.23 (creatine phosphate, pH 7.4, dimeric form [68]) [68] 0.31 (creatine phosphate, 30 C [25]) [25] 0.4 (creatine phosphate) [19] 0.4 (creatine phosphate, pH 7.0, 25 C [4]) [4, 19] 0.49-0.5 (creatine phosphate, 30 C, pH 7.4 [33]) [33] 0.54 (MgADP-, soluble enzyme from muscle [9]) [9] 0.68 (ATP, 25 C, pH 8.0, sarcomeric mitochondrial isoform [60]) [60] 0.68 (creatine phosphate, pH 7.4, octameric form [68]) [68] 0.73 (MgATP2-, 30 C, pH 7.4 [33]) [33] 0.8 (ATP, 30 C [67]) [67] 1.01 (creatine, 25 C, pH 8.0, ubiquitous mitochondrial isoform [60]) [60] 1.07 (creatine phosphate, 37 C, ubiquitous isoform [66]) [66] 1.19 (creatine phosphate, 37 C, sarcomeric isoform [66]) [66] 1.2 (MgADP- ) [46] 1.6 (MgATP2- ) [46] 1.7 (MgATP2- ) [15] 1.9-2.2 (creatine phosphate, acetylcholine receptor membraneassociated enzyme [9]) [9, 27] 2-10.6 (creatine phosphate) [42] 2.5 (creatine, soluble enzyme from muscle [9]) [9] 3 (creatine phosphate) [15] 3.4 (creatine, pH 7.4, dimeric form [68]) [68] 3.7 (creatine phosphate, 30 C [67]) [67] 4.5 (creatine, 30 C [25]) [25] 4.9-5 (creatine, 30 C, pH 7.4 [33]) [33] 7.31 (creatine, 25 C, pH 8.0, sarcomeric mitochondrial isoform [60]) [60] 8 (creatine) [15] 8.1 (creatine, pH 7.4, octameric form [68]) [68] 12 (creatine) [46] 17 (creatine phosphate, 0.5 C, pH 7.6 [54]) [54] 50 (creatine phosphate) [46] 79 (creatine, 30 C [67]) [67] Additional information ( effect of temperature on values for MgATP2- and creatine [38]; kinetics [35, 39, 44]; overview [1]; temperature dependence of reaction, in vivo measurements [51]; dextran strongly increases Km [70]) [1, 35, 36, 38, 39, 44, 48, 51, 70, 76] 373

Creatine kinase

2.7.3.2

20   6-6.5 ( synthesis of MgATP2- [25]) [25] 6-7 ( synthesis of ATP [1]) [1] 6.3 ( synthesis of MgATP2- [4]) [4] 6.7 ( synthesis of MgATP2- [15]) [15] 7-7.5 [42] 7.5-9 ( synthesis of phosphocreatine [1]) [1] 7.6-7.7 [54] 8 ( synthesis of phosphocreatine [25]) [25] 8-8.3 ( synthesis of phosphocreatine [4]) [4] 8.7 ( synthesis of phosphocreatine [15]) [15] Additional information ( substrate channelling under different pH conditions [65]) [65] )     * , 0.5 [54] 42 ( synthesis of phosphocreatine [4]) [4] 42-45 ( synthesis of ATP [4]) [4]

3  "  ' 4 % 64000 ( gel filtration [25]) [25] 76000-78000 ( gel filtration [22]) [22] 78000 ( isozyme Mi-CK, dimeric form, scanning transmission electron microscopy [6]) [6] 78000-80000 [45] 79700 ( and also 371000, gel filtration [52]) [52] 80000 ( low speed sedimentation equilibrium centrifugation [27]) [27] 82000 ( sedimentation equilibrium centrifugation [17]) [17] 84000 ( isozyme MiMi-CK, equilibrium centrifugation [7]; gel filtration [23]) [7, 23] 84000-85000 ( isozyme MiMi-CK, sedimentation equilibrium centrifugation, gel filtration [15]) [15] 84500 ( high speed and low speed sedimentation equilibrium centrifugation [22]) [22] 85000 ( isozyme Mia-CK, dimeric form, gel filtration, analytical ultracentrifugation [4]) [4] 85100 ( sedimentation equilibrium centrifugation [41]) [41] 86000 ( gel filtration, also 346000 [60]) [60] 89000 ( isozyme Mia-CK, dimeric form, scanning transmission electron microscopy [6]) [6] 100000 ( gel filtration [20]) [20] 126000-145000 ( flagellar isozyme, sucrose density gradient centrifugation, SDS-PAGE [48]) [48]

374

2.7.3.2

Creatine kinase

240000 ( head isozyme, calculation from Stokes' radius and partial specific volume [48]) [48] 306000-352000 ( isozyme Mia-CK, octameric form, gel permeation chromatography, scanning transmission electron microscopy [4]) [4] 328000-340000 ( isoenzyme Mi-CK, octameric form, sedimentation velocity analysis, sedimentation equilibrium centrifugation, scanning transmission electron microscopy [6]) [6] 346000 ( gel filtration, also 86000 [60]) [60] 360000 ( isozyme Mia-CK, octameric form, gel filtration [5]) [5] 371000 ( and also 79700, gel filtration [52]) [52] Additional information ( overview [1]; structural properties, sulfhydryl groups [32]) [1, 32]  

dimer ( 2 * 35000, bovine, SDS-PAGE [25]; 2 * 40000, bovine, SDS-PAGE [27]; 2 * 40000-43000, SDS-PAGE [9]; 1 * 41000 + 1 * 42000, isozyme CK-II [43]; 2 * 41000, isozyme CK-IV [43]; 2 * 41000, SDS-PAGE [13]; 2 * 41500, SDS-PAGE [4]; 2 * 42000, isozyme CK-III [43]; 2 * 42000, SDS-PAGE [5]; 2 * 42000, SDS-PAGE [15]; 2 * 41000, SDS-PAGE [23]; 2 * 42500, [45]; 2 * 43000, SDS-PAGE [7]; 2 * 43000, SDS-PAGE, but also octamer [60]; 2 * 43600, SDS-PAGE, but also octamer, electron microscopy [52]; 2 * 43000-44000, SDS-PAGE, presence of 2-mercaptoethanol [17]; 2 * 44000, SDS-PAGE, presence of 2-mercaptoethanol, high speed sedimentation equilibrium centrifugation of urea-treated enzyme [22]; 2 * 43195, calculated from sequence of cDNA [40]; 2 * 49000, SDS-PAGE [19]; 2 * 50000, SDS-PAGE [20]; crystallization data, without ATP [3]) [3-5, 7, 9, 13, 15, 17, 19, 20, 22, 23, 25, 27, 40, 43, 45, 60] monomer ( 1 * 145000, flagellar isozyme, SDS-PAGE [48]) [48] octamer ( crystallization data [2]; crystallization data, with ATP [3]; 8 * 42000, SDS-PAGE, octameric structure dissociates during storage at -20 C, pH above 8.5, protein concentration below 0.3 mg/ml to dimeric form [5]; 8 * 43600, SDS-PAGE, but also dimer, electron microscopy [52]; 8 * 43000, SDS-PAGE, also as dimer [60]) [2, 3, 5, 52, 60] polymer ( x * 47000, head mitochondrial isozyme, SDS-PAGE) [48] Additional information ( hydrolytic cleaveage is responsible for conversion of isoform MM1 to MM2 and MM3 [11]; overview on isoforms [57]) [11, 57] #      !   Additional information ( enzyme activity depends on free sulfhydryl groups [32,41,45]; titration of two thiol groups leads to almost complete loss of activity [38]) [38, 32, 41, 45]

375

Creatine kinase

5 $   .#   .'  .

2.7.3.2

  

.

 aorta [1] blood plasma [11] brain [1, 4, 18, 21, 46, 51, 66, 69] electric organ [9] head [48] heart ( enzyme variants I-IV [10]) [1-3, 5-7, 10, 12-15, 18, 21, 23, 25, 27, 30, 31, 33-35, 44, 47, 50, 52, 66, 68, 70] intestine [1] liver [1] muscle ( skeletal [1, 8, 10-12, 16-18, 22, 24, 26, 28, 29, 32, 37, 40, 44, 65]; commercial preparation [36]; enzyme variants I-IV [10]; skinned psoas muscle [53]; two muscle-specific isoforms [54]) [1, 8, 10-12, 16-18, 22, 24, 26, 28, 29, 32, 36, 37, 40-42, 44-46, 53-55, 64, 65, 68] placenta [1] retina [1] sperm flagellum [48, 62] spermatozoon [1, 48, 56, 62] uterus ( of immature animals [19]) [19] Additional information ( overview tissue distribution of mitochondrial enzyme [1]) [1] 6 "  cytoplasm [1, 5, 14, 16-22, 47] membrane ( acetylcholine receptor membrane [9]) [9] mitochondrion ( accumulated in contact sites between inner and outer mitochondrial membrane [1, 5]; head isozyme [48]; overview on intramitochondrial localization [68]) [1-7, 13, 15, 18, 23, 25, 31, 33, 40, 47, 48, 52, 60, 63, 68, 70] soluble [1, 11, 43] Additional information ( overview [57]) [57] # !   (2 interconvertible forms of enzyme formed by reversible oxidation of sulfhydryl groups [31]; preparation of catalytically active hybrids of brain and muscle enzymes [37]) [25, 31, 37] (isozyme Mia-CK [4]) [4, 5, 7] (5 varieties of isozyme MM-CK [10]; preparation of catalytically active hybrids of brain and muscle enzymes [37]) [10, 13, 15, 21, 37] (isozyme MM-CK [17]) [17] [19] [8, 37] [62] [9]

376

2.7.3.2

Creatine kinase

[9] (isozyme MM-CK [11]) [11, 21, 23] [20] [26] [42] [42] [42] (isozyme CK-II, CK-III, CK-IV [43]) [43] [45] [46] (2 isozymes [48]) [48] (recombinant enzyme, inclusion bodies [67]) [67] (overview [1]) [1]    (reactivators are thiols, like N-acetylcysteine, b-mercaptoethanol, dithiothreitol, monothioglycerol, glutathione [18]) [18] (reactivation of 5,5'-dithiobis-(2-nitrobenzoic acid)-modified enzyme by excess of dithiothreitol, kinetics [58]) [58]   "  (isozyme MM-CK [14]) [14] (a dimeric and a octameric isoform [3]; brain-type isoform [74]) [2, 3, 74] (isozyme MiMi-CK [15]; ubiquituos mitochondrial isoform [72]; muscle isoform [73]) [15, 72, 73] (muscle isoform [75]) [8, 28, 75] (isozyme MM-CK [41]) [41] (in complex with a transistion-state analog [71]) [71] (overview: electron microscopy, X-ray crystallography [1]) [1]   [40] (brain isoform [69]) [69] [54] [61]     DH65 ( affinity to substrates almost like wild type enzyme, very little stability [69]) [69] DH65P66 ( 8-fold decreased affinity for creatine phosphate [69]) [69] N285A ( severe loss of activity [55]) [55] N285D ( severe loss of activity, ordered binding mechanism [55]) [55] N285Q ( severe loss of activity, random order mechanism, reduced affinity for second substrate [55]) [55] Additional information ( transgenic mice lacking mitochondrial enzyme or both mitochondrial and cytoplasmic enzyme [63]) [63]

377

Creatine kinase



2.7.3.2

   biotechnology ( stability of immobilized enzyme [12]; use as biomarker of sperm cell membrane degradation [56]) [12, 56] medicine ( possible roles in pathology [1]; enzyme properties relevant for analysis [18]; role of enzyme in severe left ventricular hypertrophy [59]; transgenic mice lacking mitochondrial enzyme or both mitochondrial and cytoplasmic enzyme, myocardial energy-recruiting mechanims [63]) [1, 18, 59, 63]

7    20   4.5-10.5 ( rapid inactivation above and below [15]) [15] 6-8 ( calf brain enzyme stable, stability can be extended to pH 5.5-9 by addition of 0.01 M 2-mercaptoethanol [37]) [37] Additional information ( comparison of various enzymes of various sources [37]) [37] )     23 ( isozyme MiMi-CK: 1 h, 15% loss of activity, 2 h, 34% loss of activity, 3 h, 66% loss of activity, isozyme BB-CK: 6 h, 32% loss of activity, isozyme MM-CK: no loss of activity [23]) [23] 35 ( calf brain: 0.01 M 2-mercaptoethanol enhances stability in pHrange 6-8 [37]) [37] 37 ( isozyme MiMi-CK: 10 min, 30% loss of activity, 20 min, 62% loss of activity, 80 min, 75% loss of activity, isozyme MM-CK: 80 min, 75% loss of activity [23]) [23] 45 ( soluble enzyme: half-life 4 min, immobilized enzyme: half-life 35 min [16]; 20 min, 80% residual activity for sarcomeric isoform, 90% residual activity for ubiquotous isoform [66]) [16, 66] 51 ( inactivation above [4]) [4] Additional information ( comparison of various enzymes of various sources [37]; enzymes from marine fishes are less thermostable than that of carp, the latter being more labile than the rabbit enzyme [42]; shark muscle isozyme marginally more resistant to temperature inactivation than brain isozyme [46]) [37, 42, 46] 8     !  , 2-mercaptoethanol enhances pH-stability [37] , dimeric enzyme stable to 1-2 M urea [6] , sensitive to denaturation [26]     , 4 C, 20 mM sodium phosphate buffer, pH 8.0, 10% loss of activity in 2 weeks, faster inactivation in presence of dithiothreitol [25] , 4 C, 10 mM MOPS buffer, pH 7.2, 2% v/v glycerol, 25 mM 2-mercaptoethanol, 0.1 mM EDTA, stable for more than 4 months [7]

378

2.7.3.2

Creatine kinase

, 4 C, octameric enzyme, protein concentration above 1 mg/ml, 1 mM 2mercaptoethanol, 0.2 mM EDTA, 0.26 M NaCl, 25 mM sodium phosphate buffer, pH 7.0 [5] , liquid N2 preserves octameric stucture, dissociation to dimer at higher temperatures [5] , 4 C, pH 7.0, 1 mM dithiothreitol or 14 mM 2-mercaptoethanol, at least 3 months [15] , -10 C, 5 mM Tris/HCl buffer, pH 8.7, 1 mM 2-mercaptoethanol, saturated ammonium sulfate solution [17] , 4 C, several weeks [19] , -17 C, inactivation in presence of chloride or nitrate [32] , -70 C, 0.05 M Tris/barbital buffer, pH 7.8, 0.01 M 2-mercaptoethanol, 17% loss of activity in 6 weeks [23]

! 

[1] Wyss, M.; Smeitink, J.; Wevers, R.A.; Wallimann, T.: Mitochondrial creatine kinase: a key enzyme of aerobic energy metabolism. Biochim. Biophys. Acta, 1102, 119-166 (1992) [2] Schnyder, T.; Winkler, H.; Gross, H.; Eppenberger, H.M.; Wallimann, T.: Crystallization of mitochondrial creatine kinase. Growing of large protein crystals and electron microscopic investigation of microcrystals consisting of octamers. J. Biol. Chem., 266, 5318-5322 (1991) [3] Schnyder, T.; Sargent, D.F.; Richmond, T.J.; Eppenberger, H.M.; Wallimann, T.: Crystallization and preliminary X-ray analysis of two different forms of mitochondrial creatine kinase from chicken cardiac muscle. J. Mol. Biol., 216, 809-812 (1990) [4] Wyss, M.; Schlegel, J.; James, P.; Eppenberger, H.M.; Wallimann, T.: Mitochondrial creatine kinase from chicken brain. Purification, biophysical characterization, and generation of heterodimeric and heterooctameric molecules with subunits of other creatine kinase isoenzymes. J. Biol. Chem., 265, 15900-15908 (1990) [5] Schlegel, J.; Zurbriggen, B.; Wegmann, G.; Wyss, M.; Eppenberger, H.M.; Wallimann, T.: Native mitochondrial creatine kinase forms octameric structures. I. Isolation of two interconvertible mitochondrial creatine kinase forms, dimeric and octameric mitochondrial creatine kinase: characterization, localization, and structure-function relationships. J. Biol. Chem., 263, 16942-16953 (1988) [6] Schnyder, T.; Engel, A.; Lustig, A.; Wallimann, T.: Native mitochondrial creatine kinase forms octameric structures. II. Characterization of dimers and octamers by ultracentrifugation, direct mass measurements by scanning transmission electron microscopy, and image analysis of single mitochondrial creatine kinase octamers. J. Biol. Chem., 263, 16954-16962 (1988) [7] Brooks, S.P.; Bennett, V.D.; Suelter, C.H.: Homogeneous chicken heart mitochondrial creatine kinase purified by dye-ligand and transition-state analog-affinity chromatography. Anal. Biochem., 164, 190-198 (1987) 379

Creatine kinase

2.7.3.2

[8] Hershenson, S.; Helmers, N.; Desmueles, P.; Stroud, R.: Purification and crystallization of creatine kinase from rabbit skeletal muscle. J. Biol. Chem., 261, 3732-3736 (1986) [9] Barrantes, F.J.; Braceras, A.; Caldironi, H.A.; Mieskes, G.; Moser, H.; Toren, E.C.; Roque, M.E.; Walliman, T.; Zechel, A.: Isolation and characterization of acetylcholine receptor membrane-associated (nonreceptor v2-protein) and soluble electrocyte creatine kinases. J. Biol. Chem., 260, 3024-3034 (1985) [10] Vaidya, H.; Dietzler, D.N.; Leykam, J.F.; Ladenson, J.H.: Purification of five creatine kinase-MM variants from human heart and skeletal muscle. Biochim. Biophys. Acta, 790, 230-237 (1984) [11] George, S.; Ishikawa, Y.; Perryman, M.B.; Roberts, R.: Purification and characterization of naturally occurring and in vitro induced multiple forms of MM creatine kinase. J. Biol. Chem., 259, 2667-2674 (1984) [12] Rudge, J.; Bickerstaff, G.F.: Thermal stability of immobilized creatine kinase. Biochem. Soc. Trans., 12, 311-313 (1984) [13] Grace, A.M.; Perryman, M.B.; Roberts, R.: Purification and characterization of human mitochondrial creatine kinase. A single enzyme form. J. Biol. Chem., 258, 15346-15354 (1983) [14] Gilliland, G.L.; Sjollin, L.; Olsson, G.: Crystallization and preliminary X-ray diffraction data of two crystal forms of bovine heart creatine kinase. J. Mol. Biol., 170, 791-793 (1983) [15] Blum, H.E.; Deus, B.; Gerok, W.: Mitochondrial creatine kinase from human heart muscle: purification and characterization of the crystallized isoenzyme. J. Biochem., 94, 1247-1257 (1983) [16] Takasawa, T.; Onodera, M.; Shiokawa, H.: Properties of three creatine kinases MM from porcine skeletal muscle. J. Biochem., 93, 389-395 (1983) [17] Takasawa, T.; Shiokawa, H.: Isolation and properties of creatine kinase from porcine skeletal muscle. J. Biochem., 90, 195-204 (1981) [18] Gerhardt, W.: Creatine kinase. Methods Enzym. Anal., 3rd Ed. (Bergmeyer, H.U., ed.), 3, 508-510 (1983) [19] Kumar, S.A.; O'Connor, D.L.; Seeger, J.I.; Beach, T.A.: Purification and characterization of creatine kinase, an estrogen-induced uterine protein (IP) from immature rats. Biochem. Biophys. Res. Commun., 111, 156-165 (1983) [20] Olson, E.N.; Lathrop, B.K.; Glaser, L.: Purification and cell-free translation of a unique high molecular weight form of the brain isozyme of creatine phosphokinase from mouse. Biochem. Biophys. Res. Commun., 108, 715723 (1982) [21] Roberts, R.: Purification of human and canine creatine kinase isozymes. Methods Enzymol., 90, 185-195 (1982) [22] Takasawa, T.; Fukushi, K.; Shiokawa, H.: Crystallization and properties of creatine kinase from equine skeletal muscle. J. Biochem., 89, 1619-1631 (1981) [23] Roerts, R.; Grace, A.M.: Purification of mitochondrial creatine kinase. Biochemical and immunological characterization. J. Biol. Chem., 255, 28702877 (1980)

380

2.7.3.2

Creatine kinase

[24] Reddy, S.R.R.; Watts, D.C.: Inhibition of creatine kinase by iodoalkanes. Further appraisal of the essential nature of the reactive thiol group. Biochim. Biophys. Acta, 569, 109-113 (1979) [25] Hall, N.; Addis, P.; DeLuca, M.: Mitochondrial creatine kinase. Physical and kinetic properties of the purified enzyme from beef heart. Biochemistry, 18, 1745-1751 (1979) [26] Fisher, S.E.; Whitt, G.S.: Purification of the creatine kinase isozymes of the green sunfish (Lepomis cyanellus) with Blue Sepharose CL-6B. Anal. Biochem., 94, 89-95 (1979) [27] Herasymowych, O.S.; Mani, R.S.; Kay, C.M.: Isolation, purification and characterization of creatine kinase from bovine cardiac muscle. Biochim. Biophys. Acta, 534, 38-47 (1978) [28] Burgess, A.N.; Liddell, J.M.; Cook, W.; Tweedlie, R.M.; Swan, I.D.A.: Creatine kinase. A new crystal form providing evidence of subunit structural homogeneity. J. Mol. Biol., 123, 691-695 (1978) [29] Reddy, S.R.R.; Watts, D.C.: Inhibition of rabbit muscle creatine kinase by iodomethane [proceedings]. Biochem. Soc. Trans., 6, 553-555 (1978) [30] Saks, V.A.; Lipina, N.V.; Sharov, V.G.; Smirnov, V.N.; Chazov, E.; Grosse, R.: The localization of the MM isozyme of creatine phosphokinase on the surface membrane of myocardial cells and its functional coupling to ouabaininhibited (Na+ , K+ )-ATPase. Biochim. Biophys. Acta, 465, 550-558 (1977) [31] Hall, N.; Addis, P.; DeLuca, M.: Purification of mitochondrial creatine kinase: two interconvertible forms of the active enzyme. Biochem. Biophys. Res. Commun., 76, 950-956 (1977) [32] Madelian, V.; Warren, W.A.: Properties of a structurally and functionally altered form of creatine kinase produced in solutions containing chloride and nitrate. Arch. Biochem. Biophys., 184, 103-110 (1977) [33] Saks, V.A.; Chernousova, G.B.; Gukovsky, D.E.; Smirnov, V.N.; Chazov, E.I.: Studies of energy transport in heart cells. Mitochondrial isoenzyme of creatine phosphokinase: kinetic properties and regulatory action of Mg2+ ions. Eur. J. Biochem., 57, 273-290 (1975) [34] Gercken, G.; Dªring, V.: Inhibition of creatine kinase by creatinine phosphate. FEBS Lett., 46, 87-91 (1974) [35] Witteveen, S.A.G.J.; Sobel, B.E.; DeLuca, M.: Kinetic properties of the isoenzymes of human creatine phosphokinase. Proc. Natl. Acad. Sci. USA, 71, 1384-1387 (1974) [36] Schimerlik, M.I.; Clelend, W.W.: Inhibition of creatine kinase by chromium nucleotides. J. Biol. Chem., 248, 8418-8423 (1973) [37] Keutel, H.J.; Okabe, K.; Jacobs, H.K.; Ziter, F.; Maland, L.; Kuby, S.A.: Studies on adenosine triphosphate transphosphorylases. XI. Isolation of the crystalline adenosine triphosphate-creatine transphosphorylases from the muscle and brain of man, calf, and rabbit; and a preparation of their enzymatically active hybrids. Arch. Biochem. Biophys., 150, 648-678 (1972) [38] Simonarson, B.; Watts, D.C.: Purification and properties of adenosine triphosphate-creatine phosphotransferase from muscle of the dogfish Scylliorhinus canicula. Biochem. J., 128, 1241-1253 (1972)

381

Creatine kinase

2.7.3.2

[39] Kuby, S.A.; Noltmann, E.A.: ATP-creatine transphosphorylase. The Enzymes, 2nd. Ed. (Boyer, P.D., ed.), 6, 515-603 (1962) [40] Hossle, J.P.; Schlegel, J.; Wegmann, G.; Wyss, M.; Bohlen, P.; Eppenberger, H.M.; Wallimann, T, Perriard, J.-C.: Distinct tissue specific mitochondrial creatine kinases from chicken brain and striated muscle with a conserved CK framework. Biochem. Biophys. Res. Commun., 151, 408-416 (1988) [41] Gosselin-Rrey, C.; Gerday, C.: Isolation and molecular properties of creatine kinase from carp white muscle. Biochim. Biophys. Acta, 221, 241-254 (1970) [42] Nakagawa, T.; Nagayama, F.: Enzymic properties of fish muscle creatine kinase. Comp. Biochem. Physiol. B, 98, 349-354 (1991) [43] Robert, J.; Kobel, H.R.: Purification and characterization of cytoplasmic creatine kinase isozymes of Xenopus laevis. Biochem. Genet., 26, 543-555 (1988) [44] Schneider, C.; Stull, G.A.; Apple, F.S.: Kinetic characterization of human heart and skeletal muscle CK isoenzymes. Enzyme, 39, 220-226 (1988) [45] Afolayan, A.; Daini, O.A.: Isolation and properties of creatine kinase from the breast muscle of tropicalfruit bat, Eidolon helvum. Comp. Biochem. Physiol. B Comp. Biochem., 85, 463-468 (1986) [46] Gray, K.A.; Grossman, S.H.; Summers, D.D.: Purification and characterization of creatine kinase isozymes from the nurse shark Ginglymostoma cirratum. Comp. Biochem. Physiol. B Comp. Biochem., 83, 613-620 (1986) [47] Storey, K.B.: Purification and properties of turtle heart creatine kinase. Role for the enzyme in glykolytic control. Int. J. Biochem., 6, 54-59 (1975) [48] Tombes, R.M.; Shapiro, B.M.: Enzyme termini of a phosphocreatine shuttle. Purification and characterization of two creatine kinase isozymes from sea urchin sperm. J. Biol. Chem., 262, 16011-16019 (1987) [49] Kenyon, G.L.; Reed, G.H.: Creatine kinase: structure-activity relationships. Adv. Enzymol. Relat. Areas Mol. Biol., 54, 367-426 (1983) [50] Miura, T.; Muraoka, S.; Fujimoto, Y.: Inactivation of creatine kinase induced by quercetin with horseradish peroxidase and hydrogen peroxide pro-oxidative and anti-oxidative actions of quercetin. Food Chem. Toxicol., 41, 759-765 (2003) [51] Buist, R.; Kroeker, S.; Peeling, J.: Temperature dependence of the creatine kinase reaction measured in rat brain in vivo by 31P NMR saturation transfer. Can. J. Chem., 77, 1887-1891 (1999) [52] Walterscheid-Muller, U.; Braun, S.; Salvenmoser, W.; Meffert, G.; Dapunt, O.; Gnaiger, E.; Zierz, S.; Margreiter, R.; Wyss, M.: Purification and characterization of human sarcomeric mitochondrial creatine kinase. J. Mol. Cell. Cardiol., 29, 921-927 (1997) [53] Gregor, M.; Mejsnar, J.; Janovska, A.; Zurmanova, J.; Benada, O.; Mejsnarova, B.: Creatine kinase reaction in skinned rat psoas muscle fibers and their myofibrils. Physiol.Res., 48, 27-35 (1999) [54] Winnard, P.; Cashon, R.E.; Sidell, B.D.; Vayda, M.E.: Isolation, characterization and nucleotide sequence of the muscle isoforms of creatine kinase from the Antarctic teleost Chaenocephalus aceratus. Comp. Biochem. Physiol. B, 134B, 651-667 (2003) 382

2.7.3.2

Creatine kinase

[55] Borders, C.L., Jr.; MacGregor, K.M.; Edmiston, P.L.; Gbeddy, E.R.K.; Thomenius, M.J.; Mulligan, G.B.; Snider, M.J.: Asparagine 285 plays a key role in transition state stabilization in rabbit muscle creatine kinase. Protein Sci., 12, 532-537 (2003) [56] Grzyb, K.; Rychlowski, M.; Biegniewska, A.; Skorkowski, E.F.: Quantitative determination of creatine kinase release from herring (Clupea harengus) spermatozoa induced by tributyltin. Comp. Biochem. Physiol. C, 134C, 207-213 (2003) [57] Ventura-Clapier, R.; Kuznetsov, A.; Veksler, V.; Boehm, E.; Anflous, K.: Functional coupling of creatine kinases in muscles: species and tissue specificity. Mol. Cell. Biochem., 184, 231-247 (1998) [58] Yang, Y.; Zhou, H.-M.: Reactivation kinetics of 5,5'-dithiobis-(2-nitrobenzoic acid)-modified creatine kinase reactivated by dithiothreitol. Biochim. Biophys. Acta, 1388, 190-198 (1998) [59] Ye, Y.; Wang, C.; Zhang, J.; Cho, Y.K.; Gong, G.; Murakami, Y.; Bache, R.J.: Myocardial creatine kinase kinetics and isoform expression in hearts with severe LV hypertrophy. Am. J. Physiol., 281, H376-H386 (2001) [60] Schlattner, U.; Eder, M.; Dolder, M.; Khuchua, Z.A.; Strauss, A.W.; Wallimann, T.: Divergent enzyme kinetics and structural properties of the two human mitochondrial creatine kinase isoenzymes. Biol. Chem., 381, 10631070 (2000) [61] Harder, G.; McGowan, R.: Isolation and characterization of the muscle-specific isoform of creatine kinase from the zebrafish, Danio rerio. Biochem.Cell Biol., 79, 779-782 (2001) [62] Tombes, R.M.: Isolation and characterization of sea urchin flagellar creatine kinase. Methods Cell Biol., 47, 467-472 (1995) [63] Bonz, A.W.; Kniesch, S.; Hofmann, U.; Kullmer, S.; Bauer, L.; Wagner, H.; Ertl, G.; Spindler, M.: Functional properties and [Ca2+ ]. (I). Metabolism of creatine kinase±KO mice myocardium. Biochem. Biophys. Res. Commun., 298, 163-168 (2002) [64] Couthon, F.; Clottes, E.; Vial, C.: High salt concentrations induce dissociation of dimeric rabbit muscle creatine kinase. Physico-chemical characterization of the monomeric species. Biochim. Biophys. Acta, 1339, 277-288 (1997) [65] Gregor, M.; Janovska, A.; Stefl, B.; Zurmanova, J.; Mejsnar, J.: Substrate channelling in a creatine kinase system of rat skeletal muscle under various pH conditions. Exp. Physiol., 88, 1-6 (2003) [66] Kanemitsu, F.; Mizushima, J.; Kageoka, T.; Okigaki, T.; Taketa, K.; Kira, S.: Characterization of two types of mitochondrial creatine kinase isolated from normal human cardiac muscle and brain tissue. Electrophoresis, 21, 266-270 (2000) [67] Wang, P.-F.; Novak, W.R.P.; Cantwell, J.S.; Babbitt, P.C.; McLeish, M.J.; Kenyon, G.L.: Expression of Torpedo californica creatine kinase in Escherichia coli and purification from inclusion bodies. Protein Expr. Purif., 26, 89-95 (2002) [68] Lipskaya, T.Y.: Mitochondrial creatine kinase: properties and function. Biochemistry, 66, 1098-1111 (2001) 383

Creatine kinase

2.7.3.2

[69] Mourad-Terzian, T.; Steghens, J.P.; Min, K.L.; Collombel, C.; Bozon, D.: Creatine kinase isoenzymes specificities: histidine 65 in human CK-BB, a role in protein stability, not in catalysis. FEBS Lett., 475, 22-26 (2000) [70] Gellerich, F.N.; Laterveer, F.D.; Korzeniewski, B.; Zierz, S.; Nicolay, K.: Dextran strongly increases the Michaelis constants of oxidative phosphorylation and of mitochondrial creatine kinase in heart mitochondria. Eur. J. Biochem., 254, 172-180 (1998) [71] Lahiri, S.D.; Wang, P.-F.; Babbitt, P.C.; McLeish, M.J.; Kenyon, G.L.; Allen, K.N.: The 2.1 A structure of Torpedo californica creatine kinase complexed with the ADP-Mg2+ -NO-3 -creatine transition-state analogue complex. Biochemistry, 41, 13861-13867 (2002) [72] Eder, M.; Fritz-Wolf, K.; Kabsch, W.; Wallimann, T.; Schlattner, U.: Crystal structure of human ubiquitous mitochondrial creatine kinase. Proteins, 39, 216-225 (2000) [73] Tang, L.; Zhou, H.M.; Lin, Z.J.: Crystallization and preliminary X-ray analysis of human muscle creatine kinase. Acta Crystallogr. Sect. D, 55 (Pt 3), 669-670 (1999) [74] Eder, M.; Schlattner, U.; Becker, A.; Wallimann, T.; Kabsch, W.; Fritz-Wolf, K.: Crystal structure of brain-type creatine kinase at 1.41 A resolution. Protein Sci., 8, 2258-2269 (1999) [75] Rao, J.K.; Bujacz, G.; Wlodawer, A.: Crystal structure of rabbit muscle creatine kinase. FEBS Lett., 439, 133-137 (1998) [76] Zhu, L.; Fan, Y.-X.; Perrett, S.; Zhou, J.-M.: Relationship between Kinetic and Equilibrium Folding Intermediates of Creatine Kinase. Biochem. Biophys. Res. Commun., 285, 857-862 (2001) [77] Moreland, B.; Watts, D.C.: Phosphagen kinases and evolution in the echinodermata. Nature, 214, 458-462 (1967)

384

   



     2.7.3.3     ATP:l-arginine N-phosphotransferase    arginine kinase   

AK ArgK adenosine 5'-triphosphate-arginine phosphotransferase adenosine 5'-triphosphate: l-arginine phosphotransferase arginine phosphokinase kinase, arginine (phosphorylating)     9026-70-4

  











Penaeus japonicus [1] Portunus trituberculatus [1] Paracentrotus lividus [2] Symplectoteuthis oualaniensis [3] Manduca sexta [4, 32] Homarus vulgaris [5, 8, 9] Apis mellifera (three isoforms transcribed from a single gene: a, b, and c [28]) [6, 19, 28] Drosophila melanogaster [7] Drosophila hydei [7] Drosophila simulans [7] Drosophila bifasciata [7] Drosophila subobscura [7] Drosophila ambigua [7] Drosophila tristis [7] Zaprionus vittiger [7] Pecten maximus [10]

385

Arginine kinase

















2.7.3.3

Caudina arenicola [11] Phormia regina [12] Musca domestica [13] Homarus americanus [14] Panulirus longipes [15] Hemicentrotus pulcherrimus [16] Anthocidaris crassispina [16] Pseudocentrotus depressus [16] Jasus verreauxi [17] Sabella pavonina [18, 20] Corbicula japonica [21, 29] Trypanosoma cruzi [22, 35, 37] Trypanosoma brucei [22] Limulus polyphemus [23, 25, 30, 33] Penaeus aztecus [24] Steinernema carpocapsae [26] Plodia interpunctella [27] Solen strictus [29] Stichopus japonicus [31] Nautilus pompilius [34] Octopus vulgaris [34] Sepioteuthis lessoniana [34] Anthopleura japonicus [36]

       !   "   ATP + l-arginine = ADP + N-phospho-l-arginine ( rapid equilibrium random mechanism [6])     phospho group transfer      

 ADP + N-phospho-l-Arg ( the enzyme is an important component of the energy releasing mechanism in the visual system that has high and fluctuating energy demands [28]; the enzyme is a modulator of energetic reserves under starvation stress conditions, activity is post-transcriptionally regulated [37]) (Reversibility: ? [28, 36]) [28, 37] # ATP + l-arginine  ATP + l-Arg ( production of high-energy reserves N-phospho-l-Arg in insect muscles [4]; the enzyme is involved in the storage of the high-energy phosphate reserve phosphoarginine [27]) (Reversibility: ? [3, 4, 27]) [3, 4, 27] # ADP + N-phospho-l-Arg

386

2.7.3.3

Arginine kinase

    

 ATP + 4-guanidinebutanoic acid ( 8% of the activity with lArg [20]) (Reversibility: ? [20]) [20] # ADP + N-phospho-4-guanidinobutanoic acid  ATP + 5-guanidinopentanoic acid ( 10% of the activity with lArg [20]) (Reversibility: ? [20]) [20] # ADP + N-phospho-5-guanidinopentanoic acid  ATP + d-Arg ( d-Arg is as active as l-Arg [20]; d-Arg is phosphorylated to a lesser degree [12]; no activity [9, 13]) (Reversibility: ? [12, 20]) [12, 20] # ADP + N-phospho-d-Arg  ATP + l-Arg ( strictly specific for ATP [20]) (Reversibility: r [1, 3-6, 20, 24, 26]; ? [2, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 21, 22, 23, 25, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37]) [1-37] # ADP + N-phospho-l-Arg [1, 3-6]  ATP + l-arginine methyl ester (Reversibility: ? [6]) [6] # ADP + N-phospho-l-arginine methyl ester  ATP + l-argininic acid ( 45% of the activity with l-Arg [20]) (Reversibility: ? [20]) [20] # ADP + N-phospho-l-argininic acid  ATP + l-canavanine ( 7% of the activity with l-Arg [20]; 7.3% of the activity with l-Arg [26]) (Reversibility: r [4,20,26]) [4, 20, 26] # ADP + l-phosphocanavanine [4]  ATP + l-homoarginine ( 25% of the activity with l-Arg [20]) (Reversibility: ? [20]) [20] # ADP + N-phospho-l-homoarginine  ATP + N-acetyl-l-Arg ( 13% of the activity with l-Arg [20]) (Reversibility: ? [20]) [20] # ADP + Nw -phospho-N-a-acetyl-l-Arg  ATP + octopine ( 30% of the activity with l-Arg [20]) (Reversibility: ? [20]) [20] # ADP + N-phospho-d-octopine  GDP + N-phospho-l-Arg ( 10% of the activity with ADP [3]) (Reversibility: ? [3]) [3] # GTP + l-Arg  UDP + N-phospho-l-Arg ( 10% of the activity with ADP [3]) (Reversibility: ? [3]) [3] # UTP + l-Arg  Additional information ( strictly specific for ATP [20]) [20] # ? $ %  

ATP ( product inhibition, competitive with ADP, noncompetitive with l-Arg [6]) [6]

387

Arginine kinase

2.7.3.3

Ca2+ [3] Cl- [3] Cu2+ [3] d-Arg ( competitive to l-arginine [12]; product inhibition, competitive with arginine phosphate and noncompetitive withg ADP [6]) [6, 12] Fe2+ [3] His ( 5fold higher concentration than l-Arg, 50% inhibition [35]) [35] K+ ( 200 mM, 50% inhibition [3]) [3] l-Asp ( 5fold higher concentration than l-Arg, 25% inhibition [35]) [35] l-Glu ( 5fold higher concentration than l-Arg, 31% inhibition [35]) [35] l-Lys ( 5fold higher concentration than l-Arg, 25% inhibition [35]) [35] l-arginine methyl ester ( competitive to l-Arg [12]) [12] l-canavanine ( competitive to l-Arg [12]) [12] l-homoarginine ( 5fold higher concentration than l-Arg, 33% inhibition [35]) [35] l-nitroarginine ( 5fold higher concentration than l-Arg, 28% inhibition [35]) [35] Mg2+ ( at high concentrations noncompetitive inhibition of MgATP2- [12]) [12] MgADP- ( inhibition is potentiated by NO3 - [11]) [11] MgATP2- ( enzyme form AK2 is strongly inhibited at high concentrations [10]) [10] Mn2+ ( at high concentrations noncompetitive inhibition of MgATP2- [12]) [12] N-methyl-l-Arg ( 5fold higher concentration than l-Arg, 28% inhibition [35]) [35] NADH ( noncompetitive [3]) [3] NH+4 ( 200 mM, 50% inhibition [3]) [3] Na+ ( 200 mM, 50% inhibition [3]) [3] Zn2+ [3] agmatine ( 5fold higher concentration than l-Arg, 20% inhibition [35]) [35] canavanine ( 5fold higher concentration than l-Arg, 50% inhibition [35]) [35] ethylguanidine ( 5fold higher concentration than l-Arg, 22% inhibition [35]) [35] iodoacetamide [14] p-hydroxymercuribenzoate [14] ' ( 

Ca2+ ( 4 mM, activation [1]; 4% of the activation with Mg2+ [3]; activates [6]; 4 mM, 19% of the activation

388

2.7.3.3

Arginine kinase

with Mg2+ [9]; less effactive than Mg2+ in activation [13]; a divalent cation such as Mg2+ , Mn2+ or Ca2+ is required [35]; , activates [36]) [1, 3, 6, 9, 13, 35, 36] Co2+ ( 4 mM, activation [1]; activates [6]; 4 mM, 26% of the activation with Mg2+ [9]) [1, 6, 9] Cu2+ ( activates [6]) [6] Fe2+ ( 4 mM, activation [1]; 4 mM, 12% of the activation with Mg2+ [9]) [1, 9] Mg2+ ( 4 mM, activation [1, 9]; highest activity if ratio Mg2+ :ATP is 1:1, synthesis of arginine phosphate [1, 3, 6]; highest activity if the ratio Mg2+ :ADP is 4:1, synthesis of ATP [1, 6]; Km : 0.6 mM [3]; divalent cation requirement is satisfied by Mg2+ or Mn2+ . Km -value for Mg2+ : 0.6 mM [3]; 5-10 mM required [7]; required [12]; most effective divalent cation for activation [13]; required, Km -value: 0.476 mM, formation of ATP [26]; a divalent cation such as Mg2+ , Mn2+ or Ca2+ is required [35]; activates at concentrations greater than 1 mM, maximal effect at 1.5 mM [36]) [1, 3, 6, 7, 9, 12, 13, 26, 35, 36] Mn2+ ( 4 mM, activation [1, 6]; Km : 0.08 mM [3]; more effective than Mg2+ in activation [9]; less effective than Mg2+ in activation [1, 6, 8, 9, 12, 13]; divalent cation requirement is satisfied by Mg2+ or Mn2+ . Km -value for Mn2+ : 0.08 mM [3]; a divalent cation such as Mg2+ , Mn2+ or Ca2+ is required [35]) [1, 3, 6, 7, 9, 12, 13, 35] Sn2+ ( 4 mM, 13% of the activation with Mg2+ [9]) [9] ) &  * +, 535 (ATP, 25 C, H60G mutant of domain 2 [21]) [21] 535 (l-Arg, 25 C, H60G mutant of domain 2 [21]) [21] 571.8 (ATP, 25 C, D197G mutant of domain 2 [21]) [21] 571.8 (l-Arg, 25 C, D197G mutant of domain 2 [21]) [21] 1086 (ATP, 25 C, H60R mutant of domain 2 [21]) [21] 1086 (l-Arg, 25 C, H60R mutant of domain 2 [21]) [21] 1635 (l-canvanine) [4] 2682 (ATP, 25 C, domain 2 [21]) [21] 2682 (l-Arg, 25 C, domain 2 [21]) [21] 3040 (l-phosphocanavanine) [4] 4398 (ATP, 25 C, two-domain enzyme [21]) [21] 4398 (l-Arg, 25 C, two-domain enzyme [21]) [21] 8320 (l-Arg) [4] 25870 (N-phospho-l-Arg) [4]   !   &  *-., 0.00227 ( formation of arginine phosphate, enzyme from crude extract [26]) [26] 0.00312 ( formation of ATP, enzyme from crude extract [26]) [26] 0.245 ( homogenous muscle enzyme [30]) [30] 0.2539 (, cell extract [22]) [22] 389

Arginine kinase

2.7.3.3

0.288 ( cell extract [22]) [22] 46.3 [24] 154 [7] 212 [14] 234 [1] 240 [23] 248 [4] 318 [1] 360 [2] Additional information [31] /01 *', 0.15 (l-Arg, pH 8.6, 25 C [7]) [7] 0.2 (ADP, pH 7.2, 25 C [3]) [3] 0.26 (l-Arg, 37 C, domain 2 [21]) [21] 0.3 (ATP, 30 C, pH 7.3 [35]) [35] 0.3 (l-Arg, 30 C, pH 7.3 [35]) [35] 0.37 (l-Arg, pH 7.5, 26 C [26]) [26] 0.42 (ADP, pH 7.5, 26 C [26]) [26] 0.42 (l-Arg, 37 C, two-domain enzyme [21]) [21] 0.45 (MgADP-, 37 C [4]) [4] 0.46 (ATP, 37 C, two-domain enzyme [21]) [21] 0.5 (l-Arg, 7 C [4]) [4, 11] 0.52 (l-Arg, 37 C, D197G mutant enzyme of domain 2 [21]) [21] 0.67 (l-Arg, 25 C, mutant enzyme D62G [34]) [34] 0.68 (l-Arg, 25 C, recombinant wild-type enzyme [34]) [34] 0.7 (N-phospho-l-Arg, 37 C [4]) [4] 0.73 (N-phospho-l-Arg, pH 7.5, 26 C [26]) [26] 0.8 (ATP, pH 7.6, 25 C [3]) [3] 0.8 (l-Arg, pH 8.5, 25 C [20]) [20] 0.97 (ATP, 37 C, domain 2 [21]) [21] 1 (l-Arg, pH 7.6, 25 C [3]) [3] 1.02 (l-Arg, 37 C, H60R mutant of domain 2 [21]; 25 C, native enzyme [34]) [21, 34] 1.25 (ADP, pH 6.7, 25 C [2]) [2] 1.3 (d-Arg, pH 8.5, 25 C [20]) [20] 1.3 (MgATP2- ) [11] 1.35 (l-Arg) [13] 1.4 (MgATP2-, pH 8.5, 25 C [20]) [20] 1.74 (l-Arg, 25 C, native enzyme [34]) [34] 2.08 (N-phospho-l-Arg, pH 6.7, 25 C [2]) [2] 2.22 (ATP) [13] 2.35 (ATP, pH 7.5, 26 C [26]) [26] 2.82 (l-Arg, 25 C, native enzyme [34]) [34] 3.45 (l-Arg, 25 C, mutant enzyme S63G [34]) [34] 3.5 (N-phospho-l-Arg, pH 7.2, 25 C [3]) [3] 3.6 (l-Arg, 37 C, H60G mutant of domain 2 [21]) [21]

390

2.7.3.3

Arginine kinase

15 (5-guanidinopentanoic acid, pH 8.5, 25 C [20]) [20] 15 (octopine, pH 8.5, 25 C [20]) [20] 18 (l-arginic acid, pH 8.5, 25 C [20]) [20] 22 (l-canavanine, 37 C [4]) [4] 27 (l-phosphocanavanine, 37 C [4]) [4] 30 (N-acetyl-l-Arg) [20] Additional information [30] / 01 *', 0.05 (Zn2+ ) [3] 0.15 (NADH) [3] 0.5 (Cu2+ ) [3] 0.85 (Ca2+ ) [3] 1.5 (Fe2+ ) [3] 6 (l-canavanine, pH 7.3, 30 C [35]) [35] 7 (l-homoarginine, pH 7.3, 30 C [35]) [35] 100 (SO24- ) [3] 20   5.8 ( synthesis of ATP [1]) [1] 6 ( synthesis of arginine and ATP [24]) [24] 6.1 ( synthesis of ATP [1]) [1] 6.2-6.3 ( synthesis of ATP [20]) [20] 6.3 ( synthesis of ATP, 20 mM phosphate buffer [3]) [3] 6.9 ( synthesis of ATP, 20 mM Tris-HCl buffer [3]) [3] 7.1-7.2 ( synthesis of ATP [6]) [6] 7.2 ( formation of ATP, enzyme from third stage juveniles [26]) [26] 7.3 ( formation of ATP, enzyme from adult [26]) [26] 7.5-8 [10] 7.8 ( formation of arginine phosphate, enzyme from third-stage juveniles [26]) [26] 7.9 [9, 11] 7.9-8.5 ( formation of arginine phosphate, enzyme from adult [26]) [26] 7.9-9.1 [36] 8.2 ( synthesis of N-phospho-l-arginine, 20 mM Tris-HCl buffer or 20 mM phosphate buffer [3]) [3, 35] 8.3 ( synthesis of N-phospho-l-arginine [6]) [6] 8.4 [13] 8.5 ( synthesis of arginine phosphate and ADP [24]) [24] 8.6-8.9 [7] 8.7-8.8 ( synthesis of N-phospho-l-arginine [20]) [20] 9 ( synthesis of N-phospho-l-arginine [1]) [1] 9.2 ( synthesis of N-phospho-l-arginine [1]) [1]

391

Arginine kinase

2.7.3.3

20  5-8.2 ( pH 5.0: about 30% of maximal activity, pH 8.2: about 30% of maximal activity, ATP synthesis [6]) [6] 5.5-7.5 (, pH 5.5: about 90% of maximal activity, pH 7.5: about 60% of maximal activity, synthesis of ATP [20]) [20] 5.5-8 ( pH 5.5: about 90% of maximal activity, pH 8.0: about 70% of maximal activity, no activity at pH 5.0, synthesis of ATP [1]) [1] 5.6-7.5 ( pH 5.3; about 15% of maximal activity, pH 5.6: about 85% of maximal activity, pH 7.5: about 80% of maximal activity, synthesis of arginine and ATP [24]) [24] 5.9-6.8 ( pH 5.9: about 35% of maximal activity, pH 6.8: about 55% of maximal activity, synthesis of ATP, 20 mM phosphate buffer [3]) [3] 6.3-9 ( pH 6.3: about 65% of maximal activity, pH 9.0: about 70% of maximal activity, synthesis of N-phospho-l-Arg [6]) [6] 6.5-7.3 ( pH 6.5: about 45% of maximal activity, pH 9.0: about 45% of maximal activity, synthesis of ATP, 20 mM Tris-HCl buffer [3]) [3] 7-10 ( pH 7.0, about 30% of maximal activity, pH 10.0: about 35% of maximal activity, synthesis of N-phospho-l-arginine [20]) [20] 7.2-9 ( pH 7.2: about 45% of maximal activity, pH 9.0: about 55% of maximal activity, synthesis of arginine phosphate and ADP [24]) [24] 7.5-9 ( about 50% of maximal activity at pH 7.5 and pH 9.0, synthesis of N-phospho-l-arginine, 20 mM Tris-HCl buffer [3]) [3] 8-10.5 ( pH 8.0: about 60% of maximal activity, pH 10.0: about 70% of maximal activity, synthesis of N-phospho-l-Arg [1]) [1] )     * , 40 [1] 42 [1] 45 ( synthesis of ATP and synthesis of N-phospho-l-Arg [6]) [6] )    * , 10-60 ( 10 C: about 50% of maximal activity, 60 C: about 60% of maximal activity [1]; 10 C: about 40% of maximal activity, 60 C: about 40% of maximal activity [1]) [1]

3  "  ' 4 % 36000 ( gel filtration [6]) [6] 37400 [12] 40000 ( sedimentation equilibrium centrifugation [4]; gel filtration [7]; equilibrium sedimentation [14]; gel filtration [24]) [4, 7, 14, 24] 42000 ( sedimentation equilibrium centrifugation [8]) [8, 10] 55000 ( gel filtration [3]) [3] 80000 ( gel filtration [36]) [11, 36]

392

2.7.3.3

Arginine kinase

81000 ( gel filtration [2]) [2] 150000 ( sedimentation equilibrium centrifugation [18,20]) [18, 20]  

? ( x * 37687, calculation from amino acid sequence determined from cyanogen bromide fragments [5]; x * 40000, SDS-PAGE [1]; x * 40500, SDS-PAGE [1,34]; x * 40201, calculation from nucleotide sequence [35]; x * 40238, calculation from nucleotide sequence [25]; x * 40100, calculation from nucleotide sequence [22]; x * 80000, SDS-PAGE [29]) [1, 5, 22, 25, 29, 34, 35] dimer ( 2 * 42000, SDS-PAGE [2]; 2 * 40000 [11]) [2, 11] monomer ( 1 * 37400 [12]; 1 * 40000, SDSPAGE [4,24]; 1 * 42000 [10]; 1 * 55000, SDS-PAGE [3]; 1 * 79933, calculation from nucleotide sequence [36]; 1 * 80000, SDS-PAGE [36]) [3, 4, 10, 12, 24, 36] tetramer ( 4 * 38000-39000, sedimentation equilibrium in presence of 6 M guanidine hydrochloride, gel filtration in 8 M urea [18]) [18]

5 $   .#   .'  .

  

.

 adductor muscle [10, 29] adult [26] antenna [28] body wall muscle [20, 34, 35, 36] central nervous system [28] compound eye [28] egg ( unfertilized [2]) [2] epimastigote [22] epimastigote ( activity increases continuously during the exponential phase of growth [37]) [37] gonad ( male and female [2]) [2] gut [7] intestine [2] lantern muscle [2] larva ( third-stage juvenile [26]) [2, 4, 7, 26] mantle muscle [2] muscle [5, 7, 8, 9, 11, 18, 30, 31] oesophagus [2] posterior midgut [32] procyclic form [22] pupa ( mainly in muscle and gut [7]) [7] tail muscle [14, 24] tegument [2]

393

Arginine kinase

2.7.3.3

tube foot [2] whole body ( mainly in muscle and gut [7]) [7] Additional information ( no activity in sperm [2]) [2] 6 "  mitochondrion ( no microcompartmentation [32]) [32] soluble [22] # !   [1] [1] [2] [3] [4] [9] [19] [7] (enzyme form AK1 and enzyme form AK2 [10]) [10] [11] [12] (partial [13]) [13] [14] [18, 20] (recombinant enzyme from Escherichia coli [23]; three-dimensional crystal structure of an arginine kinase transition-state analogue complex refined at 1.2 A resolution [33]) [23, 33] [24] (recombinant enzyme [27]) [27] (recombinant enzyme [31]) [31] [34] [34] [34]   "  (polyethylene glycol precipitation of recombinant enzyme. Crystallization as a transition state analog [23]) [23]   (domain 2 is separated from the two-domain enzyme and expressed in Escherichia coli, domain 2 still exhibits activity. Expression of mutants of domain 2 in Escherichia coli: H60G, H60R and D197G [21]) [21] (expression in Escherichia coli [35]) [35] (expression in Escherichia coli [22]) [22] (expression in Escherichia coli [23,30]) [23, 25, 30] (expression in Escherichia coli as a histidine-tagged protein [27]) [27] (gene cloned and inserted into the prokaryotic expression plasmid pET21b, expression in a soluble and functional form in Escherichia coli [31]) [31]

394

2.7.3.3

Arginine kinase

    D197G ( mutant of domain 2, affinity for Arg in mutant enzyme is reduced considerably, accompanied by a decrease in Vmax [21]) [21] D62E (, 3.3% of Vmax of recombinant wild-type enzyme, Km -value for l-Arg is 99% of that of the wild-type enzyme [34]) [34] D62G (, 0.6% of Vmax of recombinant wild-type enzyme [34]) [34] H60G ( mutant of domain 2, affinity for Arg in mutant enzyme is reduced considerably, accompanied by a decrease in Vmax [21]) [21] H60R ( mutant of domain 2, affinity for Arg in mutant enzyme is reduced considerably, accompanied by a decrease in Vmax [21]) [21] R193G (,1.5% of Vmax of recombinant wild-type enzyme [34]) [34] S63G (, 5.1% of Vmax of recombinant wild-type enzyme, Km -value for l-Arg is 516% of that of the wild-type enzyme [34]) [34] S63T (, 0.3% of Vmax of recombinant wild-type enzyme [34]) [34] Y68S ( mutant enzyme without activity [34]) [34] Additional information ( double mutant Val268insertion/Phe270deletion: enzyme with significaltly decreased specific activity compared with both the native and the recombinant wild-type enzyme, no detectable change in guanidine substrate specificity [30]) [30] 

   medicine ( the enzyme is a possible target for chemotherapy [22]; the recombinant enzyme may be used to identify a group of polysensitized indoor allergic patients and for immunotheraphy of theses individuals [27]; arginine kinase is a possible chemotherapy target for Chagas` disease [35]) [22, 27, 35]

7    20   5.1 ( 25 C, 15 min, 70% loss of activity [9]) [9] 5.4 ( 25 C, 15 min, 20% loss of activity [9]) [9] 6-9.1 ( 25 C, 15 min, stable [9]) [9] 8.5 ( 30 C, 3 h, stable [9]) [9] )     20 ( 10 min, stable up to [1]) [1] 25 ( 10 min, inactivation above [1]) [1] 30 ( pH 8.5, 3 h, stable [9]) [9] 35 ( 10 min, stable [2]) [2] 40 ( 10 min, 21% loss of activity [2]; 10 min, 100 mM Tris-HCl buffer, pH 8.0, 15% loss of activity. 10 min, 100 mM Tris-HCl buffer, pH 7.0, 16% loss of activity [1]; 10 min, 100 mM Tris-HCl buffer, pH 8.0, 34% loss of activity. 10 min, 100 mM Tris-HCl buffer, pH 7.0, 46% loss of activity [1]) [1, 2] 45 ( 10 min, stable up to [7]; inactivation [2]) [2, 7] 55 ( 10 min, complete loss of activity [7]) [7] Additional information ( unstable to heat [11,12]) [11, 12] 395

Arginine kinase

2.7.3.3

8     !  , stable to repeated freezing/thawing [9] , reversible inactivation by treatment with 8 M urea, reactivation is promoted by thiols and inhibited by divalent metal ions [14] , more stable in 100 mM Tris-HCl buffer at pH 8.0 than in 100 mM phosphate buffer at pH 7.0 [1] , the substrate-bound structure of the two-domain enzyme is stabilized by the bond between H60 and D197 [29]     , -80 C, several months [2] , 4 C, slow loss of activity during prolonged storage, can partially be reactivated by addition of 0.1% w/v 2-mercaptoethanol [4] , 4 C or -10 C, at least 3 months [9] , 4 C, 3 months, 68% loss of activity [13] , refrigerated, 80% saturated ammonium sulfate, 20 mM l-Arg, 100 mM 2-mercaptoethanol, pH 7.0. 50% loss of activity in 8-10 days [20] , -20 C, 10 mM Tris-HCl buffer, pH 8.5, activity gradually decreases, but can be restored by addition of 1 mM 2-mercaptoethanol [1]

! 

[1] Livera, W.C.D.; Shimizu, C.: Comparison and characterization of arginine kinases purified from the prawn Penaeus japonicus (Kurumaebi) and the swimming crab Portunus trituberculatus. Agric. Biol. Chem., 53, 23772386 (1989) [2] Ratto, A.; Christen, R.: Purification and characterization of arginine kinase from sea-urchin eggs. Eur. J. Biochem., 173, 667-674 (1988) [3] Storey, K.B.: Purification and characterization of arginine kinase from the mantle muscle of the squid, Symplectoteuthis oualaniensis. Role of the phosphagen/phosphagen kinase system in a highly aerobic muscle. Arch. Biochem. Biophys., 179, 518-526 (1977) [4] Rosenthal, G.A.; Dahlman, D.L.; Robinson, G.W.: l-Arginine kinase from tobacco hornworm, Manduca sexta (L.). Purification, properties, and interaction with l-canavanine. J. Biol. Chem., 252, 3679-3683 (1977) [5] Regnouf, F.; Kassab, R.; Fattoum, A.: Primary structure of lobster-muscle arginine kinase. Isolation and characterization of the fragments produced by cyanogen-bromide cleavage. Eur. J. Biochem., 44, 67-79 (1974) [6] Cheung, A.C.: Kinetic properties of arginine phosphokinase from honeybees, Apis mellifera L. (Hymenoptera, Apidae). Arch. Biochem. Biophys., 154, 28-39 (1973) [7] Wallimann, T.; Eppenberger, H.M.: Properties of arginine kinase from Drosophila melanogaster. Eur. J. Biochem., 38, 180-184 (1973) [8] Landon, M.F.; Oriol, C.: Hydrodynamic properties of lobster arginine kinase. Biochim. Biophys. Acta, 278, 227-232 (1972)

396

2.7.3.3

Arginine kinase

[9] Virden, R.; Watts, D.C.; Baldwin, E.: Adenosine 5`-triphosphate-arginine phosphotransferase from Lobster muscle: purification and properties. Biochem. J., 94, 536-544 (1965) [10] Reddy, S.R.; Roustan, C.; Benyamin, Y: Purification and properties of two molecular forms of arginine kinase from the adductor muscle of the scallop, Pecten maximus. Comp. Biochem. Physiol. B, 99, 387-394 (1991) [11] Seals, J.D.; Grossman, S.H.: Purification and characterization of arginine kinase from the sea cucumber Caudina arenicola. Comp. Biochem. Physiol. B, 89, 701-707 (1988) [12] Baker, G.T.: Purification and some properties of arginine phosphokinase from the blow fly, Phormia regina. Insect Biochem., 6, 449-456 (1976) [13] Rockstein, M.; Kumar, S.S.: Arginine kinase from the housefly, Musca domestica. Purification and properties. Insect Biochem., 2, 344-352 (1972) [14] Blethen, S.L.; Kaplan, N.O.: Purification of arginine kinase from lobster and a study of some factors affecting its reactivation. Biochemistry, 6, 14131420 (1967) [15] Smith, E.; Morrison, J.F.: Kinetic studies on the arginine kinase reaction. J. Biol. Chem., 244, 4224-4234 (1969) [16] Fujimaki, H.; Yanagisawa, T.: Changes in activities of creatine kinase, arginine kinase and their multienzyme forms during embryonic and larval development of sea urchins. Dev. Growth Differ., 20, 125-131 (1978) [17] Uhr, M.L.; Marcus, F.; Morrison, J.F.: Studies on adenosine triphosphate: arginine phosphotransferase. Purification and reaction mechanism. J. Biol. Chem., 241, 5428-5435 (1966) [18] Robin, Y.; Guillou, A.; Thoai, N.V.: Unspecific arginine kinase of molecular weight 150 000. Amino acid composition, subunit structure and number of substrate binding sites. Eur. J. Biochem., 52, 531-537 (1975) [19] Cheung, A.C.: Kinetic properties of arginine phosphokinase from honeybees, Apis mellifera L. (Hymenoptera, Apidae). Arch. Biochem. Biophys., 154, 28-39 (1973) [20] Robin, Y.; Klotz, C.; van Thoai, N.: Unspecific arginine kinase of molecular weight 150000. Eur. J. Biochem., 21, 170-178 (1971) [21] Suzuki, T.; Tomoyuki, T.; Uda, K.: Kinetic properties and structural characteristics of an unusual two-domain arginine kinase of the clam Corbicula japonica. FEBS Lett., 533, 95-98 (2003) [22] Pereira, C.A.; Alonso, G.D.; Torres, H.N.; Flawia, M.M.: Arginine kinase: a common feature for management of energy reserves in African and American flagellated trypanosomatids. J. Eukaryot. Microbiol., 49, 82-85 (2002) [23] Zhou, G.; Parthasarathy, G.; Somasundaram, T.; Ables, A.; Roy, L.; Strong, S.J.; Ellington, W.R.; Chapman, M.S.: Expression, purification from inclusion bodies, and crystal characterization of a transition state analog complex of arginine kinase: a model for studying phosphagen kinases. Protein Sci., 6, 444-449 (1997) [24] France, R.M.; Sellers, D.S.; Grossman, S.H.: Purification, characterization, and hydrodynamic properties of arginine kinase from gulf shrimp (Penaeus aztecus). Arch. Biochem. Biophys., 345, 73-78 (1997)

397

Arginine kinase

2.7.3.3

[25] Strong, S.J.; Ellington, W.R.: Isolation and sequence analysis of the gene for arginine kinase from the chelicerate arthropod, Limulus polyphemus: insights into catalytically important residues. Biochim. Biophys. Acta, 1246, 197-200 (1995) [26] Platzer, E.G.; Wang, W.; Thompson, S.N.; Borchardt, D.B.: Arginine kinase and phosphoarginine, a functional phosphagen, in the rhabditoid nematode Steinernema carpocapsae. J. Parasitol., 85, 603-607 (1999) [27] Binder, M.; Mahler, V.; Hayek, B.; Sperr, W.R.; Scholler, M.; Prozell, S.; Wiedermann, G.; Valent, P.; Valenta, R.; Duchene, M.: Molecular and immunological characterization of arginine kinase from the Indianmeal moth, Plodia interpunctella, a novel cross-reactive invertebrate pan-allergen. J. Immunol., 167, 5470-5477 (2001) [28] Kucharski, R.; Maleszka, R.: Arginine kinase is highly expressed in the compound eye of the honey bee, Apis mellifera. Gene, 211, 343-349 (1998) [29] Suzuki, T.; Sugimura, N.; Taniguchi, T.; Unemi, Y.; Murata, T.; Hayashida, M.; Yokouchi, K.; Uda, K.; Furukohri, T.: Two-domain arginine kinases from the clams Solen strictus and Corbicula japonica: exceptional amino acid replacement of the functionally important D62 by G. Int. J. Biochem. Cell Biol., 34, 1221-1229 (2002) [30] Strong, S.J.; Ellington, W.R.: Expression of horseshoe crab arginine kinase in Escherichia coli and site-directed mutations of the reactive cysteine peptide. Comp. Biochem. Physiol. B, 113, 809-816 (1996) [31] Guo, S.Y.; Guo, Z.; Guo, Q.; Chen, B.Y.; Wang, X.C.: Expression, purification, and characterization of arginine kinase from the sea cucumber Stichopus japonicus. Protein Expr. Purif., 29, 230-234 (2003) [32] Chamberlin, M.E.: Mitochondrial arginine kinase in the midgut of the tobacco hornworm (Manduca sexta). J. Exp. Biol., 200, 2789-2796 (1997) [33] Yousef, M.S.; Fabiola, F.; Gattis, J.L.; Somasundaram, T.; Chapman, M.S.: Refinement of the arginine kinase transition-state analogue complex at 1.2 A resolution: mechanistic insights. Acta Crystallogr. Sect. D, 58, 2009-2017 (2002) [34] Suzuki, T.; Fukuta, H.; Nagato, H.; Umekawa, M.: Arginine kinase from Nautilus pompilius, a living fossil. Site-directed mutagenesis studies on the role of amino acid residues in the guanidino specificity region. J. Biol. Chem., 275, 23884-23890 (2000) [35] Pereira, C.A.; Alonso, G.D.; Paveto, M.C.; Iribarren, A.; Cabanas, M.L.; Torres, H.N.; Flawia, M.M.: Trypanosoma cruzi arginine kinase characterization and cloning. A novel energetic pathway in protozoan parasites. J. Biol. Chem., 275, 1495-1501 (2000) [36] Suzuki, T.; Kawasaki, Y.; Furukohri, T.: Evolution of phosphagen kinase. Isolation, characterization and cDNA-derived amino acid sequence of two-domain arginine kinase from the sea anemone Anthopleura japonicus. Biochem. J., 328 (Pt 1), 301-306 (1997) [37] Alonso, G.D.; Pereira, C.A.; Remedi, M.S.; Paveto, M.C.; Cochella, L.; Ivaldi, M.S.; Gerez de Burgos, N.M.; Torres, H.N.; Flawia, M.M.: Arginine kinase of the flagellated protozoan Trypanosoma cruzi. Regulation of its expression and catalytic activity. FEBS Lett., 498, 22-25 (2001) 398

)   

3

     2.7.3.4     ATP:taurocyamine N-phosphotransferase    taurocyamine kinase   

ATP:taurocyamine phosphotransferase TPK kinase (phosphorylating), taurocyamine kinase, taurocyamine (phosphorylating) taurocyamine phosphotransferase     9026-72-6

   Arenicola marina [1-3]

       !   "   ATP + taurocyamine = ADP + N-phosphotaurocyamine     phospho group transfer     

 ADP + phosphocreatine ( low activity [3]) (Reversibility: ? [3]) [3] # ATP + creatine  ATP + glycocyamine ( low activity [3]) (Reversibility: r [3]) [3] # ADP + N-phosphoglycocyamine [3]

399

Taurocyamine kinase

2.7.3.4

 ATP + guanidopropionic acid ( low activity [3]) (Reversibility: ? [3]) [3] # ADP + N-phosphoguanidinopropionic acid  ATP + hypotaurocyamine (Reversibility: r [3]) [3] # ADP + N-phosphohypotaurocyamine [3]  ATP + lombricine ( low activity [3]) (Reversibility: ? [1]) [3] # ADP + N-phospholombricine  ATP + taurocyamine (Reversibility: r [1]) [1-3] # ADP + N-phosphotaurocyamine [1, 3] $ %  

NEM [2] PCMB [2] chloroacetophenone [2] monoiodoacetate [2] ' ( 

Mg2+ ( required, maximal activity at 10 mM [1]) [1]   !   &  *-., 1283 [1] /01 *', 0.1 (N-taurocyamine, pH 8.0, 25 C [1]) [1] 0.83 (N-phosphotaurocyamine, pH 7.2, 25 C [1]) [1] 1.2 (ADP, pH 7.2, 25 C [1]) [1] 3.3 (ATP, pH 8.0, 25 C [1]) [1] 20   6.8 ( reaction with phosphotaurocyamine or hypophosphotaurocyamine [3]) [3] 7.2 ( synthesis of ATP [1]) [1] 8 ( synthesis of phosphotaurocyamine [1]) [1] 8.5 ( reaction with hypotaurocyamine [3]) [3] 9 ( reaction with taurocyamine [3]) [3] 20  6.2-8 ( pH 6.2: about 30% of maximal activity, pH 8.0: about 50% of maximal activity, synthesis of ATP [1]) [1] 7.2-8.7 ( pH 7.2: about 40% of maximal activity, pH 8.7: about 40% of maximal activity, synthesis of phosphotaurocyamine [1]) [1] 7.7-9.5 ( pH 7.7: about 50% of maximal activity, pH 9.5: about 70% of maximal activity, synthesis of phosphotaurocyamine [3]; pH 7.7: about 60% of maximal activity, pH 9.5: about 60% of maximal activity, synthesis of hypophosphotaurocyamine [3]) [3] )     * , 33 [1]

400

2.7.3.4

Taurocyamine kinase

)    * , 20-45 ( 20 C: 70% of maximal activity, 45 C: less than 45% of maximal activity [1]) [1]

3  "  ' 4 % 59000 ( gel filtration [1]) [1] 61000 ( ultracentrifugation [1]) [1] 80000 ( gel filtration [2]) [2]  

Additional information ( SDS-PAGE reveals 3 protein bands: 11000 Da, 13000-14000 Da and 21000-22000 Da [1]) [1]

5 $   .#   .'  .

  

.

 body wall muscle [1] muscle [2, 3] 6 "  cytosol [1] mitochondrion [1] # !   [1, 2]

7    20   6-9 ( stable [1]) [1] 8     !  , glycerol does not stabilize [1] , unstable to freezing [1] , unstable to lyophilization [1]     , -20 C, no decrease of activity when the crude extract is kept frozen for several months [1] , 0 C, 0.033 M phosphate buffer, pH 7, or 0.05 M Tris-HCl buffer, pH 7.5, or 0.01 M glycylglycine buffer, pH 7, several weeks stable, or in 75% saturated ammonium sulfate solution, pH 8, stable for several months [2] , 0 C, 0.1 M phosphate buffer, pH 7.2, saturated with mannitol, 0.02% NaN3 , stable for more than 2 months [1]

401

Taurocyamine kinase

2.7.3.4

! 

[1] Surholt, B.: Taurocyamine kinase from body-wall musculature of the lugworm Arenicola marina. Eur. J. Biochem., 93, 279-285 (1979) [2] Kassab, R.; Pradel, L.-A.; van Thoai, N.: ATP:taurocyamine and ATP:lombricine phosphotransferases. Purification and study of SH groups. Biochim. Biophys. Acta, 99, 397-405 (1965) [3] van Thoai, N.; Robin, Y.; Pradel, L.-A.: Hypotaurocyamine phosphokinase comparaison avec la taurocyamine phosphokinase. Biochim. Biophys. Acta, 73, 437-444 (1963)

402

6    

5

     2.7.3.5     ATP:lombricine N-phosphotransferase    lombricine kinase   

LK guanidinethylphosphoserine kinase kinase (phosphorylating), lombricine kinase, lombricine (phosphorylating)     9026-53-3

   Lumbricus terrestris [1, 2, 4] Megascolides cameroni [3] Urechis caupo [5]

       !   "   ATP + lombricine = ADP + N-phospholombricine     phospho group transfer     

 ATP + d-lombricine ( d-isomer, 100%, and l-isomer, 176%, are reactive [4]) (Reversibility: r [3]; ? [4]) [3, 4] # ADP + phospholombricine  ATP + l-lombricine ( d-isomer, 100%, and l-isomer, 176%, are reactive [4]) (Reversibility: r [3]; ? [4]) [3, 4] # ADP + phospholombricine

403

Lombricine kinase

2.7.3.5

 ATP + l-thalassemine (i.e. guanidinoethylphospho-O-(a-N,N-dimethyl)serine, 88% of the activity with d-lombricine [4]) (Reversibility: ? [3]) [4] # ADP + N-l-thalassemine phosphate  ATP + guanidinoethyl phosphate ( 15% of the activity with dlombricine [4]) (Reversibility: ? [4]) [4] # ADP + N-phospho-guanidinoethyl phosphate  ATP + lombricine ( i.e. guanidinoethylphospho-O-serine [3]; d-isomer, 100%, and l-isomer, 176%, are reactive [4]) (Reversibility: r [3]; ? [1,2,4]) [1-5] # ADP + phospholombricine [3]  ATP + taurocyamine ( 44% of the activity with d-lombricine [4]) (Reversibility: ? [4]) [4] # ADP + N-taurocyamine phosphate  dADP + phospholombricine ( at 16% of the activity with ADP [3]) (Reversibility: ? [3]) [3] # dATP + lombricine $ %  

2,4-dinitrofluorobenzene [2] NEM ( 0.3 mM, complete inhibition [3]) [1, 3] PCMB [1] o-iodosobenzoate ( 0.3 mM, complete inhibition [3]) [1, 3] p-hydroxymercuribenzoate ( 0.0001 mM, complete inhibition [3]) [3] phenyl iodoacetate ( 1 mM, complete inhibition [3]) [1, 3] ' ( 

Ca2+ ( 5 mM, slight activation in both directions [3]) [3] Co2+ ( 5 mM, slight activation in both directions [3]) [3] Mg2+ ( 5 mM, activation in both directions [3]; enzyme contains one binding site for MgADP- [2]) [2, 3] Mn2+ ( 5 mM, activation in both directions [3]) [3] Additional information ( not activated by Ni2+ , Cu2+ , Fe2+ , Fe3+ , Al3+ , Sn2+ , Ba2+ , Cd2+ , Zn2+ , Be2+ [3]) [3]   !   &  *-., 12.2 ( 25 C [5]) [5] 31 ( d-lombricine [3]) [3] 49 ( l-lombricine [3]) [3] /01 *', 7.4 (phospholombricine, pH 8.6, 30 C [3]) [3] 9.1 (d-lombricine, pH 8.6, 30 C [3]) [3] 11.7 (l-lombricine, pH 8.6, 30 C [3]) [3] 20   7.2 ( synthesis of ATP [3]) [3] 8.6 ( phosphorylation of lombricine [3]) [3]

404

2.7.3.5

Lombricine kinase

20  6.5-8 ( pH 6.5: about 85% of maximal activity, pH 8.0: about 50% of maximal activity, synthesis of ATP [3]) [3] 7.5-9.5 ( pH 7.5: about 65% of maximal activity, pH 9.5: about 45% of maximal activity, phosphorylation of lombricine [3]) [3]

3  "  ' 4 % 74200 ( gel filtration) [1] 80000 [2]  

? ( x * 40941, calculation from nucleotide sequence [5]) [5] dimer ( two non-identical subunits, Sepharose-mercurial chromatography [2]) [2]

5 $   .#   .'  .

  

.

 muscle [2, 3, 4] # !   [3] (recombinant enzyme [5]) [5]   (expression in Escherichia coli [5]) [5]

7    8     !  , freeze-drying of preparations in non-volatile buffers causes almost 40% loss of activity. Freezing and thawing at pH 8.6 in N-ethylmorpholine-hydrochloric acid buffer produces complete inactivation [3]     , 0 C, 0.033 M phosphate buffer, pH 7, or 0.05 M Tris-HCl buffer, pH 7.5, or 0.01 M glycylglycine buffer, pH 7, several weeks stable, or in 75% saturated ammonium sulfate solution, pH 8 stable for several months [1] , 2 C, pH 7.5, 50% v/v glycerol, several months stable [3]

405

Lombricine kinase

2.7.3.5

! 

[1] Kassab, R.; Pradel, L.-A.; van Thoai, N.: ATP:taurocyamine and ATP:lombricine phosphotransferases. Purification and study of SH groups. Biochim. Biophys. Acta, 99, 397-405 (1965) [2] Der Terrossian, E.; Pradel, L.-A.; Kassab, R.; Desvages, G.: Separation of the two non-identical subunits of lombricine kinase from Lumbricus terrestris muscle by chromatography on sepharose-mercurial. Isolation of the tryptic peptide containing its essential thiol group. Eur. J. Biochem., 45, 243-251 (1974) [3] Gaffney, T.J.; Rosenberg, H.; Ennor, A.H.: The purification and properties of adenosine triphosphate-lombricine phosphotransferase. Biochem. J., 90, 170176 (1964) [4] van Thoai, N.; Robin, Y.; Guillou, Y.: A new phosphagen, N-phosphorylguanidinoethylphospho-O-(a-N,N-dimethyl)serine (phosphothalassemine). Biochemistry, 11, 3890-3895 (1972) [5] Ross Ellington, W.; Bush, J.: Cloning and expression of a lombricine kinase from an echiuroid worm: Insights into structural correlates of substrate specificity. Biochem. Biophys. Res. Commun., 291, 939-944 (2002)

406

2    

7

     2.7.3.6     ATP:hypotaurocyamine N-phosphotransferase    hypotaurocyamine kinase   

kinase, hypotaurocyamine (phosphorylating)     9026-57-7

   Phascolosoma vulgare [1]

       !   "   ATP + hypotaurocyamine = ADP + N-phosphohypotaurocyamine     phospho group transfer     

 ATP + arginine ( very low reaction rate [1]) (Reversibility: r [1]) [1] # ADP + phosphoarginine [1]  ATP + creatine ( very low reaction rate [1]) (Reversibility: r [1]) [1] # ADP + phosphocreatine [1]  ATP + glycocyamine ( very low reaction rate [1]) (Reversibility: r [1]) [1] # ADP + phosphoglycocyamine [1]  ATP + guanidopropionic acid ( very low reaction rate [1]) (Reversibility: r [1]) [1]

407

Hypotaurocyamine kinase

2.7.3.6

ADP + N-phosphoguanidopropionic acid [1] ATP + hypotaurocyamine (Reversibility: r [1]) [1] ADP + phosphohypotaurocyamine [1] ATP + lombricine ( very low reaction rate [1]) (Reversibility: r [1]) [1] # ADP + phospholombricine [1]  ATP + taurocyamine (Reversibility: ? [1]) [1] # ADP + phosphotaurocyamine [1] #  # 

$ %  

chloroacetophenone [1] 20   6.8 ( reaction with hyotaurocyamine or taurocyamine [1]) [1] 8.9 ( phosphorylation of hypotaurocyamine [1]) [1] 20  6.5-7.7 ( pH 6.5: about 75% of maximal activity, pH 7.7: about 70% of maximal activity, reaction with phosphohypotaurocyamine [1]) [1] 7.7-9.5 ( pH 7.7: about 45% of maximal activity, pH 9.5: about 50% of maximal activity, phosphorylation of hypotaurocyamine [1]) [1]

5 $   .#   .'  .

  

.

 muscle [1] # !   [1]

7        , refrigerated, saturated ammonium sulfate solution, pH 8.1, 3-4 weeks stable [1]

! 

[1] van Thoai, N.; Robin, Y.; Pradel, L.-A.: Hypotaurocyamine phosphokinase comparaison avec la taurocyamine phosphokinase. Biochim. Biophys. Acta, 73, 437-444 (1963)

408

 %   



     2.7.3.7     ATP:guanidinoethyl-methyl-phosphate phosphotransferase    opheline kinase   

ATP:guanidinoethylmethylphosphate phosphotransferase kinase (phosphorylating), opheline kinase, opheline (phosphorylating)     37278-15-2

   Ophelia neglecta [1]

       !   "   ATP + guanidinoethylmethyl phosphate = ADP + N'-phosphoguanidinoethylmethyl phosphate     phospho group transfer     

 ATP + 2-guanidinoethyl phosphate (Reversibility: ? [1]) [1] # ADP + N-phospho-2-guanidinoethyl phosphate  ATP + lombricine (Reversibility: ? [1]) [1] # ADP + phospholombricine  ATP + opheline ( i.e. 2-guanidinoethylmethyl phosphate [1]) (Reversibility: r [1]) [1] # ADP + phosphoopheline [1]  ATP + taurocyamine (Reversibility: r [1]) [1] # ADP + phosphotaurocyamine [1] 409

Opheline kinase

2.7.3.7

 Additional information ( no phosphorylation of arginine, guanidinoacetate, no reverse reaction with their corresponding phosphagens [1]) [1] # ? $ %  

NEM [1] PCMB [1] chloroacetophenone [1] monoiodoacetate [1] /01 *', 0.85 (ADP, 30 C [1]) [1] 1.1 (phosphoopheline, 30 C [1]) [1] 1.8 (phosphotaurocyamine, 30 C [1]) [1] 3 (ATP, 30 C [1]) [1] 5.8 (opheline, 30 C [1]) [1] 13 (2-guanidinoethyl phosphate, 30 C [1]) [1] 15 (lombricine, 30 C [1]) [1] 50 (taurocyamine, 30 C [1]) [1] 20   6.8 ( reaction with phosphoopheline [1]) [1] 8.5 ( phosphorylation of opheline [1]) [1] 20  6-7.5 ( oH 6.0: about 80% of maximal activity, pH 7.5: about 65% of maximal activity, reaction with phosphoopheline [1]) [1] 7.5-9 ( pH 7.5: about 65% of maximal activity, pH 9.0: about 50% of maximal activity, phosphorylation of opheline [1]) [1] )     * , 30-35 [1]

5 $   .#   .'  .

  

.

 muscle [1] # !   [1]

! 

[1] van Thoai, N.; di Jeso, F.; Robin, Y.; der Terrossian, E.: Sur la nouvelle acide adenosine 5'-triphosphorique:guanidine phosphotransferase, l`opheline kinase. Biochim. Biophys. Acta, 113, 542-550 (1966)

410

   




     2.7.4.9     ATP:dTMP phosphotransferase    dTMP kinase   

TMK TMP kinase TMPK dTMP kinase dTMPK deoxythymidine 5'-monophosphate kinase kinase, thymidine monophosphate (phosphorylating) kinase, thymidylate (phosphorylating) thymidine 5'-monophosphate kinase thymidine monophosphate kinase thymidylate kinase thymidylate monophosphate kinase thymidylic acid kinase thymidylic kinase     9014-43-1

  





Mus musculus [1, 9] Homo sapiens [2, 3, 6, 13, 19, 21] Saccharomyces cerevisiae [4, 12] Herpes simplex virus type 1 (thymidine/thymidylate multifunctional kinase [5]) [5] Gallus gallus [7] Neurospora crassa [8] Acetabularia mediterranea [10] Escherichia coli (strain B [11]) [11, 14, 15]

555

dTMP Kinase



2.7.4.9

7ersinia pestis [14] Streptococcus pneumonia [16] Mycobacterium tuberculosis [17, 20] Strongylocentrotus intermedius [18]

       !   "   ATP + dTMP = ADP + dTDP ( addition of substrates to the enzyme is random, while release of the products is ordered [5])     phospho group transfer      

 ATP + dTMP ( reaction in both the de novo and salvage pathways of dTTP synthesis [16]) (Reversibility: ? [16]) [16] # ADP + dTDP  Additional information ( key enzyme in nucleotide synthesis [14]) [14] # ?     

 ATP + 2',3'-didehydro-2',3'-dideoxythymidine 5'-monophosphate (Reversibility: ? [13,14]) [13, 14] # ADP + 2',3'-didehydro-2',3'-dideoxythymidine 5'-diphosphate  ATP + 3'-amino-3'-deoxythymidine 5'-monophosphate (Reversibility: ? [13]) [13] # ADP + 3'-amino-3'-deoxythymidine 5'-diphosphate  ATP + 3'-azido-3`-deoxythymidine 5'-monophosphate ( good substrate [14]; Vmax is 100 times lower than with dTMP [14]; no activity [16]) (Reversibility: ? [13,14]) [13, 14] # ADP + 3'-azido-3'-deoxythymidine 5'-diphosphate  ATP + 3'-fluoro-3'-deoxythymidine 5'-monophosphate (Reversibility: ? [13]) [13] # ADP + 3'-fluoro-3'-deoxythymidine 5'-diphosphate  ATP + 5-bromo-2'-deoxyuridine 5'-monophosphate (Reversibility: ? [14,15]) [14, 15] # ADP + 5-bromo-2'-deoxyuridine 5'-diphosphate  ATP + 5-iodo-2'-dUMP ( 60% of the maximal activity with dTMP [11]) (Reversibility: ? [11]) [11] # ADP + 5-iodo-2'-dUDP  ATP + 5-iodo-dUMP ( 34% of the activity with dTMP [4]) (Reversibility: ? [4]) [4] # ADP + 5-iodo-dUDP  ATP + dCMP (Reversibility: ? [18]) [18] # ADP + dCDP

556

2.7.4.9

dTMP Kinase

 ATP + dTMP (Reversibility: r [1]; ? [1, 2, 4, 11, 13, 14, 15, 16, 18, 20]) [1, 2, 4, 11, 13, 14, 15, 16, 18, 20] # ADP + dTDP [1, 2]  ATP + dUMP ( 23% of the activity with dTMP [1]; 31% of the activity with dTMP [4]; at 15% of the activity with dTMP [11]; 15% of the activity with dTMP [16]) (Reversibility: r [1]; ? [4,11,14,15,16]) [1, 4, 11, 14, 15, 16] # ADP + dUDP [1]  CTP + dTMP ( 56% of the activity with ATP [2]; 26% of the activity with ATP [4]; about 30% of the activity with ATP [11]; 20% of the activity with ATP [16]; no activity with CTP [9]) (Reversibility: ? [2,4,11,14,16]) [2, 4, 11, 14, 16] # CDP + dTDP  GTP + dTMP ( 60% of the activity with ATP [2]; 35% of the activity with ATP [4]; 38% of maximal activity [6]; no activity with GTP [9]) (Reversibility: ? [2,4,6,9,14]) [2, 4, 6, 14] # GDP + dTDP  ITP + dTMP ( 1% of the activity with ATP and TMP [14]; 2% of the activity with ATP and TMP [14]) (Reversibility: ? [14]) [14] # IDP + dTDP  UTP + dTMP ( 2.5% of the activity with ATP and TMP [14]; 13% of the activity with ATP and TMP [14]; 60% of the activity with ATP [2]; 36% of the activity with ATP [4]; no activity with UTP [9]) (Reversibility: ? [2,4,14]) [2, 4, 14] # UDP + dTDP  dATP + dTMP ( 70.1% of the activity with ATP [1]; 99% of the activity with ATP [2]; 92% of the activity with ATP [4]; dATP is as active as ATP [6]; about 80% of the activity with ATP [11]; 80% of the activity with ATP [16]) (Reversibility: r [1]; ? [2,4,6,9,11,14,16]) [1, 2, 4, 6, 9, 11, 14, 16] # dADP + dTDP [1, 2]  dCTP + dTMP ( 45% of the activity with ATP [2]; 49% of the activity ATP [4]; about 20% of the activity with ATP [11]; no activity with dCTP [9]) (Reversibility: ? [2,4,11,18]) [2, 4, 11, 18] # dCDP + dTDP  dGTP + dTMP ( 59% of the activity with ATP [2]; 42% of the activity with ATP [4]; 42% of the activity with ATP [6]; about 10% of maximal activity with ATP [11]; no activity with dGTP [9]) (Reversibility: ? [2,4,6,11]) [2, 4, 6, 11] # dGDP + dTDP  dTTP + dTMP ( about 3% of the activity with ATP [11]) (Reversibility: ? [11,18]) [11, 18] # dTDP

557

dTMP Kinase

2.7.4.9

 Additional information ( thymidine/thymidylate multifunctional kinase [5]; direct correlation between the rate of phosphorylation of an NMP and its ability to induce a closing of the enzyme`s phosphate-binding loop [13]) [5, 13] # ? $ %  

(NH4 )2 SO4 ( 0.35 M, 50% inhibition [4]) [4] 2-mercaptoethanol ( 0.4 mM [1]) [1] 3'-azido-3`-deoxythymidine 5'-monophosphate ( potent competitive inhibitor [20]) [20] 5'-aminodeoxythymidine [9] 5'-carboxyldeoxythymidine [9] 5'-chlorodeoxythymidine [9] 5'-iododeoxythymidine [9] 5-iodo-2'-dUMP [11] 5-iodo-dUMP ( 2.5 mM, 30% inhibition [4]; competitive versus both thymidylate and thymidine [5]) [4, 5] 5-methyl iso-dCMP ( competitive [20]) [20] ADP ( 100 nM, 64% inhibition [1]; 0.5 mM, 33% inhibition [2]; 2.5 mM, 24% inhibition [4]; 25% inhibition [7]) [1, 2, 4, 7] ADP ( ADP*Mg2+ product inhibition, competitive versus MgATP2-, noncompetitive versus thymidylate [5]; ADP[b-S] [16]) [5, 6, 9, 16] CH2 ATP ( 100 nM, 23% inhibition [1]) [1] Ca2+ [1] Cu2+ [18] Fe2+ [18] GDP ( 100 nM, 21% inhibition [1]) [1] KCl ( 0.5 M, 10% inhibition [4]) [4] Mn2+ ( slight inhibition [1]) [1] NH4 Cl ( 0.4 M, 75% inhibition [11]) [11] NaCl ( 0.1 M, 25% inhibition [11]) [11] P1 -(adenosine 5')-P4 -(thymidine 5')-tetraphosphate [3] P1 -(adenosine 5')-P5 -(thymidine 5')-pentaphosphate [3] P1 -(adenosine 5')-P6 -(thymidine 5')-hexaphosphate [3] TDP [9] Zn2+ [18] dADP ( 100 nM, 57% inhibition [1]; 0.5 mM, 34% inhibition [2]; 2.5 mM, 23% inhibition [4]) [1, 2, 4, 6] dATP ( 100 nM, 15% inhibition [1]; 0.4 mM, 12% inhibition, adult liver [7]) [1, 7] dCTP [11] dGDP ( 0.5 mM, 23% inhibition [2]) [2] dGTP ( 100 nM, 12% inhibition [1]) [1] dTDP ( 100 nM, 39% inhibition [1]; 0.5 mM, 91% inhibition [2]; 2.5 mM, 75% inhibition [4]; product inhibition, com-

558

2.7.4.9

dTMP Kinase

petitive versus MgATP2- [5]; 0.4 mM, 48% inhibition [7]) [1, 2, 4, 5, 6, 7, 16] dTTP ( 2.5 mM, 17% inhibition [4]; competitive versus both thymidine and thymidylate [5]; 0.4 mM, 43% inhibition [7]; potent feedback inhibitor of dTMP [9]) [4, 5, 6, 7, 9, 10, 11] dUDP ( 0.5 mM, 42% inhibition [2]; 2.5 mM, 15% inhibition [4]) [2, 4] dUMP ( 2.5 mM, 19% inhibition [4]) [4, 11] thymidine ( 2.5 mM, 40% inhibition [4]; 0.77 mM, 27% inhibition [6]) [4, 6, 9]  &    

Additional information ( freezing at -70 C and then thawing results in an increase in activity [10]) [10] ' ( 

Ca2+ ( can partially replace Mg2+ in activation [18]) [18] Cd2+ ( can partially replace Mg2+ in activation [18]) [18] Co2+ ( divalent cation required, Co2 can fully substitute for Mg2+ [4]; 18% of maximal activation with Mg2+ [11]; can partially replace Mg2+ in activation [18]) [4, 11, 18] Fe2+ ( divalent cation required, Fe2+ can fully substitute for Mg2+ [4]) [4] Mg2+ ( required, concentration range for maximal activity is From Mg:ATP ratios of 1 to 3 with an optimum around 1.3 to 1.5 [1]; divalent cation required [4]; requirement of a Mg2+ :ATP ratio greater than 1.0 and for optimal activity there is a requirement of an additional 23 mM Mg2+ above the concentration of ATP [7]; absolute requirement for divalent cation. When Mg2+ is equal to ATP, the rate of dTMP kinase reaction is maximal [11]; activity is maximal in presence of 2-5 mM ATP and 10 mM MgCl2 [18]) [1, 4, 7, 11, 18] Mn2+ ( divalent cation required, Mn2+ can fully substitute for Mg2+ [4]; can partially replace Mg2+ requirement [7]; 41% of maximal activation with Mg2+ [11]; can partially replace Mg2+ in activation [18]) [4, 7, 11, 18] NaCl ( from 0-0.5 M does not affect activity [4]) [4] Ni2+ ( divalent cation required, Ni2+ can fully substitute for Mg2+ [4]) [4] ) &  * +, 0.6 (3'-azido-3'-deoxythymidine 5'-monophosphate, pH 7.5, 25 C [13]) [13] 1.8 (3'-fluoro-3'-deoxythymidine 5'-monophosphate, pH 7.5, 25 C [13]) [13] 1.8 (ddTMP, pH 7.5, 25 C [13]) [13] 5.4 (2',3'-didehydro-2',3'-dideoxythymidine 5'-monophosphate, pH 7.5, 25 C [13]) [13]

559

dTMP Kinase

2.7.4.9

8.4 (3'-amino-3'-deoxythymidine 5'-monophosphate, pH 7.5, 25 C [13]) [13] 24 (TMP, pH 7.5, 25 C [13]) [13] 534 (ATP, pH 8.5, 25 C [16]) [16] 534 (dTMP, pH 8.5, 25 C [16]) [16]   !   &  *-., 0.024 [9] 0.188 [7] 0.809 [6] 0.833 [2] 12.76 [1] Additional information [12] /01 *', 0.0045 (dTMP, pH 7.4, 30 C [20]) [20] 0.0049 (dTMP, pH 7.4, 37 C [2]) [2] 0.005 (TMP, pH 7.5, 25 C [13]) [13] 0.006 (ATP, pH 7.5, 25 C, reaction with dTMP [13]) [13] 0.008 (3'-fluoro-3'-deoxythymidine 5'-monophosphate, pH 7.5, 25 C [13]) [13] 0.012 (2',3'-didehydro-2',3'-dideoxythymidine 5'-monophosphate, pH 7.5, 25 C [13]) [13] 0.012 (3'-azido-3'-deoxythymidine 5'-monophosphate, pH 7.5, 25 C [13]) [13] 0.015 (TMP, pH 7.4, 30 C [14]; pH 7.4, 30 C, wild-type enzyme [15]) [14, 15] 0.027 (ATP, pH 7.5, 25 C, reaction with 3'-fluoro-3'-deoxythymidine 5'-monophosphate [13]) [13] 0.033 (ATP, pH 7.5, 25 C, reaction with 2',3'-didehydro-2',3'-dideoxythymidine 5'-monophosphate [13]) [13] 0.04 (ATP, pH 7.4, 30 C, independent of cosubstrate [15]) [15] 0.045 (TMP, pH 7.4, 30 C [14]) [14] 0.066 (dTMP, pH 8.5, 25 C [16]) [16] 0.069 (ATP, pH 7.5, 25 C, reaction with 3'-azido-3'-deoxythymidine 5'-monophosphate [13]) [13] 0.08 (5-bromo-2'-deoxyuridine 5'-monophosphate, pH 7.4, 30 C [14]; pH 7.4, 30 C, wild-type enzyme [15]) [14, 15] 0.09 (3'-azido-3'-deoxythymidine 5'-monophosphate, pH 7.4, 30 C [14]) [14] 0.1 (5-bromo-2'-deoxyuridine 5'-monophosphate, pH 7.4, 30 C [14]) [14] 0.1 (ATP, pH 7.4, 30 C [20]) [20] 0.13 (dATP, pH 7.2, 37 C [6]) [6] 0.17 (3'-azido-3'-deoxythymidine 5'-monophosphate, pH 7.4, 30 C [14]) [14] 0.17 (3'-azido-3'-deoxythymidine 5'-monophosphate, pH 7.4, 30 C [14]) [14] 560

2.7.4.9

dTMP Kinase

0.19 (dTMP, pH 7.4, 37 C [1]) [1] 0.22 (2',3'-didehydro-2',3'-dideoxythymidine 5'-monophosphate, pH 7.4, 30 C [14]) [14] 0.235 (ATP, pH 8.5, 25 C [16]) [16] 0.24 (dTMP, pH 7.8, 37 C [11]) [11] 0.25 (ATP, pH 7.2, 37 C [6]) [6] 0.25 (dTMP, pH 7.4, 30 C, mutant enzyme G146A [15]) [15] 0.27 (2',3'-didehydro-2',3'-dideoxythymidine 5'-monophosphate, pH 7.4, 30 C [14]) [14] 0.28 (5-bromo-2'-deoxyuridine 5'-monophosphate, pH 7.4, 30 C, mutant enzyme G146A [15]) [15] 0.33 (GTP, pH 7.2, 37 C [6]) [6] 0.45 (dGTP, pH 7.2, 37 C [6]) [6] 0.62 (MgATP2-, pH 7.4, 37 C [2]) [2] 1.1 (MgdATP2- ) [9] 1.2 (ATP, pH 7.8, 37 C [11]) [11] 1.5 (MgATP2-, pH 7.5, 37 C [9]) [9] 2.5 (dUMP, pH 7.4, 30 C [14]; pH 7.4, 30 C, wild-type enzyme [15]) [14, 15] 3.8 (dUMP, pH 7.4, 30 C [14]) [14] 4 (dUMP, pH 7.4, 30 C, mutant enzyme G146A [15]) [15] Additional information [7] / 01 *', 0.00018 (P1 -(adenosine 5')-P6 -(thymidine 5')-hexaphosphate, pH 7.5, 37 C, with dTMP as variable substrate and ATP as cosubstrate [3]) [3] 0.0002 (P1 -(adenosine 5')-P6 -(thymidine 5')-hexaphosphate, pH 7.5, 37 C, with ATP as variable substrate and dTMP as cosubstrate [3]) [3] 0.00047 (P1 -(adenosine 5')-P5 -(thymidine 5')-pentaphosphate, pH 7.5, 37 C, with dTMP as variable substrate and ATP as cosubstrate [3]) [3] 0.0006 (5-iodo-5'-fluoro-2',5'-dideoxyuridine, versus thymidylate [5]) [5] 0.0006 (P1 -(adenosine 5')-P5 -(thymidine 5')-pentaphosphate, pH 7.5, 37 C, with ATP as variable substrate and dTMP as cosubstrate [3]) [3] 0.0009 (5-iodo-5'-fluoro-2',5'-dideoxyuridine, versus thymidine [5]) [5] 0.0175 (P1 -(adenosine 5')-P4 -(thymidine 5')-tetraphosphate, pH 7.5, 37 C, with dTMP as variable substrate and ATP as cosubstrate [3]) [3] 0.02 (3'-azido-3'-deoxythymidine monophosphate, pH 7.4, 30 C [20]) [20] 0.02 (5'-chlorodeoxythymidine, pH 7.5, 37 C [9]) [9] 0.0209 (P1 -(adenosine 5')-P4 -(thymidine 5')-tetraphosphate, pH 7.5, 37 C, with ATP as variable substrate and dTMP as cosubstrate [3]) [3] 0.027 (dTTP, pH 7.5, 37 C [9]) [9] 0.03 (5'-carboxyldeoxythymidine, pH 7.5, 37 C [9]) [9] 0.03 (dTDP, pH 7.5, 37 C [9]) [9] 0.047 (ADP, pH 8.5, 25 C, with ATP as variable substrate [16]) [16]

561

dTMP Kinase

2.7.4.9

0.05 (dTTP, verus thymidylate [5]) [5] 0.059 (dTDP, versus thymidylate [5]) [5] 0.06 (MgADP-, versus MgATP2- [5]) [5] 0.062 (dTTP, versus thymidine [5]) [5] 0.1 (MgADP-, versus thymidylate [5]) [5] 0.13 (5'-aminodeoxythymidine, pH 7.5, 37 C [9]) [9] 0.13 (5-methyl iso-dCMP, pH 7.4, 30 C [20]) [20] 0.16 (dTMP, pH 8.5, 25 C, dTMP as variable substrate [16]) [16] 0.25 (ADP, pH 7.4, 37 C [1]) [1] 0.4 (5-iodo-2'-dUMP, pH 7.8, 37 C [11]) [11] 0.415 (ADP, pH 8.5, 25 C, with dTMP as variable substrate [16]) [16] 0.62 (dTDP, pH 8.0, 37 C [4]) [4] 0.75 (dTTP) [6] 20   6-9 [4] 7 ( and a second optimum at pH 8.8 [10]) [1, 10] 7.5 [7] 7.8 [11] 8-8.5 [18] 8.5 [16] 8.8 ( and a second optimum at pH 7.0 [10]) [10] 20  5-9 ( pH 5: about 45% of maximal activity, pH 6-9: optimum [4]) [4] 7-8.7 ( pH 7.0: about 50% of maximal activity, pH 8.7: about 40% of maximal activity [11]) [11] 7-9.5 ( pH 7.0: about 40% of maximal activity, pH 9.5: about 55% of maximal activity [16]) [16] )     * , 45 [10]

3  "  ' 4 % 33000 ( sucrose density gradient centrifugation [9]) [9] 46000 ( gel filtration [7]) [7] 46600 ( sedimentation equilibrium ultracentrifugation [20]) [20] 48000 ( gel filtration [15]) [15] 50000 ( gel filtration [2]) [2] 65000 ( gel filtration, sucrose density gradient centrifugation [11]) [11] 100000 [18] Additional information ( thymidylate kinase is the product of the CDC8 gene [12]) [12]

562

2.7.4.9

dTMP Kinase

 

? ( x * 24792, calculation from nucleotide sequence [12]; x * 25000, SDS-PAGE [12]) [12] dimer ( 2 * 22635, electrospray ionization mass spectrometry [20]; 2 * 23794, electrospray ionization mass spectrometry, G146A mutant enzyme [15]; 2 * 23779, electrospray ionization mass spectrometry, wild-type enzyme [15]; 2 * 24000, SDS-PAGE [2]) [2, 15, 20]

5 $   .#   .'  .

  

.

 ascites hepatoma [1] ascites sarcoma 180 cell [9] blast cell ( of chronic myelocytic leukemia [6]; peripheral blast cell obtained from acute myelocytic leukemia patients [3]) [3, 6] egg [18] liver ( embryonic and adult [7]) [7] peripheral blood mononuclear cells ( from HIV-infected patients and healthy noninfected individuals. Enzyme activity is 10fold lower in extracts from infected as compared to uninfected persons [19]) [19] placenta ( term placenta [2]) [2] 6 "  chloroplast [10] cytoplasm [4] mitochondrion ( no activity in mitochondria [4]; the enzyme is loosely bound to mitochondrial membrane [8]) [8] nucleus [4] # !   (the stability of the enzyme is maintained during purification by the constant presence of dTMP and 2-mercaptoethanol and by the elimination of substrate-destoying phosphatase activity [1]; partial [9]) [1, 9] [6] [12] (partial [7]) [7] [11] [14] [16]   "  (vapor diffusion method using hanging-drop geometry, crystal structures of the enzyme with dTMP and ADP, dTMP and AppNHp, dTMP with ADP and AlF3, dTDP and ADP, and Tp5 A [21]) [21] [17]

563

dTMP Kinase

2.7.4.9

  (expression of wild-type enzyme and G146A variant in Escherichia coli BL21(DE3)/pDIA17 [15]) [15] (expression in Escherichia coli [14]) [14] (histidine-tagged enzyme, overexpression in Escherichia coli [16]) [16] (overexpression in Escherichia coli [20]) [20]     G146A ( mutation is accompanied by a small but significant enhancement of the thermodynamic stability, midpoint denaturation temperature is 3 C higher than that of the wild-type enzyme, midpoint transition is 3.3 M urea, compared to 3.0 M for the wild-type enzyme [15]) [15] 

   medicine ( the enzyme is a promising target for developing drugs against tuberculosis because the configuration of its active site is unique in the TMPK family [17]) [17]

7    )     41 ( 10 min, inactivation when dTMP concentration is below 0.05 mM [9]) [9] 49 ( Tm -value [14]) [14] 50 ( 10 min, in absence of substrate activity decreases to 32% of the initial activity, completely stable in presence of 10 mM dTMP [4]) [4] 57 ( Tm -value [14]; midpoint denaturation temperature for wild-type enzyme [15]) [14, 15] 60 ( 10 min, in absence of substrate activity decreases to 1% of the initial activity, partial protection in presence of 10 mM dTMP [4]; midpoint denaturation temperature for the G146A mutant enzyme [15]) [4, 15] 65 ( 10 min, 50% inactivation, irreversible [20]) [20] Additional information ( the denaturation temperature of the enzyme increase by 2.9 C in the presence of 5 mM ATP and by 8 C in the presence of 5 mM dTMP [14]) [14] 8     !  , dTDP and dTTP are effective as enzyme stabilizer [1] , the enzyme is very unstable and may be stabilized in the presence of 2mercaptoethanol with dTMP, dTDP, or ADP but not with ATP, MgATP2- or mercaptoethanol alone [9] , the stability of the enzyme is maintained during purification by the constant presence of dTMP and 2-mercaptoethanol and by the elimination of substrate-destoying phosphatase activity [1] , the enzyme is very labile and can be stabilized for long periods of time by its substrate thymidine 5'-monophosphate in the presence of 2-mercaptoethanol [7]

564

2.7.4.9

dTMP Kinase

, midpoint transition is 3.0 M urea for the wild-type enzyme and 3.3 M for the G146A mutant enzyme [15] , dissociation of the native dimeric species of the enzyme occurs at an urea concentration of around 4 M, leading to the accumulation of partially folded monomers that unfold totally at urea concentrations above 5.5 M [20]     , -70 C, 1 week without significant loss of activity [9] , -70 C, 1 year, 20-30% loss of activity [11] , 2 C, half-life of about 1 month [11]

! 

[1] Kielley, R.K.: Purification and properties of thymidine monophosphate kinase from mouse hepatoma. J. Biol. Chem., 245, 4204-4212 (1970) [2] Tamiya, N.; Yusa, T.; Yamaguchi, Y.; Tsukifuji, R.; Kuroiwa, N.; Moriyama, Y.; Fujimura, S.: Co-purification of thymidylate kinase and cytosolic thymidine kinase from human term placenta by affinity chromatography. Biochim. Biophys. Acta, 995, 28-35 (1989) [3] Bone, R.; Cheng, Y.C.; Wolfenden, R.: Inhibition of adenosine and thymidylate kinases by bisubstrate analogs. J. Biol. Chem., 261, 16410-16413 (1986) [4] Jong, A.Y.S.; Campbell, J.L.: Characterization of Saccharomyces cerevisiae thymidylate kinase, the CDC8 gene product. General properties, kinetic analysis, and subcellular localization. J. Biol. Chem., 259, 14394-14398 (1984) [5] Chen, M.C.; Walker, J.; Prusoff, W.H.: Kinetic studies of herpes simplex virus type 1-encoded thymidine and thymidylate kinase, a multifunctional enzyme. J. Biol. Chem., 254, 10747-10753 (1979) [6] Lee, L.S.; Cheng, Y.C.: Human thymidylate kinase. Purification, characterization, and kinetic behavior of the thymidylate kinase derived from chronic myelocytic leukemia. J. Biol. Chem., 252, 5686-5691 (1977) [7] Smith, L.K.; Eakin, R.E.: Partial purification and characterization of thymidylate kinase from embryonic chick liver. Arch. Biochem. Biophys., 167, 6171 (1975) [8] Rossi, M.; Woodward, D.O.: Enzymes of deoxythymidine triphosphate biosynthesis in Neurospora crassa mitochondria. J. Bacteriol., 121, 640-647 (1975) [9] Cheng, Y.C.; Prusoff, W.H.: Mouse ascites sarcoma 180 thymidylate kinase. General properties, kinetic analysis, and inhibition studies. Biochemistry, 12, 2612-2619 (1973) [10] de Groot, E.J.; Schweiger, H.G.: Thymidylate kinase from Acetabularia. I. Properties of the enzyme. J. Cell Sci., 64, 13-25 (1983) [11] Nelson, D.J.; Carter, C.E.: Purification and characterization of thymidine 5monophosphate kinase from Escherichia coli B. J. Biol. Chem., 244, 52545262 (1969)

565

dTMP Kinase

2.7.4.9

[12] Jong, A.Y.S.; Kuo, C.l.; Campbell, J.L.: The CDC8 gene of yeast encodes thymidylate kinase. J. Biol. Chem., 259, 11052-11059 (1984) [13] Ostermann, N.; Segura-Pena, D.; Meier, C.; Veit, T.; Monnerjahn, C.; Konrad, M.; Lavie, A.: Structures of human thymidylate kinase in complex with prodrugs: implications for the structure-based design of novel compounds. Biochemistry, 42, 2568-2577 (2003) [14] Chenal-Francisque, V.; Tourneux, L.; Carniel, E.; Christova, P.; de la Sierra, I.; Barzu, O.; Gilles, A.-M.: The highly similar TMP kinase of Yersinia pestis and Escherichia coli differ markedly in their AZTMP phosphorylating activity. Eur. J. Biochem., 265, 112-119 (1999) [15] Tourneux, L.; Bucurenci, N.; Lascu, I.; sakamoto, H.; Briand, G.; Gilles, A.M.: Substitution of an alanine residue for glycine 146 in TMP kinase from Escherichia coli is responsible for bacterial hypersensitivity to bromodeoxyuridine. J. Bacteriol., 180, 4291-4293 (1998) [16] Petit, C.M.; Koretke, K.K.: Characterization of Streptococcus pneumoniae thymidylate kinase: steady-state kinetics of the forward reaction and isothermal titration calorimetry. Biochem. J., 363, 825-831 (2002) [17] Fioravanti, E.; Haouz, A.; Ursby, T.; Munier-Lehmann, H.; Delarue, M.; Bourgeois, D.: Mycobacterium tuberculosis thymidylate kinase: structural studies of intermediates along the reaction pathway. J. Mol. Biol., 327, 1077-1092 (2003) [18] terentyev, L.L.; Terentyeva, N.A.; Rasskazov, V.A.: Purification and some properties of thymidylate kinase from sea urchin. Biochemistry, 64, 80-85 (1999) [19] Jacobsson, B.; Britton, S.; Törnevik, Y.; Eriksson, S.: Decreas in thymidylate kinase activity in peripheral blood mononuclear cells from HIV-infected individuals. Biochem. Pharmacol., 56, 389-395 (1998) [20] Munier-lehmann, H.; Chaffotte, A.; Pochet, S.; Labesse, G.: Thymidylate kinase of Mycobacterium tuberculosis: a chimera sharing properties common to eukaryotic and bacterial enzymes. Protein Sci., 10, 1195-1205 (2001) [21] Ostermann, N.; Schlichting, I.; Brundiers, R.; Konrad, M.; Reinstein, J.; Veit, T.; Goody, R.S.; Lavie, A.: Insights into the phosphoryltransfer mechanism of human thymidylate kinase gained from crystal structures of enzyme complexes along the reaction coordinate. Structure, 8, 629-642 (2000)

566

 0 % %0   

3?

     2.7.4.10     nucleoside-triphosphate:AMP phosphotransferase    nucleoside-triphosphate-adenylate kinase   

GTP:AMP phosphotransferase guanosine triphosphate-adenylate kinase kinase, nucleoside triphosphate-adenylate (phosphorylating) nucleoside triphosphate-adenosine monophosphate transphosphorylase nucleoside triphosphate-adenylate kinase isozyme 3 of adenylate kinase     9026-74-8

   Bos taurus (calf [7]) [1-4, 7] Sus scrofa [5] Homo sapiens [6]

       !   "   nucleoside triphosphate + AMP = nucleoside diphosphate + ADP ( mechanism [1])     phospho group transfer      

 nucleoside triphosphate + AMP ( involved in reaction sequence of substrate level phosphorylation [2,3]) (Reversibility: r [2, 3]) [2, 3] # nucleoside diphosphate + ADP

567

Nucleoside-triphosphate-adenylate kinase

2.7.4.10

    

 ATP + AMP ( poor substrate, highly specific for AMP, phosphorylation at about 5% the rate of ITP [1]; not [5]) (Reversibility: r [1,7]) [1, 7] # ADP [7]  CTP + AMP ( highly specific for AMP, phosphorylation at about 11% the rate of ITP [1]) (Reversibility: r [1,7]) [1, 7] # CDP + ADP [7]  GTP + AMP ( highly specific for AMP and ADP, in reverse reaction [1]; phosphorylation at about 90% the rate of ITP [1]; dAMP can replace AMP [5]; no acceptor substrates are 2'-AMP, 3'AMP [5]; not: CMP, GMP, IMP, UMP [5,7]) (Reversibility: r [1-3,5,7]) [1-5, 7] # GDP + ADP ( via nucleotide-enzyme complex, no phosphorylated enzyme intermediate [1]) [1, 5]  ITP + AMP ( best substrate [1]; highly specific for AMP and ADP in reverse reaction [1]; dAMP can replace AMP [5]; no donor substrate is ribose 5'-triphosphate [7]; no acceptor substrates are 3'-GMP [1]; not: GMP [7]; not: IMP, CMP, UMP [1,7]; not: dCMP, dGMP, TDP, deoxyadenosine [5]) (Reversibility: r [1,5,7]) [1, 5, 7] # IDP + ADP ( via nucleotide-enzyme complex, no phosphorylated enzyme intermediate [1]) [1, 5]  UTP + AMP ( highly specific for AMP, phosphorylation at about 19% the rate of ITP [1]) (Reversibility: r [1,7]) [1, 7] # UDP + ADP [7]  dGTP + dAMP (Reversibility: r [5]) [5] # dGDP + dADP $ %  

(adenylyl)5-adenosine [4] ADP ( reverse reaction, kinetics [1]) [1] AMP ( free form, Mg2+ reverses [1]) [1] AgNO3 ( weak [5]) [5] GDP ( reverse reaction, kinetics [1]) [1] GTP ( kinetics [1]) [1] HgCl2 ( weak [5]) [5] Mg2+ ( weak, above 4 mM, activates below [1]) [1] N-ethylmaleimide ( weak [5]) [5] p-hydroxymercuribenzoate ( weak [5]) [5] Additional information ( no inhibition by EDTA, GSH, cysteamine or high phosphate concentrations [1]) [1] ' ( 

Ca2+ ( activation, 50% as effective as Mg2+ [1]) [1] Mg2+ ( requirement, 5-10 mM [7]; activation, 4 mM, as good as Mn2+ , slightly inibitory above 4 mM [1]; actual substrate: MgNTP [2-5]) [1-5, 7] 568

2.7.4.10

Nucleoside-triphosphate-adenylate kinase

Mn2+ ( activation, as good as Mg2+ [1]; 75% as effective as Mg2+ [5]) [1, 5] Additional information ( no activation by EDTA, cysteamine, GSH or high phosphate concentrations [1]) [1]   !   &  *-., 0.28 (UTP) [7] 17.5 [5] 135 [1] /01 *', 0.0012 (GDP, pH 8.0, with ADP [1]) [1] 0.033 (AMP) [1] 0.056 (GTP, pH 8.0, with AMP [1]) [1] 0.29 (ADP, pH 8.0, with GDP [1]) [1] 0.63 (ITP, pH 8.0, with AMP [1]) [1] 1 (ATP, pH 8.0, with of AMP [1]) [1] 7.4 (UTP, pH 8.0, with of AMP [1]) [1] 9.1 (CTP, pH 8.0, with of AMP [1]) [1] / 01 *', 0.73 (ATP, pH 8.5, competitive with AMP [1]) [1] 0.74 (ATP, pH 8.5, competitive with GTP [1]) [1] 0.77 (GDP, pH 8.5 [1]) [1] 0.8 (GTP, pH 8.5 [1]) [1] 0.9 (ADP, pH 8.5 [1]) [1] 1.6 (AMP, pH 8.5 [1]) [1] 20   7.4 ( IDP, ADP [5]) [5] 7.5 ( in presence of ITP + AMP [7]) [7] 8.5 [1] 20  6-9 ( about half-maximal activity at pH 6 and about 80% of maximal activity at pH 9 [5]; half-maximal activity at pH 6 and pH 9 [7]; at pH 4.5 the activity was only about 2% of that at pH 7.4 [5]) [5, 7] 7-9.7 ( about half-maximal activity at pH 7 and 9.7 [1]) [1] )     * , 25 ( assay at [5]) [5]

3  "  ' 4 % 52000 ( gel filtration [1]) [1] Additional information ( amino acid sequence [3]) [2-4]

569

Nucleoside-triphosphate-adenylate kinase

5 $   .#   .'  .

2.7.4.10

  

.

 heart [1-4] liver [5, 7] 6 "  mitochondrion ( matrix [1-4]) [1-4] # !   [1] [5]   "  (X-ray diffraction analysis [2,4]) [2, 4]   (isozyme 3 [6]) [6]

7    )     0 ( 5 min, 2 mg protein/ml, stable in 0.1 N HCl [5]) [5] 40 ( 5 min, 2 mg protein/ml, in 0.1 N HCl, 70% loss of activity [5]) [5] 95 ( 5 min, 95% inactivation at pH 7.4, 0.15 M imidazole buffer [5]) [5] Additional information ( 35% ammonium sulfate, AMP or other substrates enhance thermal stability [1]) [1] 8     !  , ammonium sulfate or substrates enhance stability [1] , freeze-thawing, dilutions, or low ionic strength decreases activity rapidly [1] , stability increases during purification [1]     , -15 C, 3 years [7] , 0 C, partially purified preparation, several weeks [1] , room temperature, 1 week [1] , -20 C to 4 C, a few weeks [5]

! 

[1] Albrecht, G.J.: Purification and properties of nucleoside triphosphate-adenosine monophosphate transphosphorylase from beef heart mitochondria. Biochemistry, 9, 2462-2470 (1970)

570

2.7.4.10

Nucleoside-triphosphate-adenylate kinase

[2] Pai, E.F.; Schulz, G.E.; Tomasselli, A.G.; Noda, L.H.: Preliminary X-ray studies on the GTP: AMP phosphotransferase from beef heart mitochondria. J. Mol. Biol., 164, 347-350 (1983) [3] Wieland, B.; Tomasselli, A.G.; Noda, L.H.; Frank, R.; Schulz, G.E.: The amino acid sequence of GTP:AMP phosphotransferase from beef-heart mitochondria. Extensive homology with cytosolic adenylate kinase. Eur. J. Biochem., 143, 331-339 (1984) [4] Diederichs, K.; Schulz, G.E.: Three-dimensional structure of the complex between the mitochondrial matrix adenylate kinase and its substrate AMP. Biochemistry, 29, 8138-8144 (1990) [5] Chiga, M.; Rogers, A.E.; Plaut, G.W.E.: Nucleotide transphosphorylases from liver. J. Biol. Chem., 236, 1800-1805 (1961) [6] Xu, G.; O'Connell, P.; Stevens, J.; White, R.: Characterization of human adenylate kinase 3 (AK3) cDNA and mapping of the AK3 pseudogene to an intron of the NF1 gene. Genomics, 13, 537-542 (1992) [7] Heppel, L.A.; Strominger, J.L.; Maxwell, E.S.: Nucleoside monophosphate kinase, II. Transphosphorylation between adenosine monophophate and nucleoside triphosphates. Biochim. Biophys. Acta, 32, 422-430 (1959)

571

*,   

3

     2.7.4.11     ATP:(d)AMP phosphotransferase    (deoxy)adenylate kinase   

kinase, deoxyadenylate (phosphorylating)     37278-19-6

   Mus musculus [1] Escherichia coli (strain 201 infected with bacteriophage T4amBL292, a maturation defective phage mutant, host-coded activity which is a component of T4 dNTP-synthesizing enzyme complex [2]) [2]

       !   "   ATP + dAMP = ADP + dADP (AMP can also act as acceptor)     phospho group transfer      

 ATP + dAMP (Reversibility: ? [1, 2]) [1, 2] # ADP + dADP [1]     

 ATP + AMP (Reversibility: ? [1]) [1] # ADP + ADP [1]  ATP + dAMP ( no substrates are GMP or dGMP [1]) (Reversibility: ? [1,2]) [1, 2] # ADP + dADP [1]

572

2.7.4.11

(Deoxy)adenylate kinase

$ %  

ATP ( substrate inhibition [1]) [1] EDTA [1] Additional information ( no inhibition by p-hydroxymercuribenzoate [1]) [1] ' ( 

Co2+ ( requirement, half as effective as Mg2+ or Mn2+ [1]) [1] Fe2+ ( requirement, half as effective as Mg2+ or Mn2+ [1]) [1] Fe3+ ( activation, only about 15% as effective as Mg2+ or Mn2+ [1]) [1] Mg2+ ( requirement, as good as Mn2+ [1]) [1] Mn2+ ( requirement, as good as Mg2+ [1]) [1] Additional information ( no activation by Ca2+ , Zn2+ or KCl [1]) [1]   !   &  *-., 0.6 [1] /01 *', 0.1 (ATP) [1] 1 (AMP, plus ATP [1]) [1] 1 (dAMP, plus ATP [1]) [1] 20   7-9 ( broad, Tris-HCl preferred to 3,3-dimethylglutarate buffer [1]) [1] )     * , 37 ( assay at [1]) [1]

5 $   .#   .'  .

  

.

 fibroblast (i.e. l-cells, strain L60TM, a subline of Earle's l-strain [1]) [1] # !   (partial [1]) [1]

7    )     80 ( 94%, 97%, 98% or 100% loss of activity within 2, 5, 20 or 30 min, respectively [1]) [1]

573

(Deoxy)adenylate kinase

2.7.4.11

! 

[1] Griffith, T.J.; Helleiner, C.W.: The partial purification of deoxynucleoside monophosphate kinases from L cells. Biochim. Biophys. Acta, 108, 114-124 (1965) [2] Allen, J.R.; Lasser, G.W.; Goldman, D.A.; Booth, J.W.; Mathews, C.K.: T4 phage deoxyribonucleotide-synthesizing enzyme complex. Further studies on enzyme composition and regulation. J. Biol. Chem., 258, 5746-5753 (1983)

574

) 0$      

3

     2.7.4.12     ATP:(d)NMP phosphotransferase    T2-induced deoxynucleotide kinase   

deoxynucleotide kinase kinase, deoxynucleotide (phosphorylating, T2-induced)     37278-99-2

   Escherichia coli (infected with bacteriophage T2 [1]) [1]

       !   "   ATP + dGMP (or dTMP) = ADP + dGDP (or dTDP)     phospho group transfer      

 ATP + dGMP ( or dTMP [1]) (Reversibility: r [1]) [1] # ADP + dGDP [1]     

 ATP + 5-hydroxymethyl-dCMP ( phosphorylation at about half the rate of dGMP or dTMP [1]) (Reversibility: r [1]) [1] # ADP + 5-hydroxymethyl-dCDP [1]  ATP + dGMP ( as good as dTMP [1]) (Reversibility: r [1]) [1] # ADP + dGDP [1]

575

T2-Induced deoxynucleotide kinase

2.7.4.12

 ATP + dTMP ( as good as dGMP [1]) (Reversibility: r [1]) [1] # ADP + dTDP [1]  dATP + dGMP ( phosphorylation at about 80-90% the rate of ATP [1]) (Reversibility: r [1]) [1] # dADP + dGDP  dATP + dTMP ( phosphorylation at about 80-90% the rate of ATP [1]) (Reversibility: r [1]) [1] # dADP + dTDP  Additional information ( poor substrates are dUMP or 5-methyldCMP, no donor substrates are GTP, CTP, dGTP, dCTP, or dTTP, no acceptor substrates are dCMP, dAMP, GMP, or UMP [1]) [1] # ? $ %  

5-hydroxymethyl-dCMP (dGMP or dTMP as substrate [1]) [1] dGMP ( dTMP or 5-hydroxymethyl-dCMP as substrate [1]) [1] dTMP ( dGMP or 5-hydroxymethyl-dCMP as substrate [1]) [1] Additional information ( tryptic digestion inactivates [1]) [1] ' ( 

Ca2+ ( requirement, about 60% as effective as Mn2+ , dGMP as substrate [1]) [1] Mg2+ ( requirement, about 90% as effective as Mn2+ , dGMP as substrate [1]) [1] Mn2+ ( requirement, dGMP as substrate) [1]   !   &  *-., 24.16 [1] /01 *', 0.056 (5-hydroxymethyl-dCMP, pH 7.4, 37 C [1]) [1] 0.085 (dGMP, pH 7.4, 37 C [1]) [1] 0.278-0.313 (dTMP, pH 7.4, 37 C [1]) [1] 0.82 (ATP, pH 7.4, 37 C, with dTMP [1]) [1] 1.25 (ATP, pH 7.4, 37 C, with dGMP [1]) [1] 5 (ATP, pH 7.4, 37 C, with 5-hydroxymethyl-dCMP [1]) [1] / 01 *', 0.04 (5-hydroxymethyl-dCMP, pH 7.4, 37 C, dTMP is substrate [1]) [1] 0.044 (5-hydroxymethyl-dCMP, pH 7.4, 37 C, dGMP is substrate [1]) [1] 0.064 (dGMP, pH 7.4, 37 C, dTMP is substrate [1]) [1] 0.25 (dTMP, pH 7.4, 37 C, 5-hydroxymethyl-dCMP is substrate [1]) [1] 0.314 (dTMP, pH 7.4, 37 C, dGMP is substrate [1]) [1] 1.28 (dGMP, pH 7.4, 37 C, 5-hydroxymethyl-dCMP is substrate [1]) [1]

576

2.7.4.12

T2-Induced deoxynucleotide kinase

20   8 ( dTMP or dGMP [1]) [1] 8.6 ( 5-hydroxymethyl-dCMP [1]) [1] 20  7-9.3 ( pH 7: about 75% of maximal activity of 5-hydroxymethyldCMP, 84% of maximal activity dTMP or 87% of maximal activity of dGMP, pH 9.3: about 79% of maximal activity of dTMP, 83% of maximal activity of dGMP or 90% of maximal activity of 5-hydroxymethyl-dCMP [1]) [1] )     * , 37 ( assay at [1]) [1]

5 $   .#   .'  .

  

# !   (infected with bacteriophage T2 [1]) [1]

7    20   4 ( 3 min, about 60% loss of activity at 37 C [1]) [1] 4.4 ( 37 C, 3 min, about 30% loss of activity, and about 45% loss of activity within 6 min [1]) [1] 4.7 ( 15 min, about 20% loss of activity at 37 C [1]) [1] 5.4 ( 15 min stable at 37 C [1]) [1]     , -10 C, crude preparation, at least a year [1] , 4 C, partially purified preparation, at least 2 months [1] , at -10 C to 4 C, 1 mM EDTA, up to 20% loss of activity within 6 months [1]

! 

[1] Bello, L.J.; Bessman, M.J.: The enzymology of virus-infected bacteria, IV. Purification and properties of the deoxynucleotide kinase induced by bacteriophage T2. J. Biol. Chem., 238, 1777-1787 (1963)

577

*,  0 % %  

3

     2.7.4.13     ATP:deoxynucleoside-phosphate phosphotransferase    (deoxy)nucleoside-phosphate kinase   

deoxynucleoside monophosphate kinase deoxynucleoside-5'-monophosphate kinase deoxyribonucleoside monophosphokinase kinase (phosphorylating), deoxynucleoside monophosphate     37278-20-9

   no activity in Escherichia coli [1-3] bacteriophage T5 (mutant T5amH128a [3]; from infected Escherichia coli cells [1,3]) [1, 3] bacteriophage T4 (maturation defective phage mutant T4amBL292, phagecoded activity which is a component of T4 dNTP-synthezising enzyme complex [2]; from infected Escherichia coli cells [2]) [2]

       !   "   ATP + deoxynucleoside phosphate = ADP + deoxynucleoside diphosphate     phospho group transfer      

 ATP + deoxynucleoside phosphate ( enzyme induced during viral infection [1-3]) (Reversibility: r [1, 3]; ? [2]) [1-3] # ADP + deoxynucleoside diphosphate [1, 3]

578

2.7.4.13

(Deoxy)nucleoside-phosphate kinase

    

 ATP + 5-bromo-dUMP ( phosphorylated at 45% the rate of dAMP [1]) (Reversibility: r [1]) [1] # ADP + 5-bromo-dUDP  ATP + dAMP ( dAMP is the best acceptor substrate [1,3]; dATP can replace ATP with less efficiency [1]) (Reversibility: r [1,3]; ? [2]) [1, 2] # ADP + dADP [1, 3]  ATP + dCMP ( phosphorylated at 36% the rate of dAMP [3]; phosphorylated at 30% the rate of dAMP, dATP can replace ATP [1]) (Reversibility: r [1,3]) [1, 3] # ADP + dCDP [1]  ATP + dGMP ( phosphorylated at 65% the rate of dAMP [1,3]; dATP can replace ATP [1]) (Reversibility: r [1,3]) [1, 3] # ADP + dGDP [1]  ATP + dTMP ( phosphorylated at 70% the rate of dAMP [1,3]; dATP can replace ATP with less efficiency [1]) (Reversibility: r [1,3]) [1, 3] # ADP + dTDP [1, 3]  Additional information ( very poor acceptor substrates are AMP, CMP or dUMP [1]; no acceptor substrates are dIMP, deoxyxanthylate, 5-hydroxymethyldeoxycytidylate, UMP, GMP, 5-bromo-dUMP, 5-methyldCMP or deoxythymidine [1]; no donor substrates are UTP, GTP, CTP, dTTP, dGTP or dCTP [1]) [1] # ? $ %  

dAMP ( competitive, dTMP as substrate [1]) [1] dCMP ( competitive, dTMP as substrate [1]) [1] dGMP ( competitive, dTMP as substrate [1]) [1] ' ( 

Co2+ ( requirement, can replace Mg2+ [1]) [1] Fe2+ ( activation, less effective than Mg2+ , Co2+, Mn2+ [1]) [1] Mg2+ ( requirement, can replace Co2+ [1]) [1] Mn2+ ( requirement, can replace Co2+ or Mg2+ [1]) [1] Additional information ( no activation by Ba2+ , Ca2+ , Cd2+ , Cr3+, Cu2+ , Fe3+ , Hg2+ , Ni2+ , Zn2+ [1]) [1]   !   &  *-., 14.7 ( purified enzyme [1]) [1] 166.8 ( purified enzyme, substrate dTMP [3]) [3] 232 ( purified enzyme, substrate dAMP [3]) [3] /01 *', 0.034 (dCMP, pH 7.5, 37 C [1]) [1] 0.173 (dTMP, pH 7.5, 37 C [1]) [1] 0.22 (dAMP, pH 7.5, 37 C [1]) [1]

579

(Deoxy)nucleoside-phosphate kinase

2.7.4.13

0.22 (dGMP, pH 7.5, 37 C [1]) [1] 0.36 (ATP, pH 7.5, 37 C [1]) [1] 2.8 (dUMP, pH 7.5, 37 C [1]) [1] / 01 *', 0.032 (dCMP, dTMP as substrate, pH 7.6, 37 C [1]) [1] 0.15 (dAMP, dTMP as substrate, pH 7.6, 37 C [1]) [1] 0.15 (dGMP, dTMP as substrate, pH 7.6, 37 C [1]) [1] 20   7 [1] 7-7.5 [3] 7.6 ( substrate dTMP [1]) [1] 20  6.2-9 ( about 80% of maximal activity at pH 6.2 and about 65% of maximal activity at pH 9 with substrate dGMP [1]) [1] 6.2-9.4 ( about 65% of maximal activity at pH 6.2 and about 70% of maximal activity at pH 9.4 with substrate dAMP [1]) [1] 6.3-9 ( at pH 6.3: about 70% of maximal activity with substrate dTMP, about 85% of maximal activity with substrate dCMP, at pH 9.0: about 50% of maximal activity with substrate dTMP, 65% of maximal activity with substrate dCMP [1]) [1] Additional information ( active over a broad range [3]) [3] )     * , 37 ( assay at [1]) [1]

3  "  ' 4 % 22000 [2] 29140 ( analytical equilibrium ultracentrifugation [3]) [3]  

monomer ( 1 * 29000, SDS-PAGE [3]) [3]

5 $   .#   .'  .

  

# !   (500fold [1]; 1190fold to homogeneity [3]) [1, 3]

580

2.7.4.13

(Deoxy)nucleoside-phosphate kinase

7    )     37 ( 60 min, stable [3]) [3] 42 ( 10% loss of activity after 30 min [3]) [3] 47 ( 50% loss of activity after 30 min [3]) [3] 52 ( rapid inactivation, t1=2 : 5 min [3]) [3]     , -20 C, purified enzyme, 15% glycerol, only loss of a small part of activity after 6 months [3] , 0 C, 25% loss of activity per day in crude cell extract [1] , 4 C, several months, ionic strength 0.1 or above [1]

! 

[1] Bessman, M.J.; Herriott, S.T.; Van Bibber Orr, M.J.: The enzymology of virusinfected bacteria. VI. Purification and properties of the deoxynucleotide kinase induced by bacteriophage T5. J. Biol. Chem., 240, 439-445 (1965) [2] Allen, J.R.; Lasser, G.W.; Goldman, D.A.; Booth, J.W.; Mathews, C.K.: T4 phage deoxyribonucleotide-synthesizing enzyme complex. Further studies on enzyme composition and regulation. J. Biol. Chem., 258, 5746-5753 (1983) [3] Mikoulinskaia, G.V.; Gubanov, S.I.; Zimin, A.A.; Kolesnikov, I.V.; Feofanov, S.A.; Miroshnikov, A.I.: Purification and characterization of the deoxynucleoside monophosphate kinase of bacteriophage T5. Protein Expr. Purif., 27, 195-201 (2003)

581

   

     2.7.4.14     ATP:CMP phosphotransferase    cytidylate kinase   

ATP:UMP-CMP phosphotransferase CMP kinase CMPK CTP:CMP phosphotransferase MssA protein P25 UCK UMP-CMP kinase UMP/CMP kinase UMPK cytidine monophosphate kinase dCMP kinase deoxycytidine monophosphokinase deoxycytidylate kinase kinase, cytidylate (phosphorylating) pyrimidine nucleoside monophosphate kinase Additional information (cf. EC 2.7.4.4 and EC 2.7.4.8)     37278-21-0

  





582

Tetrahymena pyriformis [1, 4, 12] Escherichia coli (strain B [2]) [2, 15, 18, 23, 27] Rattus norvegicus [3, 6, 7, 8, 11, 14] Homo sapiens [5, 10, 20, 24, 25, 26] Mycoplasma mycoides (subsp. mycoides [9]) [9] Saccharomyces cerevisiae [13]

33

2.7.4.14





Cytidylate kinase

Dictyostelium discoideum [16] Arabidopsis thaliana [17] Drosophila melanogaster [19] Sus scrofa [21] Arabidopsis thaliana [22] Bacillus subtilis [23] Dictyostelium sp. [26]

       !   "   ATP + (d)CMP = ADP + (d)CDP ( formation of a ternary complex, addition of substrates is random [5]; reaction proceeds by a sequential mechanism, a ternary complex of the enzyme with both substrates is formed as the central intermediate in the reaction [12]; reaction mechanism is sequential and nonequilibrium in nature, substrates bind to the enzyme in a random order, substrate binding is cooperative [14]; the mechanism is analogous to the phosphoryl transfer mechanism in cAMP-dependent protein kinase that phosphorylates the hydroxyl groups of serine residues [16]; random bi-bi mechanism [17])     phospho group transfer      

 ATP + CMP ( the enzyme is required during rapid cell growth [24]; the enzyme plays a crucial role in the formation of UDP, CDP and dCDP which are required for cellular nucleic acid synthesis [25]; the enzyme catalyses an important step in the phosphorylation of UTP, CTP and dCTP. It is also involved in the necessary phosphorylation by cellular kinases of nucleoside analogs used in antiviral therapies [26]) (Reversibility: ? [24, 25, 26]) [24, 25, 26] # ADP + CDP  ATP + UMP ( the enzyme plays a crucial role in the formation of UDP, CDP and dCDP which are required for cellular nucleic acid synthesis [25]; the enzyme catalyses an important step in the phosphorylation of UTP, CTP and cCTP. It is also involved in the necessary phosphorylation by cellular kinases of nucleoside analogs used in antiviral therapies [26]) (Reversibility: ? [25, 26]) [25, 26] # ADP + UDP  ATP + dCMP ( the enzyme plays a crucial role in the formation of UDP, CDP and dCDP which are required for cellular nucleic acid synthesis [25]; the enzyme catalyses an important step in the phosphorylation of UTP, CTP and cCTP. It is also involved in the necessary phosphorylation by cellular kinases of nucleoside analogs used in antiviral therapies [26]) (Reversibility: ? [25, 26]) [25, 26] # ADP + dCDP 583

Cytidylate kinase

2.7.4.14

    

 ATP + 1-b-d-arabinofuranosylcytosine (Reversibility: ? [20]) [20] # ADP + ?  ATP + 2',2'-difluorodeoxycytidine (Reversibility: ? [20]) [20] # ADP + ?  ATP + AMP (Reversibility: r [5,10,20]) [5, 10, 20] # ADP + ADP  ATP + CMP (Reversibility: r [5]; ? [1, 2, 3, 4, 7, 9, 10, 13, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28]) [1, 2, 3, 4, 5, 7, 9, 10, 13, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27] # ADP + CDP [1, 2, 3, 7]  ATP + l-(-)-2',3'-dideoxy-5-fluoro-3'-thia-CMP (Reversibility: ? [25]) [25] # ADP + ?  ATP + UMP ( 0.8% of the activity with CMP [27]) (Reversibility: r [5]; ? [1, 2, 3, 4, 7, 10, 13, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27]) [1, 2, 3, 4, 5, 7, 10, 13, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27] # ADP + UDP [1, 2, 3, 7]  ATP + ara-CMP (Reversibility: ? [18, 20, 25, 26]) [18, 20, 25, 26] # ADP + ara-CDP  ATP + b-d-2',3'-dideoxy-CMP + H2 O (Reversibility: ? [25]) [25] # ADP + ?  ATP + b-l-2',3'-dideoxy-2',3'-didehydro-5-fluoro-CMP (Reversibility: ? [25]) [25] # ADP + ?  ATP + b-l-2',3'-dideoxy-3'-thiacytidine monophosphate (Reversibility: ? [26]) [26] # ADP + b-l-2',3'-dideoxy-3'-thiacytidine diphosphate  ATP + b-l-2',3'-dideoxy-CMP + H2 O (Reversibility: ? [25]) [25] # ADP + ?  ATP + b-l-dioxolane-cytidine (Reversibility: ? [25]) [25] # ADP + ?  ATP + dAMP (Reversibility: ? [20]) [20] # ADP + dADP  ATP + dCMP ( 105% of the activity with CMP [27]; no evidence for reversal of the reaction [9]) (Reversibility: ir [9]; ? [1, 3, 4, 7, 9, 10, 13, 18, 19, 20, 25, 27]) [1, 3, 4, 7, 9, 13, 18, 19, 20, 25, 27] # ADP + dCDP [1, 3, 7, 9]  ATP + dUMP (Reversibility: ? [10,19,20]) [10, 19, 20] # ADP + dUDP  ATP + gemcitabine monophosphate (Reversibility: ? [25]) [25] # ADP + ? 584

2.7.4.14

 #  # 

#  #  #  #  #  #  #  #  #  #  #  #  #  #  #  #  #  #  # 

Cytidylate kinase

ATP +l-(-)-2',3'-dideoxy-3'-thia-CMP (Reversibility: ? [25]) [25] ADP + ? CTP + CMP (Reversibility: ? [7,25]) [7, 25] CDP + CDP GTP + CMP ( poor substrate [23]; ATP is equally effective as ATP [23]) (Reversibility: ? [7, 18, 23, 25]) [7, 18, 23, 25] GDP + CDP GTP + UMP (Reversibility: ? [25]) [25] GDP + UDP GTP + dCMP (Reversibility: ? [25]) [25] GDP + dCDP ITP + CMP (Reversibility: ? [7]) [7] IDP + CDP ITP + UMP (Reversibility: ? [3,7]) [3, 7] IDP + UDP TTP + CMP (Reversibility: ? [25]) [25] TDP + CDP TTP + UMP (Reversibility: ? [25]) [25] TDP + UDP TTP + dCMP (Reversibility: ? [25]) [25] TDP + dCMP UTP + CMP (Reversibility: ? [7]) [7] UDP + CDP XTP + CMP (Reversibility: ? [7]) [7] XDP + CDP dATP + CMP (Reversibility: ? [3, 7, 23, 25]) [3, 7, 23, 25] dADP + CDP dATP + UMP ( dATP shows 10% of the activity with ATP [4]) (Reversibility: r [5]; ? [3, 4, 7, 25]) [3, 4, 5, 7, 25] dADP + ADP dATP + dCMP (Reversibility: ? [3,7]) [3, 7] dADP + dCDP dCTP + CMP (Reversibility: ? [3,7]) [3, 7] dCDP + CDP dCTP + UMP (Reversibility: ? [25]) [25] dGDP + UDP dCTP + dCMP (Reversibility: ? [25]) [25] dCDP + dCDP dGTP + CMP (Reversibility: ? [7,25]) [7, 25] dGDP + CDP dGTP + UMP (Reversibility: ? [25]) [25] dGDP + UDP dTTP + CMP (Reversibility: ? [7]) [7] dTDP + CDP dUTP + CMP (Reversibility: ? [7]) [7] 585

Cytidylate kinase

2.7.4.14

# dUDP + CDP  Additional information ( formation of a ternary complex, addition of substrates is random [5]; ATP-mediated induced-fit of LID in CMPKcoli modulated by CMP leading to a closed conformation of the active site, protected from water [15]; the UMP-CMP kinase has a relaxed enantiospecificity for the nucleoside monophosphate acceptor site, but it is restricted to d-nucleotides at the donor site [26]) [5, 15, 26] # ? $ %  

ADP [1, 14] CDP ( competitive [1]; competitive withCMP [4]) [1, 4] CMP ( above 0.13 mM, substrate inhibition [3,7]; competitive inhibition of UMP phosphorylation [13]; substrate inhibition above 0.2 mM [26]) [3, 7, 13, 26] CTP ( inhibits reaction with ATP and UMP, CMP or dCMP [25]) [25] CuSO4 ( 0.25 mM, 96% inhibition of enzyme form UMPK1 and 92% inhibition of enzyme form UMPK2 [10]) [10] DTNB ( 0.009 mM, 50% inhibition [11]) [11] F- ( complete inhibition at 25 mM [3]) [3] HgCl2 ( 0.25 mM, complete inhibition of enzyme form UMPK1 and UMPK2 [10]; 0.1 mM, 88% inhibition [13]) [10, 13] MgATP2- ( competitive with UMP [12]) [12] NEM ( 0.035 mM, 50% inhibition [11]; 1.0 mM; 41% inhibition [13]) [11, 13] NaClO ( 250 mM [3]) [3] NaSCN ( 250 mM [3]) [3] P1,P5 -di(adenosine-5')pentaphosphate [17] PCMB ( 0.1 mM, 69% loss of activity [13]) [13] TTP ( inhibits reaction with ATP and UMP, CMP or dCMP [25]) [25] UDP ( product inhibition [14]) [14] UMP ( substrate inhibition above 0.2 mM [26]) [26] UTP ( inhibits reaction with ATP and UMP, CMP or dCMP [25]) [25] ZnCl2 ( 0.25 mM, 55% inhibition of enzyme form UMPK1 and 30% inhibition of enzyme form UMPK2 [10]) [10] dADP [1] dCDP [1] dCMP ( competitive inhibition of UMP phosphorylation [13]) [13] dCTP ( inhibits reaction with ATP and UMP, CMP or dCMP [25]) [25] dTDP [1] iodoacetamide [6] iodoacetate ( 0.05 mM, 50% inhibition [11]) [11] lead nitrate ( 0.25 mM, 52% inhibition of enzyme form UMPK1 and 40% inhibition of enzyme form UMPK2 [10]) [10] p-hydroxymercuribenzoate ( 0.02 mM, 50% inhibition [11]) [11]

586

2.7.4.14

Cytidylate kinase

p-hydroxymercuriphenyl sulfonate ( 0.02 mM, 50% inhibition [11]) [11] Additional information ( no substrate inhibition with 3 mM dCMP and 1 mM UMP [3,7]) [3, 7]  &    

2-mercaptoethanol ( at 5 mM: reduction in molecular weight from approximately 53000 Da to 17000 Da. This low molecular weight form is partially active in the presence of 2-mercaptoethanol. In absence of 2-mercaptoethanol the low molecular weight form is inactive. At 50 mM: full reactivation of the CMP(ATP) kinase activity followed by dCMP(ATP) and CMP(dCTP) [6]; activates [7]) [6, 7] l-Cys ( activates [7]) [7] NADPH (NADPH-dependent activation system is composed of at least two protein factors: one is heat-stable and the other is indistinguishable from NADPH-dependent disulfide reductase [8]) [8] dithiothreitol ( activates [7,13]; 1 mM, activates [13]) [7, 13] glutathione ( activates [7]) [7] reduced dl-a-lipoic acid ( activates [7]) [7] thioredoxin ( activity is strictly dependent upon sulfhydryl reducing agents. Reduced thioredoxin is by far the most effective [3,7]; activates [6]) [3, 6, 7] ' ( 

Ca2+ ( required for the phosphorylation of CMP, IUMP and dCMP by either ATP or dCTP. With CMP as phosphate acceptor and ATP as phosphate donor, Mn2+ , Ni2+ and Ca2+ are able to substitute for Mg2+ but are less effective. The relative rates are Mg2+ (100%), Mn2+ (42%), Ni2+ (16%), and Ca2+ (13%) [3]) [3] Co2+ ( divalent cation required, Mn2+ can substitute for Mg2+ [13]) [13] Mg2+ ( required for the phosphorylation of CMP, IUMP and dCMP by either ATP or dCTP. With CMP as phosphate acceptor and ATP as phosphate donor, Mn2+ , Ni2+ and Ca2+ are able to substitute for Mg2+ but are less effective. The relative rates are Mg2+ (100%), Mn2+ (42%), Ni2+ (16%), and Ca2+ (13%) [3]; optimal concentration is 2-3 mM [5]; required for phosphorylation of CMP, UMP and dCMP by either ATP or dCTP [7]; divalent cation required, Mg2+ is most effective [13]; strong requirement, maximal activity at 3 mM [24]) [3, 5, 7, 13, 24] Mn2+ ( required for the phosphorylation of CMP, IUMP and dCMP by either ATP or dCTP. With CMP as phosphate acceptor and ATP as phosphate donor, Mn2+ , Ni2+ and Ca2+ are able to substitute for Mg2+ but are less effective. The relative rates are Mg2+ (100%), Mn2+ (42%), Ni2+ (16%), and Ca2+ (13%) [3]; with CMP as phosphate donor, Mn2+ , Ni2+ and Ca2+ are able to substitute for Mg2+ , 100%, Mn2+ , 42%, Ni2+ , 15% and Ca2+ , 13% [7]; divalent cation required, Mn2+ can substitute for Mg2+ [13]) [3, 7, 13] Na2 SO4 ( 250 mM, stimulates [3]) [3] 587

Cytidylate kinase

2.7.4.14

NaCl ( 250 mM, stimulates [3]) [3] Ni2+ ( required for the phosphorylation of CMP, IUMP and dCMP by either ATP or dCTP. With CMP as phosphate acceptor and ATP as phosphate donor, Mn2+ , Ni2+ and Ca2+ are able to substitute for Mg2+ but are less effective. The relative rates are Mg2+ (100%), Mn2+ (42%), Ni2+ (16%), and Ca2+ (13%) [3]) [3] ) &  * +, 39 (UMP, pH 7.0, 25 C, AUA chimeric enzyme [21]) [21] 458.4 (UMP, pH 6.5 [17]) [17] 514.2 (CMP, pH 6.5 [17]) [17] 600 (b-l-2',3'-dideoxy-3'-thiacytidine monophosphate, pH 7.4, 37 C [26]) [26] 822 (UMP, pH 7.0, 25 C, UAU chimeric enzyme [21]) [21] 840 (b-l-2',3'-dideoxy-3'-thiacytidine monophosphate, pH 7.4, 37 C, GST fusion UMP/CMP kinase [26]) [26] 1320 (dCMP, pH 7.4, 37 C [26]) [26] 1800 (dCMP, pH 7.4, 37 C, GST fusion UMP-CMP kinase [26]) [26] 2160 (b-l-2',3'-dideoxy-3'-thiacytidine monophosphate, pH 7.4, 37 C, His-tagged UMP-CMP kinase [26]) [26] 2700 (ara-CMP, pH 7.4, 37 C, GST fusion UMP-CMP kinase [26]) [26] 4320 (CMP, pH 7.4, 37 C, GST fusion UMP-CMP kinase [26]) [26] 4380 (dCMP, pH 7.4, 37 C, His-tagged UMP-CMP kinase [26]) [26] 5520 (UMP, pH 7.4, 37 C, GST fusion UMP-CMP kinase [26]) [26] 7800 (CMP, pH 7.4, 37 C, His-tagged UMP-CMP kinase [26]) [26] 7800 (UMP, pH 7.4, 37 C, His-tagged UMP-CMP kinase [26]) [26] 9000 (ara-CMP, pH 7.4, 37 C, His-tagged UMP-CMP kinase [26]) [26] 10440 (UMP, pH 7.0, 25 C, UMP-CMP kinase [21]) [21] 13020 (UMP, pH 7.4, 37 C [26]) [26] 24600 (CMP, pH 7.4, 37 C [26]) [26]   !   &  *-., 4.2 ( reaction with CMP and ATP [3]) [3] 4.98 ( reaction with dCMP and ATP [3]) [3] 8.76 ( reaction with UMP and dCMP [3]) [3] 11.1 [13] 27.5 [1, 4] 30 [3] 30.17 [7] 337 [5] /01 *', 0.0053 (CMP, reaction with ATP, enzyme from Novikoff ascites hepatoma [3]) [3] 0.015 (CMP, 37 C, 196-aa and 228-aa enzyme form, in presence of 8 mM MgATP2- [25]) [25]

588

2.7.4.14

Cytidylate kinase

0.017 (dCMP, pH 8.0, 37 C [19]) [19] 0.02 (UMP, pH 7.4, 37 C, His-tagged UMP/CMP kinase [26]) [26] 0.022 (CMP, pH 7.4, 37 C [26]) [26] 0.023 (CMP, pH 8.0, 37 C [19]) [19] 0.023 (dCMP, 37 C, 196-aa enzyme form, in presence of 8 mM MgATP2- [25]) [25] 0.026 (CMP, 37 C enzyme form UMPK 1 [10]) [10] 0.027 (dCMP, reaction with dATP, liver enzyme [3]) [3, 7] 0.028 (CMP, 37 C, enzyme form UMPK2 [10]) [10] 0.029 (ATP, pH 6.5, when UMP is the other substrate [17]) [17] 0.03 (CMP, reaction with ATP, liver enzyme [3]) [3, 7] 0.035 (CMP, pH 7.4, 30 C [23]; reaction with ATP [18]) [18, 23] 0.038 (ATP, pH 7.4, reaction with CMP [18]) [18] 0.04 (CMP, pH 7.4, 30 C [23]) [23] 0.04 (UMP, reaction with ATP, liver enzyme [3]) [3, 7] 0.043 (UMP, reaction with ATP, enzyme from Novikoff ascites hepatoma [3]) [3] 0.044 (UMP, pH 7.4, 37 C [26]) [26] 0.045 (UMP, pH 7.4, 37 C, His-tagged UMP/CMP kinase [26]) [26] 0.052 (UMP, pH 7.5, 25 C [13]) [13] 0.053 (UMP, reaction with dATP, liver enzyme [3]) [3, 7] 0.053 (UMP, 37 C, enzyme form UMPK1 [10]) [10] 0.064 (UMP, 37 C, enzyme form UMPK2 [10]) [10] 0.067 (ATP, pH 7.1, 37 C [14]) [14] 0.067 (UMP, 37 C, 196-aa enzyme form, in presence of 8 mM MgATP2- [25]) [25] 0.069 (UMP, 37 C, 228-aa enzyme form, in presence of 8 mM MgATP2- [25]) [25] 0.07 (CMP, pH 7.4, 37 C, GST fusion UMP-CMP kinase [26]) [26] 0.071 (CMP, pH 7.5, 25 C [13]) [13] 0.074 (dATP, reaction with CMP, liver enzyme [3]) [3] 0.087 (dATP, pH 7.4, reaction with CMP [18]) [18] 0.094 (dCMP, pH 7.4, 30 C [23]) [23] 0.094 (dGMP, reaction with ATP [18]) [18] 0.095 (UMP, pH 8.0, 37 C [19]) [19] 0.13 (CMP, pH 7.1, 37 C [14]) [14] 0.134 (ATP, reaction with dCMP, enzyme from Novikoff ascites hepatoma [3]) [3] 0.14 (UMP, pH 7.4, 37 C, GST fusion UMP-CMP kinase [26]) [26] 0.15 (b-l-2',3'-dideoxy-3'-thiacytidine monophosphate, pH 7.4, 37 C, His-tagged UMP/CMP kinase [26]) [26] 0.153 (UMP, pH 6.5 [17]) [17] 0.2 (CMP, pH 7.4, 37 C [26]) [26] 0.2 (UMP, pH 7.4, 37 C [26]) [26] 0.204 (ATP, 37 C, 196-aa enzyme form, in presence of 8 mM MgATP2- [25]) [25]

589

Cytidylate kinase

2.7.4.14

0.211 (CMP, 37 C, 196-aa enzyme form, in presence of 8 mM MgATP2- [25]) [25] 0.228 (b-l-(-)-2',3'-dideoxy-2',3'-didehydro-5-fluoro-CMP, 37 C [25]) [25] 0.25 (ATP, 37 C, enzyme form UMPK2 [10]) [10] 0.25 (l-(-)-2',3'-dideoxy-5-fluoro-3'-thia-CMP, 37 C, His-tagged UMP/CMP kinase [25]) [25] 0.26 (ara-CMP, pH 7.4, 37 C, His-tagged UMP/CMP kinase [26]) [26] 0.266 (CMP, pH 6.5 [17]) [17] 0.272 (b-d-2',3'-dideoxy-CMP, 37 C [25]) [25] 0.292 (ATP, pH 6.5, reaction with CMP [17]) [17] 0.3 (b-l-2',3'-dideoxy-3'-thiacytidine monophosphate, pH 7.4, 37 C, GST fusion UMP-CMP kinase [26]) [26] 0.32 (ATP, reaction with CMP, liver enzyme [3]) [3] 0.33 (dCMP, pH 7.4, 30 C [23]) [23] 0.34 (ara-CMP, pH 7.4, 37 C, GST fusion UMP-CMP kinase [26]) [26] 0.35 (ATP, pH 7.1, 37 C [14]) [14] 0.354 (dCMP, 37 C, 196-aa enzyme form, in presence of 8 mM MgATP2- [25]) [25] 0.36 (ATP, 37 C, enzyme form UMPK1 [10]) [10] 0.36 (araCMP, pH 7.4, 30 C [23]; reaction with ATP [18]) [18, 23] 0.37 (araCMP, pH 7.4, 30 C [23]) [23] 0.42 (dATP, reaction with UMP, liver enzyme [3]) [3] 0.43 (dUMP, pH 8.0, 37 C [19]) [19] 0.45 (2',2'-difluorodeoxycytidine, pH 8.0, 37 C [20]) [20] 0.494 (l-(-)-2',3'-dideoxy-3'-thia-CMP, 37 C [25]) [25] 0.5 (ATP) [4] 0.5 (CMP, pH 8.0, 37 C [20]) [20] 0.513 (dCMP, 37 C, 196-aa enzyme form, in presence of 8 mM MgATP2- [25]) [25] 0.54 (dCMP, pH 7.5, 25 C [13]) [13] 0.58 (ATP, reaction with UMP, liver enzyme [3]) [3] 0.581 (gemcitabine monophosphate, 37 C [25]) [25] 0.61 (dATP, reaction with dCMP, liver enzyme [3]) [3] 0.64 (GTP, reaction with CMP [18]) [18] 0.68 (ATP, reaction with dCMP, liver enzyme [3]) [3] 0.697 (b-l-2',3'-dideoxy-CMP, 37 C [25]) [25] 0.715 (dCMP, reaction with ATP, enzyme from Novikoff ascites hepatoma [3]) [3] 0.8 (CMP) [4] 0.82 (dCTP, reaction with CMP, liver enzyme [3]) [3] 0.9 (dCMP, pH 7.4, 37 C, His-tagged UMP/CMP kinase [26]) [26] 0.917 (ara-CMP, 37 C [25]) [25] 0.93 (UMP, pH 7.4, 30 C [23]; reaction with ATP [18]) [18, 23] 590

2.7.4.14

Cytidylate kinase

0.98 (CMP, reaction with dCTP, liver enzyme [3]) [3, 7] 1.037 (b-d-2',3'-dideoxy-CMP, 37 C [25]) [25] 1.1 (dCMP, reaction with dATP, liver enzyme [3]) [3] 1.1 (dCMP, pH 7.4, 37 C, GST fusion UMP-CMP kinase [26]) [26] 1.25 (CMP) [4] 1.4 (UMP, pH 7.1, 37 C [14]) [14] 1.4 (araCMP, pH 8.0, 37 C [20]) [20] 1.6 (UMP, pH 8.0, 37 C [20]) [20] 2 (b-l-2',3'-dideoxy-3'-thiacytidine monophosphate, pH 7.4, 37 C [26]) [26] 2 (cCMP, pH 7.4, 37 C [26]) [26] 2.77 (dCMP, reaction with ATP, liver enzyme [3]) [3, 7] 3.6 (UMP, pH 7.4, 30 C [23]) [23] 5.9 (dUMP, pH 8.0, 37 C [20]) [20] 8.5 (dUMP, pH 7.5, 25 C [13]) [13] / 01 *', 0.0012 (P1,P5 -di(adeenosine-5')pentaphosphate, versus ATP [17]) [17] 0.00653 (P1,P5 -di(adeenosine-5')pentaphosphate, versus UMP [17]) [17] 0.12 (CMP, pH 7.4, 37 C, GST fusion UMP/CMP kinase [26]) [26] 0.5 (CMP, pH 7.4, 37 C, His-tagged UMP/CMP kinase [26]) [26] 0.5 (UMP, pH 7.4, 37 C, GST fusion UMP/CMP kinase [26]) [26] 1.5 (UMP, pH 7.4, 37 C, His-tagged UMP/CMP kinase [26]) [26] 20   6.5 [17] 7-8 [1, 4] 7.5 ( reaction with UMP and ATP [13]) [13] 20  4-9 ( pH 4.0: about 40% of maximal activity, pH 9.0: about 65% of maximal activity, reaction with UMP and ATP [13]) [13] 6.2-8.6 ( 50% of maximal activity at pH 6.2 and at pH 8.6 [1]) [1]

3  "  ' 4 % 22500 ( sucrose density gradient centrifugation [11]) [11] 26000 ( gel filtration [11,13]) [11, 13]  

? ( x * 22222, calculation from nucleotide sequence [24]; x * 22448, mass spectroscopy, calculation from nucleotide sequence [17]) [17, 24]

591

Cytidylate kinase

5 $   .#   .'  .

2.7.4.14

  

.

 A-549 cell ( lung carcinoma [24]) [24] Burkitt lymphoma cell [24] G-361 cell ( melanoma cell [24]) [24] H-660 cell ( promyelocytic leukemia [24]) [24] HeLa cell 53 [24, 25] K-562 cell ( chronic myelogenous leukemia cell [24]) [24] MOLT-4 cell ( lymphoblastic leukemia cell [24]) [24] Novikoff ascites hepatoma cell [3] Raji cell [24] SW-480 cell ( colorectal adenocarcinoma cell [24]) [24] appendix [24] bone marrow [11, 14, 24] brain [21] erythrocyte [5, 10] fetal liver [24] liver [3, 6, 7, 8] lymph node [24] macrophage [24] peripheral blood leukocyte [24] spleen [24] thymus [24] Additional information ( two mRNA species are expressed in all immune tissues examined, an all cases the 3.4 kb form is the more prominent RNA species [24]) [24] 6 "  cytoplasm [25] cytosol [20] mitochondrion ( N-terminal signal targets the enzyme to mitochondria [19]) [19] nucleus [20] # !   [1, 4, 11] (partial [2]) [2, 18] [3, 7] (partial, two allelic gene products UMPK1 and UMPK2 [10]) [5, 10, 25] [13] [17] [23]   "  (hanging drop vapor diffusion technique [18]; enzyme in complex with CDP [27]) [18, 27]

592

2.7.4.14

Cytidylate kinase

  (hanging-drop method in 50 mM Tris-HCl buffer, pH 7.4, 20 C, with ammonium sulfate as a precipitant [27]) [27] (expression in Escherichia coli. The 228-aa and the 196-11 form are expressed as His-tagged fusion protein. The 196-aa UMP/CMP kinase is the actual form of the enzyme [20,25]; expression of His-tagged UMP-CMP kinase and UMP-CMP kinase fusion protein with glutathione S-transferase [26]) [20, 25, 26] (cDNA is subcloned into pGEX-4T-3 and expressed as a glutathione Stransferase fusion protein in Escherichia coli [17]) [17, 24] (expression in Escherichia coli [19]) [19]     G21A ( mutant enzyme is degraded during the purification phase [22]) [22] G22A ( mutant enzyme with decreased turnover-number/Km ATP value. Turnover-number is 59% of that of the wild-type enzyme [22]) [22] G24A ( mutant enzyme with decreased turnover-number/Km ATP value. Turnover-number is 48% of that of the wild-type enzyme [22]) [22] G26A ( mutant enzyme with decreased turnover-number/Km ATP value [22]) [22] G27R ( mutant enzyme is degraded during the purification phase. Turnover-number is 45% of that of the wild-type enzyme [22]) [22] K27E ( mutant enzyme with 2600fold decreased turnover-number/Km ATP value. Turnover-number is 21% of that of the wild-type enzyme [22]) [22] K27M ( mutant enzyme with 1000fold decreased turnover-number/Km ATP value. Turnover-number is 22% of that of the wild-type enzyme [22]) [22] Additional information ( two types of chimeric enzymes have been constructed by genetic engineering of chicken cytosolic adenylate kinase and porcine brain UMP/CMP kinase. One designated as UAU carries an AMP-binding domain of AK in the remaining body of UMP/CMP kinase, and the other, designated as AUA, carries a UMP/CMP-binding domain in the remaining body of adenylate kinase. UAU is 4fold more active with AMP, 40fold less active with UMP, and 4fold less active with CMP than the parental UMP/CMP kinase, although AUA has considerably lowered reactivity for both AMP and UMP. AUA has a Tm -value 11 C lower than adenylate kinase, whereas UAU has a Tm -value similar to that of UMP/CMP kinase. Expression in Escherichia coli JM109 [21]) [21]

593

Cytidylate kinase

2.7.4.14

7    20   6.5 ( more stable than at higher pH [5]) [5] 7 ( at pH 7.0, the enzyme is most stable when kept at 4 C [13]) [13] Additional information ( more stable in histidine buffer than in phosphate buffer [5]) [5] )     4 ( 24 h, in absence of dithiothreitol the purified enzyme shows considerable loss of activity [11]; at pH 7 the enzyme is most stable when kept at 4 C [13]) [11, 13] 20 ( in absence of glycerol, the half-life of a preparation with a specific activity of 80 is about 10 min, inactivation is partially reversed by addition of 2-mercaptoethanol [5]) [5] 25 ( 30 min, stable [10]) [10] 40 ( 30 min, enzyme form UMPK2 loses 70% of maximal activity, enzyme form UMPK1 loses 30% of its activity [10]) [10] 45 ( 30 min, enzyme form UMPK2 loses more than 80% of maximal activity, enzyme form UMPK1 loses more than 60% of its activity [10]) [10] 48 ( midpoint denaturation temperature in absence of nucleotide substrates or in presence of ATP [23]) [23] 49 ( midpoint denaturation temperature in absence of nucleotide substrates [23]; midpoint denaturation temperature in presence of CMP [23]) [23] 50 ( 30 min, enzyme forms UMPK2 and UMPK1 lose 90% of its activity [10]; midpoint denaturation temperature of the AUA chimeric enzyme [21]) [10, 21] 51 ( midpoint denaturation temperature of UMP/CMP kinase [21]; midpoint denaturation temperature in presence of CMP [23]) [21, 23] 52 ( midpoint denaturation temperature of the UAU chimeric enzyme [21]) [21] 58 ( 10 min, 50% loss of activity [17]; midpoint denaturation temperature in presence of ATP [23]) [17, 23] 8     !  , bovine serum albumin, 0.1 mg/ml, is completely effective in preventing the loss of activity in the dilute preparation [1] , three-fold dilution of a preparation of 0.2 mg of protein per ml results in less of 50% of the activity in 1 h [1, 4] , dialysis against 20 mM phosphate, 1 mM MgCl2 , 20% ethylene glycol, pH 8.0, 90% loss of activity [11] , freeze-thawing inactivates [11] , the enzyme is unstable when fully activated, anions promoting hydrophobic interactions stabilize the active conformation [6] , more stable in histidine than in phosphate buffer [5] , the purified enzyme is notably unstable [5] 594

2.7.4.14

Cytidylate kinase

    , 4 C, 0.2 mg/ml, 2 weeks, 20% loss of activity [1] , -10 C, 30% loss of activity within 2 months [2] , -20 C, in 25 mM Tris-acetate buffer, pH 7.5, 50 mM 2-mercaptoethanol, 50% glycerol, stable for at least 2 months [3] , -80 C, up to 12 months [11] , 4 C, considerable loss of activity within 24 h, DTT stabilizes, more stable in 20 mM phosphate buffer, pH 8 than in Tris-HCl buffer [11] , -20 C, concentrated enzyme solution in 30% glycerol, less than 5% loss of activity per month [5]

! 

[1] Ruffner, B.W.; Anderson, E.P.: Adenosine triphosphate: uridine monophosphate-cytidine monophosphate phosphotransferase from Tetrahymena pyriformis. J. Biol. Chem., 244, 5994-6002 (1969) [2] Hurwitz, J.: The enzymatic incorporation of ribonucleotides into polydeoxynucleotide material. J. Biol. Chem., 234, 2351-2358 (1959) [3] Orengo, A.; Maness, P.: Pyrimidine nucleoside monophosphate kinase from rat liver and rat Novikoff ascites hepatoma (EC 2.7.4.14). Methods Enzymol., 51, 321-331 (1978) [4] Anderson, E.P.: UMP-CMP kinase from Tetrahymena pyriformis. Methods Enzymol., 51, 331-337 (1978) [5] Scott, E.M.; Wright, R.C.: Kinetics and equilibria of pyrimidine nucleoside monophosphate kinase from human erythrocytes. Biochim. Biophys. Acta, 571, 45-54 (1979) [6] Maness, P.; Orengo, A.: Activation of rat liver pyrimidine nucleoside monophosphate kinase. Biochim. Biophys. Acta, 429, 182-190 (1976) [7] Maness, P.; Orengo, A.: A pyrimidine nucleoside monophosphate kinase from rat liver. Biochemistry, 14, 1484-1489 (1975) [8] Kobayashi, S.; Kanayama, K.: NADPH activation of deoxycytidylate kinase in rat liver extract: involvement of an endogenous disulfide reductase system. Biochem. Biophys. Res. Commun., 74, 1249-1255 (1977) [9] Neale, G.A.M.; Mitchell, A.; Finch, L.R.: Enzymes of pyrimidine deoxyribonucleotide metabolism in Mycoplasma mycoides subsp. mycoides. J. Bacteriol., 156, 1001-1005 (1983) [10] Teng, Y.-S.; Chen, S.-H.; Scott, C.R.: Human erythrocyte pyrimidine nucleoside monophosphate kinase. Partial purification and properties of two allelic gene products. J. Biol. Chem., 251, 4179-4183 (1976) [11] Seagrave, J.; Reyes, P.: Pyrimidine nucleoside monophosphate kinase from rat bone marrow cells: chromatographic, electrophoretic, and sedimentation behavior of active and inactive enzyme forms. Arch. Biochem. Biophys., 247, 76-83 (1986) [12] Gravey, T.Q.; Millar, F.K.; Anderson, E.P.: ATP:UMP-CMP phosphotransferase from Tetrahymena pyriformis. II. Kinetic studies and reaction mechanism with UMP. Biochim. Biophys. Acta, 302, 38-49 (1973) 595

Cytidylate kinase

2.7.4.14

[13] Kohno, H.; Kumagai, H.; Tochikura, T.: Purification and properties of pyrimidine nucleoside monophosphate kinase from baker`s yeast. Agric. Biol. Chem., 47, 19-24 (1983) [14] Seagrave, J.; Reyes, P.: Pyrimidine nucleoside monophosphate kinase from rat bone marrow cells: a kinetic analysis of the reaction mechanism. Arch. Biochem. Biophys., 254, 518-525 (1987) [15] Li de La Sierra, I.M.; Gallay, J.; Vincent, M.; Bertrand, T.; Briozzo, P.; Barzu, O.; Gilles, A.M.: Substrate-induced fit of the ATP binding site of cytidine monophosphate kinase from Escherichia coli: time-resolved fluorescence of 3'-anthraniloyl-2'-deoxy-ADP and molecular modeling. Biochemistry, 39, 15870-15878 (2000) [16] Hutter, M.C.; Helms, V.: Phosphoryl transfer by a concerted reaction mechanism in UMP/CMP-kinase. Protein Sci., 9, 2225-2231 (2000) [17] Zhou, L.; Lacroute, F.; Thornburg, R.: Cloning, expression in Escherichia coli, and characterization of Arabidopsis thaliana UMP/CMP kinase. Plant Physiol., 117, 245-254 (1998) [18] Bucurenci, N.; Sakamoto, H.; Briozzo, P.; Palibroda, N.; Serina, L.; Sarfati, R.S.; Labesse, G.; Briand, G.; Danchin, A.; Barzu, O.; Gilles, A.M.: CMP kinase from Escherichia coli is structurally related to other nucleoside monophosphate kinases. J. Biol. Chem., 271, 2856-2862 (1996) [19] Curbo, S.; Amiri, M.; Foroogh, F.; Johansson, M.; Karlsson, A.: The Drosophila melanogaster UMP-CMP kinase cDNA encodes an N-terminal mitochondrial import signal. Biochem. Biophys. Res. Commun., 311, 440-445 (2003) [20] Van Rompay, A.R.; Johansson, M.; Karlsson, A.: Phosphorylation of deoxycytidine analog monophosphates by UMP-CMP kinase: molecular characterization of the human enzyme. Mol. Pharmacol., 56, 562-569 (1999) [21] Okajima, T.; Fukamizo, T.; Goto, S.; Fukui, T.; Tanizawa, K.: Exchange of nucleoside monophosphate-binding domains in adenylate kinase and UMP/CMP kinase. J. Biochem., 124, 359-367 (1998) [22] Zhou, L.; Thornburg, R.: Site-specific mutations of conserved residues in the phosphate-binding loop of the Arabidopsis UMP/CMP kinase alter ATP and UMP binding. Arch. Biochem. Biophys., 358, 297-302 (1998) [23] Schultz, C.P.; Ylisastigui-Pons, L.; Serina, L.; Sakamoto, H.; Mantsch, H.H.; Neuhard, J.; Barzu, O.; Gilles, A.M.: Structural and catalytic properties of CMP kinase from Bacillus subtilis: a comparative analysis with the homologous enzyme from Escherichia coli. Arch. Biochem. Biophys., 340, 144153 (1997) [24] Pearman, A.T.; Castro-Faria-Neto, H.C.; McIntyre, T.M.; Prescott, S.M.; Stafforini, D.M.: Characterization of human UMP-CMP kinase enzymatic activity and 5' untranslated region. Life Sci., 69, 2361-2370 (2001) [25] Liou, J.Y.; Dutschman, G.E.; Lam, W.; Jiang, Z.; Cheng, Y.C.: Characterization of human UMP/CMP kinase and its phosphorylation of d- and l-form deoxycytidine analogue monophosphates. Cancer Res., 62, 1624-1631 (2002)

596

2.7.4.14

Cytidylate kinase

[26] Pasti, C.; Gallois-Montbrun, S.; Munier-Lehmann, H.; Veron, M.; Gilles, A.M.; Deville-Bonne, D.: Reaction of human UMP-CMP kinase with natural and analog substrates. Eur. J. Biochem., 270, 1784-1790 (2003) [27] Briozzo, P.; Golinelli-Pimpaneau, B.; Gilles, A.M.; Gaucher, J.F.; BurlacuMiron, S.; Sakamoto, H.; Janin, J.; Barzu, O.: Structures of Escherichia coli CMP kinase alone and in complex with CDP: a new fold of the nucleoside monophosphate binding domain and insights into cytosine nucleotide specificity. Structure, 6, 1517-1527 (1998)

597

)%  0 % %  

     2.7.4.15     ATP:thiamine-diphosphate phosphotransferase    thiamine-diphosphate kinase   

ATP:thiamin-diphosphate phosphotransferase TDP kinase kinase (phosphorylating), thiamin diphosphate kinase, thiamin diphosphate (phosphorylating) protein bound thiamin diphosphate:ATP phosphoryltransferase thiamin diphosphate kinase thiamin diphosphate phosphotransferase thiamin pyrophosphate kinase thiamin-diphosphate kinase thiamine diphosphate kinase thiamine pyrophosphate-ATP phosphoryltransferase     9075-79-0

  



Rattus norvegicus [1, 5] Bos taurus [2, 6] Cavia porcellus [3] Sus scrofa [4] Homo sapiens [7]

       !   "   ATP + thiamine diphosphate = ADP + thiamine triphosphate

598

35

2.7.4.15

Thiamine-diphosphate kinase

    phospho group transfer     

 ATP + thiamine diphosphate (Reversibility: r [1]; ? [27]) [1-7] # ADP + thiamine triphosphate  CTP + thiamine diphosphate ( 6% of the activity with ATP [6]) (Reversibility: ? [6]) [6] # CDP + thiamine triphosphate $ %  

HgCl2 ( 10 mM, complete inhibition [7]) [7] NEM ( 10 mM, 64% inhibition [2,6]) [2, 6] PCMB ( 0.5 mM, 79.5% inhibition [2,6]; 10 mM, complete inhibition [7]) [2, 6, 7]  &    

creatine ( the enzyme is dependent on creatine [4]) [4] Additional information ( a low molecular weight cofactor is required [6]) [6] ' ( 

Co2+ ( about 50% of the activation with Mg2+ [6]) [6] Fe2+ ( 30 mM FeCl2 , maximal activation of approximately 30% [7]) [7] Fe3+ ( 30 mM FeCl3 2, maximal activation of approximately 30% [7]) [7] Mg2+ ( required [2,3,4,6]) [2, 3, 4, 6] Mn2+ ( about 50% of the activation with Mg2+ [6]) [6] /01 *', 0.01 (ATP) [3] 0.6 (ATP, pH 7.4, 37 C [6]) [6] 1.1 (thiamine diphosphate) [3] 20   6-6.5 [5] 7.5 ( in Tris-HCl buffer slightly higher activity than in phosphate buffer [2,6]) [2, 6] 11 [3] )     * , 25 [3]

3  "  ' 4 % 103000 [2]

599

Thiamine-diphosphate kinase

5 $   .#   .'  .

2.7.4.15

  

.

 brain ( cortex [2,6]) [1, 2, 3, 6] erythrocyte [7] heart [3] liver [5] skeletal muscle [3, 4] 6 "  cytosol [3, 4] microsome [1] mitochondrion ( primarily localized in [1]) [1] # !   [2, 6] (partial [3]) [3]

! 

[1] Itokawa, Y.; Cooper, J.R.: The enzymatic synthesis of triphosphothiamin. Biochim. Biophys. Acta, 158, 180-182 (1968) [2] Cooper, J.R.; Nishino, K.: Enzymatic synthesis of thiamin triphosphate. Methods Enzymol., 122, 24-29 (1986) [3] Koyama, S.; Egi, Y.; Shikata, H.; Yamada, K.; Kawasaki, T.: Existence in animal tissues of adenosine triphosphate thiamin diphosphate phosphotransferase [EC 2.7.4.15]. Biochem. Int., 11, 371-380 (1985) [4] Shikata, H.; Koyama, S.; Egi, Y.; Yamada, K.; Kawasaki, T.: Identification of creatine as a cofactor of thiamin-diphosphate kinase. FEBS Lett., 201, 101104 (1986) [5] Ruenwongsa, P.; Cooper, J.R.: The role of bound thiamine pyrophosphate in the synthesis of thiamine triphosphate in rat liver. Biochim. Biophys. Acta, 482, 64-70 (1977) [6] Nishino, K.; Itokawa, Y.; Nishino, N.; Piros, K.; Cooper, J.R.: Enzyme system involved in the synthesis of thiamin triphosphate. I. Purification and characterization of protein-bound thiamin diphosphate: ATP phosphoryltransferase. J. Biol. Chem., 258, 11871-11878 (1983) [7] Yamaguchi, T.; Uchimura, K.; Mishiro, N.; Watanabe, K.: Evidence for the presence of thiamin diphosphate kinase in human erythrocytes. JPN. J. Toxicol. Environ. Health, 42, 524-528 (1996)

600

)%  0 % %  

37

     2.7.4.16     ATP:thiamine-phosphate phosphotransferase    thiamine-phosphate kinase   

ATP:thiamin-phosphate phosphotransferase kinase, thiamin monophosphate (phosphorylating) thiamin monophosphatase thiamin monophosphate kinase thiamin monophosphokinase thiamin phosphate kinase thiamin-monophosphate kinase thiamin-phosphate kinase thiamine monophosphate kinase thiamine monophosphokinase     9068-23-9

   Escherichia coli (K12 [1]) [1] Salmonella typhimurium (LT2 [2]) [2]

       !   "   ATP + thiamine phosphate = ADP + thiamine diphosphate     phospho group transfer

601

Thiamine-phosphate kinase

2.7.4.16

     

 ATP + thiamine phosphate ( enzyme plays an important role in the thiamine diphosphate biosynthetic pathway, thiamine diphosphate is the regulatory molecule for thiamine synthesis and predicts the existence of a sensor/regulatory protein [2]) (Reversibility: ? [1, 2]) [1, 2] # ADP + thiamine diphosphate [1, 2]     

 ADP + thiamine phosphate ( poor substrate [1]) [1] # ?  ATP + thiamine phosphate ( no substrates: thiamine, GTP, CTP, UTP, AMP [1,2]) [1, 2] # ADP + thiamine diphosphate [1, 2]  ITP + thiamine phosphate ( poor substrate [1]) [1] # ? $ %  

ADP ( product inhibition [1]) [1] AMP [1] Cs+ ( antagonizes stimulation by K+ [1]) [1] diphosphate [1] EDTA [1] Li+ ( antagonizes stimulation by K+ [1]) [1] N-ethylmaleimide [1] Na+ ( antagonizes stimulation by K+ [1]) [1] NaF ( weak [1]) [1] PCMB ( 2-mercaptoethanol reverses [1]) [1] oxythiamine [1] pyrithiamine [1] pyrithiamine phosphate [1] thiamine [1] Additional information ( no inhibition by phosphate, KCN, iodoacetic acid, arsenate or arsenite [1]) [1] ' ( 

Co2+ ( requirement, about 95% as effective as Mg2+ [1]) [1] Fe2+ ( activation, about 10% as effective as Mg2+ [1]) [1] K+ ( activation [1]) [1] Mg2+ ( requirement [1]) [1] Mn2+ ( activation, about 30% as effective as Mg2+ [1]) [1] NH+4 ( activation [1]) [1] Rb+ ( activation [1]) [1] Zn2+ ( activation, about 30% as effective as Mg2+ [1]) [1] Additional information ( no activation by Ba2+ , Ca2+ , Cu2+ , Cs+ , Li+ , Na+ [1]) [1]   !   &  *-., 0.000197 [1] 602

2.7.4.16

Thiamine-phosphate kinase

/01 *', 0.0011 (thiamine phosphate, pH 7.5, 37 C [1]) [1] 0.27 (ATP, pH 7.5, 37 C [1]) [1] / 01 *', 0.36 (pyrithiamine, pH 7.5, 37 C [1]) [1] 20   8 [1]

3  "  ' 4 % 35000 ( recombinant enzyme, amino acid sequence [2]) [2]  

monomer ( 1 * 35000, SDS-PAGE [2]) [2]

5 $   .#   .'  .

  

6 "  soluble [1] # !   (partial [1]) [1]   (encoded by thiL, thiL is not transcriptionally regulated by thiamine or thiamine diphosphate [2]) [2]

7    8     !  , glycerol stabilizes during purification [1]     , -20 C, 3 days, t1=2 : 10 days [1]

! 

[1] Nishino, H.: Biogenesis of cocarboxylase in Escherichia coli. Partial purification and some properties of thiamine monophosphate kinase. J. Biochem., 72, 1093-1100 (1972) [2] Webb, E.; Downs, D.: Characterization of thiL, encoding thiamin-monophosphate kinase, in Salmonella typhimurium. J. Biol. Chem., 272, 15702-15707 (1997) 603

0#% %0 % %0  % % % % ! 

3

     2.7.4.17     3-phospho-d-glyceroyl-phosphate:polyphosphate phosphotransferase    3-phosphoglyceroyl-phosphate-polyphosphate phosphotransferase   

1,3-diphosphoglycerate-polyphosphate phosphotransferase 1,3-diphosphoglycerate:polyphosphate-phosphotransferase 3-phosphoglycerol phosphate-polyphosphate phosphotransferase phosphotransferase, diphosphoglycerate-polyphosphate     9055-36-1

  



Escherichia coli (strain B [1]) [1] Micrococcus lysodeikticus [1] Neurospora crassa (wild type and 286-10HSa mutant [1]) [1] Penicillium chrysogenum (Q-176 [1]) [1] Propionibacterium shermanii [1]

       !   "   3-phospho-d-glyceroyl phosphate + (phosphate)n = 3-phosphoglycerate + (phosphate)n+1     phospho group transfer      

 1,3-diphosphoglyceric acid + (polyphosphate)n ( n is 180 [1]) (Reversibility: ? [1]) [1] # 3-phosphoglycerate + (polyphosphate)n+1 [1]

604

2.7.4.17

3-Phosphoglyceroyl-phosphate-polyphosphate phosphotransferase

    

 1,3-diphosphoglyceric acid + (polyphosphate)n ( n is 180 [1]) (Reversibility: ? [1]) [1] # 3-phosphoglycerate + (polyphosphate)n+1 [1] 20   7.2 ( assay at [1]) [1] )     * , 37 ( assay at [1]) [1]

5 $   .#   .'  .

  

# !   (strain B, partial [1]) [1] (partial [1]) [1] (wild type and 286-10HSa mutant, partial [1]) [1] (Q-176, partial [1]) [1] (partial [1]) [1]

7        , 2-4 C, crude extract, storage for 5-7 h [1]

! 

[1] Kulaev, I.S.; Bobyk, M.A.; Nikolaev, N.N.; Sergeev, N.S.; Uryson, S.O.: Polyphosphate-synthesizing enzymes of some fungi and bacteria. Biokhimiya, 36, 943-949 (1971)

605

B  0 % %  

3