identifying unknown substrates of s-adenosyl-methionine ...rx915f228/... · dissociation (cid) and...

155
Identifying Unknown Substrates of S-Adenosyl-methionine-dependent Methyltransferases via in situ Formation of Tight Binding Bisubstrate Adducts by Wanlu Qu A dissertation submitted to The Faculty of the College of Science of Northeastern University in partial fulfillment of the requirements for the degree of Doctor of Philosophy April 10, 2015 Dissertation directed by Zhaohui Sunny Zhou Professor of Chemistry and Chemical Biology

Upload: phamphuc

Post on 27-Nov-2018

222 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

Identifying Unknown Substrates of S-Adenosyl-methionine-dependent Methyltransferases via in

situ Formation of Tight Binding Bisubstrate Adducts

by Wanlu Qu

A dissertation submitted to

The Faculty of

the College of Science of

Northeastern University

in partial fulfillment of the requirements

for the degree of Doctor of Philosophy

April 10, 2015

Dissertation directed by

Zhaohui Sunny Zhou

Professor of Chemistry and Chemical Biology

Page 2: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

ii

Acknowledgements

First of all, I would like to thank my family, my grandparents and my parents for always

supporting me at my every decision and for helping me out at my every stage. They are the best

family in the world!

I would like to thank my advisor, Professor Zhaohui Sunny Zhou for offering me the

opportunity to pursue my PhD study at Northeastern University, for his many years of leading me

to think and perform as a scientist, for all his suggestions, and for always being critical, caring and

understanding. Sunny is one of the greatest mentors that I have ever worked with.

I would like to thank my labmates, Dr. Tianzhu Indi Zang, Shanshan Liu, Kalli Catcott,

Kevin Moulton, Michael Pablo, Dillon Cleary, Dr. Chris Chumsae, Dr. Min Liu, Lihua Yang,

Minjie Pan, Nathaniel Kenton, Yuan Xiu, and Tianyi Kevin Bai, for their wonderful suggestions

in both my research and life. I would like to thank Professor Richard Duclos, for his patience and

understanding when working with me. I am very lucky to work with them, lovely Sunnylanders.

I would like to thank Professor Vicki Wysocki at Ohio State University for her brilliant

ideas. I would like to thank Jing Yan in Professor Vicki Wysocki group, for her hardworking

during the collaboration with us.

I would like to thank Professor Penny Beuning, Professor Michael Pollastri and Professor

Ke Zhang, for being my committee members, for offering helpful suggestions, and for helping me

out on my research journey.

I would like to thank Professor Paul Vouros and Professor Roger Giese, for their support

in Mass Spectrometry. I would like to thank the scientists in Barnett Institute, Dr. Poguang Wang,

Dr. Shujia Daniel Dai, Dr. James Glick, Dr. David Bush, and Dr. Joey Salisbury, for their excellent

Page 3: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

iii

ideas regarding my experiments and training me in analytical techniques. I would also like to thank

all the graduate students in the laboratories of Professors Beuning, Pollastri and Zhang, for offering

me great help with my experiments in biology and organic chemistry.

I would like to thank Andrew Bean, Cara Shockley, and Richard Pumphrey, for their help

on the daily life in Department of Chemistry and Chemical Biology.

And last, but not the least, I would like to thank all my friends, Dr. Wenqin Ni, Fan Anna

Zhang, Yu Annie Wang, Dr. Yashan Zhong, Pei Shi and Dr. Kun Zhang for their support in my

life. Their help and caring are one of the best memories.

Page 4: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

iv

Abstract of Dissertation

Identification of enzyme substrates is an essential step toward understanding the

physiological functions of enzymes. However, progress in substrate elucidation is hampered by

the transient nature of enzyme-substrate interactions. This challenge is exemplified by S-adenosyl-

methionine (AdoMet or SAM) dependent methyltransferases (MTases). AdoMet dependent

MTases catalyze the methyl-transfer reaction from AdoMet to a wide array of nucleophilic

substrates. These nucleophilic substrates, however, are only transiently associated to the enzyme.

Inspired by the concept of multisubstrate adduct inhibition, a novel approach of enzyme-catalyzed

in situ bisubstrate-adduct formation is presented for capturing substrates of MTases via tight

binding MTase-adduct complexes. In our approach, the electrophilic methyl sulfonium in AdoMet

is replaced with a vinyl sulfonium in S-adenosyl-vinthionine (AdoVin). AdoVin forms a covalent

bisubstrate-adduct with the nucleophilic substrate and the resulting adduct tightly binds the

enzyme. Using this strategy, an unknown substrate was readily identified in crude cell lysates, and

was found to be a derivative of exogenous substrates.

Another significant achievement is the detection of the complex between the bisubstrate-

adduct and enzyme using native mass spectrometry directly from the reaction mixture, without

prior purification even using crude cell lysates. The subsequent identification of substrates was

achieved by employing various tandem mass spectrometric techniques, such as collision induced

dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our

knowledge, this is the first example of MTase substrate identification from cell lysates without any

separation prior to mass spectrometric analysis. The approaches outlined here should be generally

applicable for identifying substrates of other group transferases or enzymes sharing similar

mechanisms.

Page 5: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

v

Table of Contents

Acknowledgements ......................................................................................................................... ii

Abstract of Dissertation ................................................................................................................. iv

Table of Contents ............................................................................................................................ v

List of Schemes .............................................................................................................................. ix

List of Figures ................................................................................................................................ xi

List of Tables ................................................................................................................................ xv

List of Abbreviations and Symbols.............................................................................................. xvi

Chapter 1: Overview of Methylation and Methyltransferases ........................................................ 1

1.1. Protein post-translational modifications ................................................................................ 1

1.2. Methylation and S-Adenosyl-Methionine Dependent Methyltransferases ............................ 3

1.3. Analytical Methods for Methylation ...................................................................................... 5

1.3.1. Radioactivity Assay ............................................................................................................ 5

1.3.2. Mass Spectrometry with Stable Isotope Labeling .............................................................. 5

1.3.3. Design of Traceable S-adenosyl-methionine Analogues .................................................... 7

1.4. Multisubstrate Adduct Inhibitors ......................................................................................... 10

1.5. Bisubstrate-adduct Formation via S-adenosyl-vinthionine .................................................. 12

1.6. Conclusions .......................................................................................................................... 14

1.7. References ............................................................................................................................ 15

Chapter 2: Affinity Capturing Substrates via in situ Formation of Bisubstrate-Adducts ............. 20

Page 6: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

vi

2.1 Introduction .......................................................................................................................... 21

2.1.1 S-Adenosyl-methionine Synthetase .................................................................................. 21

2.2 Results and Discussions ....................................................................................................... 24

2.2.1 Enzymatic Synthesis of S-adenosyl-vinthionine ............................................................... 24

2.2.2 in situ formation of AdoVin Adduct Catalyzed by Methyltransferases ........................... 31

2.2.3 Substrate Specificity toward AdoVin ............................................................................... 38

2.2.4 Tight Binding between Bisubstrate-Adduct and Methyltransferases ............................... 42

2.2.5 Kinetics of Methyltransferases-catalyzed AdoVin Adduct Formation ............................. 47

2.2.6 Discovery and Identification of Unknown Substrates of Methyltransferases .................. 51

2.2.6.1 Discovery of Unknown Adduct ..................................................................................... 51

2.2.6.2 Elucidation and Confirmation of Unknown Adduct ...................................................... 56

2.2.6.3 Confirmation of 2-Amino-5-Mercaptobenzoic Acid as TPMT Substrate ..................... 61

2.2.7 No Modification of Enzymes by AdoVin ......................................................................... 64

2.3 Conclusions .......................................................................................................................... 67

2.4 Experimental Section ........................................................................................................... 68

2.4.1 General Procedures ........................................................................................................... 68

2.4.2 Expression and Purification of Enzymes .......................................................................... 69

2.4.3 Protein Precipitation.......................................................................................................... 71

2.4.4 Preparation of Vinthionine ................................................................................................ 72

2.4.5 Enzymatic Synthesis of AdoVin ....................................................................................... 75

Page 7: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

vii

2.4.6 in vitro Formation of AdoVin Adduct in MAT-TPMT Coupled Reactions ..................... 75

2.4.7 Binding Assay ................................................................................................................... 77

2.4.8 ex vivo AdoVin Adduct Formation Catalyzed by TPMT ................................................. 78

2.4.9 Elucidation and Confirmation of Unknown Adduct ......................................................... 79

2.4.10 Confirmation of 2-Amino-5-Mercaptobenzoic Acid as TPMT Substrate ........................ 80

2.4.11 Modification of MAT and TPMT by AdoVin .................................................................. 80

2.4.12 Kinetics Analysis of TPMT-Catalyzed AdoVin-TNB Adduct Formation ....................... 81

2.4.13 HPLC Analysis of AdoVin and AdoVin reactions ........................................................... 81

2.4.14 Mass spectrometry ............................................................................................................ 83

2.4.15 MALDI-TOF/TOF Analysis ............................................................................................. 84

2.5 References ............................................................................................................................ 85

Chapter 3: Identification of Substrates by Native Mass Spectrometry ......................................... 88

3.1 Introduction .......................................................................................................................... 89

3.1.1 Native Mass Spectrometry ................................................................................................ 89

3.1.2 Tandem Mass Spectrometry and Fragmentation in Tandem MS ..................................... 90

3.1.3 Ion Mobility Spectrometry with Mass Spectrometry........................................................ 91

3.1.4 Identification of Enzyme Substrates ................................................................................. 92

3.1.5 Identification of Enzyme Substrates via Enzyme-Substrate Complex ............................. 93

3.2 Results and Discussions ....................................................................................................... 95

3.2.1 TPMT in Gas Phase .......................................................................................................... 95

Page 8: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

viii

3.2.2 Detection of Enzyme-Substrate Complex ......................................................................... 98

3.2.3 Detection of Enzyme-Substrate Complex in Crude Biological Samples........................ 103

3.2.4 Ion Mobility and Mobiligram ......................................................................................... 114

3.2.5 Detection of Potential Modification on MTases by AdoVin .......................................... 118

3.3 Conclusions ........................................................................................................................ 119

3.4 Experimental Section ......................................................................................................... 120

3.4.1 General Procedures ......................................................................................................... 120

3.4.2 HPLC Analysis ............................................................................................................... 120

3.4.3 Mass Spectrometry.......................................................................................................... 121

3.4.4 Preparation of in vitro Enzyme-Substrate Complex ....................................................... 123

3.4.5 Preparation of Enzyme-Substrate Complex in Crude Biological Samples ..................... 123

3.5 References .......................................................................................................................... 124

Chapter 4: Conclusions and Future Directions ........................................................................... 128

4.1. AdoVin Bisubstrate-adduct Formation in Whole Cells ..................................................... 129

4.2. Affinity Capturing Unknown Methyltransferases ............................................................. 130

4.3. Substrate Identification for Other MTases using Native Mass Spectrometry via Enhanced

Binding without Formation of Covalent Adducts .............................................................. 132

4.4. Facile Generation of Bisubstrate-adduct for Structural Investigation ............................... 133

4.5. References .......................................................................................................................... 135

Page 9: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

ix

List of Schemes

Scheme 1.1. Deamidation of asparagine and formation of aspartic acid and isoaspartic acid. ...... 2

Scheme 1.2. AdoMet-dependent MTases catalyze methyl transfer from AdoMet to the nucleophilic

substrates. ................................................................................................................... 3

Scheme 1.3. in vivo methylation workflow, showing uptake of methionine into the cells, enzymatic

synthesis of AdoMet, and subsequent methylation.................................................... 6

Scheme 1.4. MTase catalyzed transfer of a methyl, propargyl or ketone group. The traceable

products can be detected via click or oxime chemistry ............................................. 7

Scheme 1.5. Transient interactions between the substrates and products with methyltransferases.

.................................................................................................................................... 9

Scheme 1.6. COMT catalyzed methylation of epinephrine via a SN2 colinear transition state and

structural design of multisubstrate adduct for COMT. ............................................ 11

Scheme 1.7. Aziridinium ion formed from the N-mustard in situ reacted with arginine catalyzed

by PRMT1 to produce bisubstrate-adduct. .............................................................. 12

Scheme 1.8. Transient interactions between the substrates and products with methyltransferases,

and “permanent” interaction between an enzyme and the bisubstrate-adduct formed

in situ. ....................................................................................................................... 13

Scheme 1.9. AdoMet-dependent MTases catalyze bisubstrate-adduct formation between AdoVin

and nucleophilic substrates. ..................................................................................... 13

Scheme 2.1. MAT catalyzed AdoMet synthesis stoichiometrically from ATP and methionine. . 21

Scheme 2.2. Formation of isotope labeled bisubstrate-adduct via isotope labeled ATP. ............. 22

Page 10: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

x

Scheme 2.3. Synthesis of vinthionine. .......................................................................................... 24

Scheme 2.4. Synthesis of S-adenosyl-vinthionine catalyzed by MAT. ........................................ 26

Scheme 2.5. TPMT catalyzed methylation of TNB and formation of bisubstrate-adduct between

AdoVin and TNB. .................................................................................................... 32

Scheme 2.6. Synthesis of 2-amino-5-mercaptobenzoic acid. ....................................................... 56

Scheme 2.7. in vitro bisubstrate-adduct formation between AdoVin and AMBA catalyzed by

thiopurine S-methyltransferase. ............................................................................... 57

Scheme 2.8. Proposed methylation of 2-amino-5-mercaptobenzoic acid catalyzed by thiopurine

methyltransferase. .................................................................................................... 61

Scheme 3.1. Detection of enzyme-ligand complex by using native mass spectrometry and

subsequent structural elucidation by tandem mass spectrometry. ........................... 94

Scheme 3.2. Workflow of the Q-IM-TOF instrument. The arrow shows the ion trajectory. ..... 121

Scheme 4.1. in vivo methylation and bisubstrate-adduct formation starting from methionine and

vinthionine, respectively. ....................................................................................... 129

Scheme 4.2. Proposed AdoVin bisubstrate-adduct formation catalyzed by TMT and proposed

isolation of TMT catalyzed by affinity tag (red) on AdoVin bisubstrate-adduct. . 131

Scheme 4.3. Substrate binding in methylation using AdoMet and proposed binding between

nucleophilic substrates and methyltransferases enhanced by AdoVin. ................. 132

Scheme 4.4. Potential generation of bisubstrate-adduct inhibitors by derivatizing other

nucleophilic substrates (such as -NH2 and -OH) to thiol substrates (-SH).. .......... 134

Page 11: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

xi

List of Figures

Figure 2.1. Mass spectrum of vinthionine. ................................................................................... 25

Figure 2.2. CID MS/MS spectrum of vinthionine. ....................................................................... 25

Figure 2.3. Reverse-phase HPLC chromatogram of AdoVin synthesis with or without MAT. ... 27

Figure 2.4. UV/Vis spectra of synthesized AdoVin and AdoMet. ............................................... 27

Figure 2.5. Strong cation exchange HPLC chromatogram showing the MAT-catalyzed synthesis

of AdoVin and AdoMet. ............................................................................................. 28

Figure 2.6. Extracted ion chromatogram of m/z 411.0. ................................................................ 29

Figure 2.7. Mass spectrum of AdoVin. ......................................................................................... 30

Figure 2.8. CID MS/MS spectrum of AdoVin. ............................................................................. 30

Figure 2.9. HPLC chromatogram of AdoVin-TNB adduct formation.......................................... 33

Figure 2.10. UV/Vis absorbance spectra of adenosine, methylated TNB and AdoVin-TNB adduct.

.................................................................................................................................. 34

Figure 2.11. Extracted ion chromatogram of m/z 411.0, and 610.0 in AdoVin-TNB reaction. ... 36

Figure 2.12. Mass spectrum of AdoVin-TNB adduct. .................................................................. 37

Figure 2.13. CID MS/MS spectrum of AdoVin-TNB adduct ....................................................... 37

Figure 2.14. Selected ion chromatogram of m/z 599.0 in AdoVin-2-Bromothiophenol and AdoVin-

3-Bromothiophenol reactions................................................................................... 39

Figure 2.15. Extracted ion chromatogram of m/z 599.0 in AdoVin-2BrPhSH reaction and negative

control. ..................................................................................................................... 40

Page 12: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

xii

Figure 2.16. HPLC chromatogram of in vitro AdoVin-Nitrophenol reactions and negative control

at 3 hours incubation. ............................................................................................... 41

Figure 2.17. HPLC chromatogram of the TPMT complex and free ligand fraction with UV

detection at 325 nm. ................................................................................................. 43

Figure 2.18. Extracted ion chromatogram of m/z 162.0 and 610.0. ............................................. 44

Figure 2.19. Selected ion chromatogram of m/z 599.0 for the binding assay between AdoVin-

BrPhSH adduct and TPMT.. .................................................................................... 45

Figure 2.20. CID MS/MS spectra of AdoVin-2BrPhSH and AdoVin-3BrPhSH adduct. ............ 46

Figure 2.21. Absorbance change of TNB at 411 nm of AdoVin-TNB reactions catalyzed by TPMT

under various concentrations ................................................................................... 48

Figure 2.22. The overall concentration change of TNB at 411 nm at different concentration of

TPMT, indicating a single turnover of the reaction. ................................................ 49

Figure 2.23. Selected ion chromatogram of AdoVin adducts in isolated TPMT fractions from crude

cell lysates of ex vivo AdoVin-TNB and negative control. ..................................... 52

Figure 2.24. HPLC chromatogram of ex vivo AdoVin-TNB reaction at 1 hour, elution containing

TPMT, and free ligands fraction. ............................................................................. 53

Figure 2.25. UV-Vis spectra of AdoVin-TNB adduct and unknown adduct. ............................... 54

Figure 2.26. Extracted ion chromatogram of m/z 610.0 and 580.0 in TPMT fraction of ex vivo

AdoVin-TNB and ex vivo negative control. ............................................................ 54

Figure 2.27. CID MS/MS spectra of AdoVin-TNB adduct, regular and isotope labeled unknown

adduct. ...................................................................................................................... 55

Page 13: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

xiii

Figure 2.28. HPLC chromatogram of ex vivo AdoVin-TNB reaction; co-injection of ex vivo

AdoVin-TNB reaction and authentic AdoVin-AMBA adduct; and authentic AdoVin-

AMBA adduct. ......................................................................................................... 58

Figure 2.29. UV spectra of ex vivo AdoVin-AMBA adduct and authentic AdoVin-AMBA adduct

standard. ................................................................................................................... 59

Figure 2.30. Extracted ion chromatogram of m/z 580.00 and 595.00. ......................................... 59

Figure 2.31. CID MS/MS spectra of AdoVin-TNB adduct, regular and isotope labeled AdoVin-

AMBA adduct. ......................................................................................................... 60

Figure 2.32. HPLC chromatogram of TPMT catalyzed methylation of 2-amino-5-mercaptobenzoic

acid. .......................................................................................................................... 62

Figure 2.33. UV spectra of methylated AMBA and AMBA. ....................................................... 63

Figure 2.34. MALDI mass spectrum of native MAT and treatment with AdoVin. ..................... 65

Figure 2.35. MALDI mass spectrum of native TPMT and treatment with AdoVin. .................... 66

Figure 2.36. 1H-NMR spectrum of vinthionine. ........................................................................... 73

Figure 2.37. 13C-NMRspectrum of vinthionine. ........................................................................... 74

Figure 3.1. Mass spectra of TPMT and zoomed in mass spectra of TPMT. ................................ 96

Figure 3.2. Tandem mass spectrum of +9 precursor ions by CID. ............................................... 97

Figure 3.3. Mass spectrum of reaction mixture of TPMT catalyzed AdoVin-TNB adduct formation,

and zoomed in mass spectrum of + 10 charged TPMT complex. ............................. 99

Figure 3.4. Tandem mass spectra of +10 charged TPMT-ligand complex in AdoVin-TNB reaction

mixture under different CID energy. ....................................................................... 101

Page 14: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

xiv

Figure 3.5. Tandem mass spectra of +10 charged TPMT-ligand complex in AdoVin-TNB reaction

mixture under different SID energy. ......................................................................... 102

Figure 3.6. Mass spectrum of ex vivo reaction mixture of TPMT catalyzed AdoVin-TNB adduct

formation. ................................................................................................................ 103

Figure 3.7. Tandem mass spectra of +10 charged TPMT-ligand complex in ex vivo AdoVin-TNB

reaction by CID and SID. ........................................................................................ 104

Figure 3.8. Mass spectrum of ex vivo reaction mixture of TPMT catalyzed AdoVin-TNB adduct

formation in TPMT lysate from non-induced E. coli cells. ..................................... 105

Figure 3.9. Tandem mass spectrum of +10 charged TPMT-ligand complex in ex vivo AdoVin-

TNB reaction with non-induced TPMT lysate by CID. .......................................... 106

Figure 3.10. Mass spectra of TPMT-AdoMet complex. ............................................................. 108

Figure 3.11. Tandem mass spectrum of +10 TPMT-AdoMet peak dissociated by low energy CID.

................................................................................................................................ 109

Figure 3.12. Tandem mass spectra of +10 TPMT-AdoMet peak under CID and SID. .............. 110

Figure 3.13. Mass spectra of in vitro AdoVin-TNB reaction with and without MTAN. ........... 111

Figure 3.14. Tandem mass spectra of +10 TPMT-AdoHcy peak under various CID energies. . 112

Figure 3.15. Tandem mass spectra of AdoHcy under various CID energies. ............................. 113

Figure 3.16. Mobiligram of reaction mixture of in vitro AdoVin-TNB adduct formation catalyzed

by thiopurine methyltransferase. ............................................................................ 115

Figure 3.17. Mobiligram of +10 TPMT-AdoVin-TNB complex under CID. ............................ 116

Figure 3.18. Mobiligram of +10 TPMT-AdoVin-TNB complex under SID. ............................. 117

Page 15: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

xv

List of Tables

Table 2.1. Calculation of extinction coefficient of the AdoVin-TNB Adduct. ............................ 35

Table 2.2. Similar substrate specificity toward AdoVin and AdoMet .......................................... 38

Table 2.3. Main components in the fractions of isolated TPMT and free ligands. ....................... 43

Table 2.4. Fitting parameters of first order kinetics...................................................................... 50

Page 16: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

xvi

List of Abbreviations and Symbols

°C

13C

15N

µL

µM

β

AdoMet

AdoHcy

AdoVin

AMBA

ATP

CADD

CASS

CID

COMT

D

Degree Celsius

Carbon-13

Nitrogen-15

Microliter

Micromolar

Beta

S-adenosyl methionine

S-adenosyl homocysteine

S-adenosyl vinthionine

2-amino-5-mercaptobenzoic acid

Adenosine-5’-triphosphate

Computer-aided drug design

Computer-aided substrate screening

Collision induced dissociation

Catechol O-methyltransferase

Deuterium

Page 17: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

xvii

D (Asp)

Da

DNA

ETD

Hcy

HPLC

h

i.d.

IMAC

IMS

K (Lys)

KCl

kV

LC-MS

m/z

M-1cm-1

Aspartate acid

Dalton

Deoxyribonucleic acid

Electron transfer dissociation

Homocysteine

High-performance liquid chromatography

Hour

Internal Diameter

Immobilized metal ion affinity

chromatography

Ion mobility spectrometry

Lysine

Potassium Chloride

Kilovolt

Liquid chromatography-mass spectrometry

Mass to charge ratio

Per molar per centimeter

Page 18: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

xviii

MALDI

MAT

MgCl2

mg/mL

min

mM

MS

MS/MS

MTAN

MTases

N (Asn)

NaCl

Nu

PIMT

PTMs

R (Arg)

RNA

rpm

Matrix-assisted laser desorption/ionization

L-methionine S-adenosyltransferase

Magnesium chloride

Milligram per milliliter

Minute

Millimolar

Mass spectrometry

Tandem mass spectrometry

5’-methylthioadenosine nucleosidase

Methyltransferases

Asparagine

Sodium Chloride

Nucleophile

Protein L-isoaspartyl O-methyltransferase

Protein posttranslational modifications

Arginine

Ribonucleic acid

Revolutions per minute

Page 19: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

xix

SDS-PAGE

SID

TCA

TFA

TCEP

TIC

TLC

TMT

TNB

TOF

TPMT

Tris

UV-Vis

v

XIC

Sodium dodecyl sulfate polyacrylamide gel

electrophoresis

Surface induced dissociation

Trichloroacetic acid

Trifluoroacetic acid

Tris(2-carboxyethyl)phosphine

Total ion chromatography

Thin layer chromatography

Thiol S-methyltransferase

2-nitro-5-mercaptobenzoic acid

Time of flight

Thiopurine S-methyltransferase

Tris(hydroxymethyl)aminomethane

Ultraviolet-visible spectroscopy

Volume

Extracted ion chromatogram

Page 20: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

1

Chapter 1: Overview of Methylation and Methyltransferases

1.1. Protein post-translational modifications

Protein post-translational modifications (PTMs) are one of the essential routes for protein

diversification. PTMs are covalent modifications of proteins, and can be classified based on

different classification methods.1-2 One classification scheme is based on the modified amino acid

side chains of proteins; for examples, modification of lysine by phosphorylation, N-methylation

and N-acylation; oxidation to sulfoxide from the methionine; potential modifications on asparagine

by deamidation, N-glycosylation, N-ADP-ribosylation and protein splicing; while the potential

covalent modifications for aspartic acid (Asp) are phosphorylation and isomerization to isoaspartic

acid (isoAsp). In a different classification scheme, PTMs can be sorted into five common covalent

additions: phosphorylation,3-5 acylation,6-8 alkylation,8-11 glycosylation,2, 12 and oxidation.2, 13

PTMs are found in numerous classes of proteins and have strong influences with protein

structures and biological functions.2, 14 Thereby, the discovery and characterization of PTMs are

required for the elucidation of protein structure modifications and the study of protein-mediated

biological functions.14-16 However, the biological significance of most of protein side chain

modifications has not been established. Moreover, multiple PTMs often occur at multiple sites of

proteins, which are crucial to their physiological functions.2

Even the smallest PTMs, by changes in mass, can make big differences to protein structures

and functional activities, for example, 0 Da mass change of isomerization between aspartic acid

(Asp) and isoaspartic acid (IsoAsp), and 1 Da mass change of deamidation from Asparagine (Asn)

to Asp (Scheme 1.1).17 Therefore, the detection and analysis of PTMs are important for uncovering

the nature of proteins. Two efficient analysis methods for PTMs are mass spectrometry and

Page 21: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

2

antibody based detection. Mass spectrometry, antibody based detection and their combinations

with other techniques have been widely applied to the determination of site specific PTMs and

quantification.18-21 This is exemplified by the detection and quantification of isoAsp in Asp/Asn

containing proteins22 and glycosylation in protein therapeutics.18 With the rapid development of

biological chemistry, computational chemistry and analytical chemistry, many PTMs have been

discovered, like eliminylation, a PTM of host cells signaling proteins performed by

phosphotheronine lyases.23

Scheme 1.1. Deamidation of asparagine and formation of aspartic acid and isoaspartic acid.

Page 22: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

3

1.2. Methylation and S-Adenosyl-Methionine Dependent Methyltransferases

Methylation involves a transfer of methyl group from the methyl donor S-adenosyl-

methionine (AdoMet) to nucleophilic substrates (Scheme 1.2) through an SN2 reaction with a linear

transition state,24 catalyzed by S-Adenosyl-methionine dependent methyltransferases (AdoMet

dependent MTases). MTases belong to a large and diverse family, catalyzing over 300 different

reactions (EC 2.1.1.1-313), and the number is still increasing. Varied in size, the substrates of

MTases range from small molecules to biological macromolecules, such as proteins, mRNA and

DNA.25-26 Atomic targets of methylation include nitrogen (amines and guanidines), oxygen

(alcohols and carboxylic acids), sulfur (thiols) and even carbon (alkanes).

Scheme 1.2. AdoMet-dependent MTases catalyze methyl transfer from AdoMet to the nucleophilic

substrates.

Page 23: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

4

Currently, MTases have been cataloged into five structurally distinct classes. Only two

families of MTases exhibit definable substrate specificity. One is the Class V SET-domain

containing family of protein lysine MTases and the other is the Class IV SPOUT family of RNA

MTases.26 While the other classes of MTases do not correlate to any certain chemical natures of

their substrates, some of them have considerable overlapping of substrates. As such, the structures

of many putative MTases have been determined, but their substrates and functions remain elusive.

Even though all MTases are considered to directly transfer a methyl group to substrates via

SN2 mechanism with proton transfer throughout the methylation process, a wide variety of

mechanisms have been described to activate the nucleophilic substrates, mostly dependent on the

polarizability of the atomic targets, i.e. N-methylation, O-methylation, S-methylation and C-

methylation.26

The detection of methylation remains challenging, due to their intrinsic properties: the

modification is small relative to overall protein structures (-CH3, 15 Da), has no charge and is

chemically inert. Despite its inert chemical properties, methylation plays an essential role in in

biosynthesis, signal transduction, protein repair, chromatin regulation and gene silencing.25, 27 The

study of MTase substrates is critical to the full understanding and functional analysis of MTases.

Page 24: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

5

1.3. Analytical Methods for Methylation

1.3.1. Radioactivity Assay

Incorporation of radioactive [3H] methyl group is commonly used in the study of in vitro

and in vivo DNA methylation, protein methylation, and even methylation of small molecules.28-29

To be more precise, a [3H] methyl group is transferred from [3H]-CH3-AdoMet to nucleophilic

substrate via the catalysis of methyltransferases, and then the [3H]-CH3 containing products can

be quantified by liquid scintillation, while the unreacted from [3H] methyl-AdoMet can be easily

separated from labeled products using gel filtration,30 filter-binding,31 or thin layer

chromatography.32

[14C] labeling could also be used with higher energy and higher efficiency in liquid

scintillation counting, compared to [3H].33 Compared to radiolabeling by chemical reagents, this

“radioenzymatic” methylation is more specific. Taking the advantage of radiolabeling, not only in

vitro studies but in vivo identification of all methyltransferase substrates is underway. However,

this method has numerous drawbacks, including radioactive wastes, high cost, highly laborious

work, and low throughput. Moreover, the direct connection between the enzymes and the

substrates cannot be established using this methods, aside from the complicated operation

procedure of radio labeling.

1.3.2. Mass Spectrometry with Stable Isotope Labeling

Stable isotope labeling combined with mass spectrometry is an efficient and effective tool

for identification and quantification, especially in proteomics screening, because of specifically

detectable mass shift but consistent properties, i.e. LC retention time, ionization efficiency and

fragmentation patterns.22, 34-38 -CD3 and -13CH have been used for the detection of AdoMet or

Page 25: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

6

methylated products. Combined with mass spectrometry, stable isotope labeling was available to

determine nanomolar concentrations of AdoMet in complicated environment, such as plasma and

even in the cells.39 As shown in Scheme 1.3, a rapid screening for in vivo methylation products

with isotope labeling has been developed by Zhou’s group.40 The cells take up isotope labeled

methionine through the membrane, and AdoMet synthetase catalyzes the synthesis of AdoMet

starting from methionine. Therefore, the isotope -CD3, is introduced into the cells after the

methylation catalyzed by AdoMet-dependent MTases in the cells. The 1:1 ratio of CH3-methionine

and CD3-methionine is introduced in the media, and the isotopic ratio of the methyl group

throughout the transformations will be maintained as a detection signature from the methionine

auxotroph strain E. coli B834 used in the experiments.

Scheme 1.3. in vivo methylation workflow, showing uptake of methionine into the cells, enzymatic

synthesis of AdoMet, and subsequent methylation.

Page 26: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

7

1.3.3. Design of Traceable S-adenosyl-methionine Analogues

Due to the intrinsic properties of the methyl group, small and chemically inert (-CH3, 15

Da), AdoMet analogues have been developed with traceable tags. The methyl group in AdoMet

has been replaced by propargyl,41 ketone,42 and other functional groups.43 After the transfer of

these alkyl groups to the nucleophilic substrates, the resulting products can be labeled and then

traced by click (azide-alkyne) or oxime (ketone-hydroxylamine) chemistry (Scheme 1.4).

Moreover, most AdoMet analogues share similar substrate specificity with AdoMet, which makes

the replacement of methyl group remain similar to native biological activities.

Scheme 1.4. MTase catalyzed transfer of a methyl (AdoMet, natural substrate), propargyl or ketone

group (substrate surrogates). The traceable products can be detected via click or oxime chemistry

Page 27: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

8

As shown in Scheme 1.4, an AdoMet analogue with a propargyl group has been shown to

be a substrate for protein arginine N-methyltransferases 1 (PRMT1), which catalyzes the

methylation of arginine in proteins and peptides. Click chemistry has been applied to label the

propargyl-transferred nucleophilic substrates, which then allows fluorescence detection.41

Similarly, in the methodology utilizing keto-AdoMet, the ketone group was first transferred onto

the nucleophilic substrates, and oxime chemistry was applied for the labeling and detection of

alkylated substrates. This method was used to discover a previously unidentified, 4-

methoxybenzylthiol (an aliphatic thiol), for thiopurine S-methyltransferase (TPMT).42 Thus, the

design of new traceable AdoMet analogues contributes to the identification of substrate specificity

of MTases, which is crucial to fully understand their biological activities.

In practice, a major limitation for such an approach is that the substrates of multiple

enzymes in the same family may be labeled non-specifically, thereby the direct association

between a particular substrate-enzyme pair cannot be readily established. This is exacerbated for

MTases as this large family of enzymes catalyzes over 300 different reactions with considerable

overlap of substrates. For instance, more than fifty protein lysine44 and nine arginine MTases27, 45

exist in humans alone.

Page 28: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

9

Another problem for methylation and for all enzyme-catalyzed reactions, is the transient

interaction between products and enzymes. The direct connection between the substrates and

enzymes is lost due to the immediate dissociation of products from the enzymes. Thus, the

traceable products mentioned often cannot be directly linked to a particular MTase (Scheme 1.5).

The direct connection between substrates and their corresponding enzymes is very important but

challenging for MTases.

catalytic

cycle

Scheme 1.5. Transient interactions between the substrates and products with methyltransferases.

Page 29: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

10

1.4. Multisubstrate Adduct Inhibitors

In drug discovery, different methods have been developed to create new inhibitors to

achieve satisfactory biological activity. One such method is the multisubstrate adduct inhibitor

approach that consists of a covalent link between the substrate analogues, simultaneously present

and reacting in the enzyme active site.46 Potentially, the multisubstrate adduct inhibitor approach

can be utilized for most enzymes that catalyze the reactions with two or more substrates (including

cofactors). A multisubstrate adduct inhibitor contains the key structural features in one molecule,

and moreover, enhanced binding affinity to enzymes due to the synergistic effect from the

substrates.46-48 Therefore, the multisubstrate adduct inhibitor approach is an efficient method to

obtain structural information about enzymes and to create new inhibitors with potentially high

potency. The concept of multisubstrate adduct inhibitor has been successfully applied as a potential

tool to identify unknown enzymes and substrates, i.e. the formation of kinase-substrate complex

via ATP-based cross-linkers.49-52

Many efforts have been made towards design and synthesis of multisubstrate adduct

inhibitor for AdoMet-dependent MTases, such as catechol O-MTase (COMT)24 and protein N-

MTase (PNMT),47 since the mechanism of AdoMet-dependent methylation matches well with the

requirement of multisubstrate adduct inhibitor, a SN2 addition reaction with a colinear transition

state when the nucleophilic substrate attacks the methyl group in AdoMet (Scheme 1.6).

Page 30: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

11

Scheme 1.6. (a) COMT catalyzed methylation of epinephrine via a SN2 linear transition state; (b)

structural design of multisubstrate adduct for COMT, containing the catechol portion, sulfonium

ion and homocysteine (left) or adenosyl moiety (right).

Utilizing the concept of multisubstrate adduct inhibitor, the Rajski and Thompson group

has performed in situ formation of bisubstrate-adduct catalyzed by protein arginine

methyltransferases 1, via an AdoMet analogue with 5’-aziridinyl adenylates,53-54 produced from

5’-(diaminobutyric acid)-N-iodoethyl-5’-deoxyadenosine ammonium hydrochloride (AAI)

(Scheme 1.7). This bisubstrate analogue was expected to be a potential bisubstrate-adduct inhibitor

to PRMT1.53 Even though its ability to identify novel PRMT substrates is not clear, the idea of

multisubstrate adduct inhibitor brings light to the design of new AdoMet analogues.

Page 31: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

12

Scheme 1.7. Aziridinium ion formed from the N-mustard (AAI) in situ reacted with arginine

catalyzed by PRMT1 to produce bisubstrate-adduct.

1.5. Bisubstrate-adduct Formation via S-adenosyl-vinthionine

A conceptually different approach to study methyltransferases is to capture nucleophilic

substrates with the corresponding enzyme by forming bisubstrate-adducts, which should bind

tightly with the enzyme due to the synergistic binding interactions between the enzyme and

moieties in both substrates (Scheme 1.8). This is the premise of multisubstrate adduct inhibitor, a

venerable idea championed by Coward and Pegg,24, 48 as mentioned before.

Our new approach is demonstrated herein with S-adenosyl-vinthionine (AdoVin, Scheme

1.9), in which a vinyl sulfonium replaces the methyl sulfonium in AdoMet. Via an addition reaction

to the vinyl sulfonium, AdoVin and the nucleophilic substrate forms a covalent tight-binding

adduct.

Page 32: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

13

single

turnover

catalytic

cycle

Scheme 1.8. a) Transient interactions between the substrates and products with methyltransferases;

b) “Permanent” interaction (tight binding) between an enzyme and the bisubstrate-adduct formed

in situ.

Scheme 1.9. AdoMet-dependent MTases catalyze bisubstrate-adduct formation between AdoVin

and nucleophilic substrates.

Page 33: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

14

1.6. Conclusions

The detection of methylation remains challenging due to small and chemically inert

properties of the methyl group. Most efforts have been made to develop traceable AdoMet

analogues, in order to facilitate the functional analysis of AdoMet dependent methyltransferases.

However, substrates cannot be linked directly to their corresponding MTases, due to substrate

overlap and transience of interactions between MTases and their substrates/products. Using a

conceptually distinct approach, we designed a new AdoMet analogue to capture nucleophilic

substrates with their corresponding MTases via in situ formation of bisubstrate-adducts.

Page 34: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

15

1.7. References

1. Seo, J.; Lee, K. J., Post-translational modifications and their biological functions:

proteomic analysis and systematic approaches. J Biochem Mol Biol 2004, 37 (1), 35-44.

2. Walsh, C. T.; Garneau-Tsodikova, S.; Gatto, G. J., Jr., Protein posttranslational

modifications: the chemistry of proteome diversifications. Angew Chem Int Ed Engl 2005,

44 (45), 7342-72.

3. Alemany, R.; Perona, J. S.; Sanchez-Dominguez, J. M.; Montero, E.; Canizares, J.;

Bressani, R.; Escriba, P. V.; Ruiz-Gutierrez, V., G protein-coupled receptor systems and

their lipid environment in health disorders during aging. Biochim Biophys Acta 2007, 1768

(4), 964-75.

4. Hubbard, S. R.; Miller, W. T., Receptor tyrosine kinases: mechanisms of activation and

signaling. Curr Opin Cell Biol 2007, 19 (2), 117-23.

5. Beene, D. L.; Scott, J. D., A-kinase anchoring proteins take shape. Curr Opin Cell Biol

2007, 19 (2), 192-8.

6. Couture, J. F.; Trievel, R. C., Histone-modifying enzymes: encrypting an enigmatic

epigenetic code. Curr Opin Struct Biol 2006, 16 (6), 753-60.

7. Shen, S.; Casaccia-Bonnefil, P., Post-translational modifications of nucleosomal histones

in oligodendrocyte lineage cells in development and disease. J Mol Neurosci 2008, 35 (1),

13-22.

8. Margueron, R.; Trojer, P.; Reinberg, D., The key to development: interpreting the histone

code? Curr Opin Genet Dev 2005, 15 (2), 163-76.

9. Smith, B. C.; Denu, J. M., Chemical mechanisms of histone lysine and arginine

modifications. Biochim Biophys Acta 2009, 1789 (1), 45-57.

10. Basso, A. D.; Kirschmeier, P.; Bishop, W. R., Lipid posttranslational modifications.

Farnesyl transferase inhibitors. J Lipid Res 2006, 47 (1), 15-31.

11. Lane, K. T.; Beese, L. S., Thematic review series: lipid posttranslational modifications.

Structural biology of protein farnesyltransferase and geranylgeranyltransferase type I. J

Lipid Res 2006, 47 (4), 681-99.

12. Knorre, D. G.; Kudryashova, N. V.; Godovikova, T. S., Chemical and functional aspects

of posttranslational modification of proteins. Acta Naturae 2009, 1 (3), 29-51.

Page 35: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

16

13. Giles, N. M.; Giles, G. I.; Jacob, C., Multiple roles of cysteine in biocatalysis. Biochem

Biophys Res Commun 2003, 300 (1), 1-4.

14. Blume-Jensen, P.; Hunter, T., Oncogenic kinase signalling. Nature 2001, 411 (6835), 355-

65.

15. Norris, K. L.; Lee, J. Y.; Yao, T. P., Acetylation goes global: the emergence of acetylation

biology. Sci Signal 2009, 2 (97), pe76.

16. Ullrich, A.; Schlessinger, J., Signal transduction by receptors with tyrosine kinase activity.

Cell 1990, 61 (2), 203-12.

17. Noguchi, S., Structural changes induced by the deamidation and isomerization of

asparagine revealed by the crystal structure of Ustilago sphaerogena ribonuclease U2B.

Biopolymers 2010, 93 (11), 1003-10.

18. Alley, W. R., Jr.; Mechref, Y.; Novotny, M. V., Characterization of glycopeptides by

combining collision-induced dissociation and electron-transfer dissociation mass

spectrometry data. Rapid Commun Mass Spectrom 2009, 23 (1), 161-70.

19. Scigelova, M.; Makarov, A., Orbitrap mass analyzer--overview and applications in

proteomics. Proteomics 2006, 6 Suppl 2, 16-21.

20. Perla-Kajan, J.; Stanger, O.; Luczak, M.; Ziolkowska, A.; Malendowicz, L. K.;

Twardowski, T.; Lhotak, S.; Austin, R. C.; Jakubowski, H., Immunohistochemical

detection of N-homocysteinylated proteins in humans and mice. Biomed Pharmacother

2008, 62 (7), 473-9.

21. Perla, J.; Undas, A.; Twardowski, T.; Jakubowski, H., Purification of antibodies against N-

homocysteinylated proteins by affinity chromatography on Nomega-homocysteinyl-

aminohexyl-Agarose. J Chromatogr B Analyt Technol Biomed Life Sci 2004, 807 (2), 257-

61.

22. Liu, M.; Cheetham, J.; Cauchon, N.; Ostovic, J.; Ni, W.; Ren, D.; Zhou, Z. S., Protein

isoaspartate methyltransferase-mediated 18O-labeling of isoaspartic acid for mass

spectrometry analysis. Anal Chem 2012, 84 (2), 1056-62.

23. Ke, Z.; Smith, G. K.; Zhang, Y.; Guo, H., Molecular mechanism for eliminylation, a newly

discovered post-translational modification. J Am Chem Soc 2011, 133 (29), 11103-5.

24. Anderson, G. L.; Bussolotti, D. L.; Coward, J. K., Synthesis and evaluation of some stable

multisubstrate adducts as inhibitors of catechol O-methyltransferase. J Med Chem 1981,

24 (11), 1271-7.

Page 36: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

17

25. Klimasauskas, S.; Weinhold, E., A new tool for biotechnology: AdoMet-dependent

methyltransferases. Trends Biotechnol 2007, 25 (3), 99-104.

26. Schubert, H. L.; Blumenthal, R. M.; Cheng, X., Many paths to methyltransfer: a chronicle

of convergence. Trends Biochem Sci 2003, 28 (6), 329-35.

27. Bedford, M. T.; Clarke, S. G., Protein arginine methylation in mammals: who, what, and

why. Mol Cell 2009, 33 (1), 1-13.

28. Gros, C.; Chauvigne, L.; Poulet, A.; Menon, Y.; Ausseil, F.; Dufau, I.; Arimondo, P. B.,

Development of a universal radioactive DNA methyltransferase inhibition test for high-

throughput screening and mechanistic studies. Nucleic Acids Res 2013, 41 (19), e185.

29. Hortnagl, H.; Benedict, C. R.; Grahame-Smith, D. G., A sensitive radioenzymatic assay

for adrenaline and noradrenaline in plasma. Br J Clin Pharmacol 1977, 4 (5), 553-8.

30. Halby, L.; Champion, C.; Senamaud-Beaufort, C.; Ajjan, S.; Drujon, T.; Rajavelu, A.;

Ceccaldi, A.; Jurkowska, R.; Lequin, O.; Nelson, W. G.; Guy, A.; Jeltsch, A.; Guianvarc'h,

D.; Ferroud, C.; Arimondo, P. B., Rapid synthesis of new DNMT inhibitors derivatives of

procainamide. Chembiochem 2012, 13 (1), 157-65.

31. Rubin, R. A.; Modrich, P., EcoRI methylase. Physical and catalytic properties of the

homogeneous enzyme. J Biol Chem 1977, 252 (20), 7265-72.

32. Jeltsch, A.; Friedrich, T.; Roth, M., Kinetics of methylation and binding of DNA by the

EcoRV adenine-N6 methyltransferase. J Mol Biol 1998, 275 (5), 747-58.

33. Cornforth, J. W.; Reichard, S. A.; Talalay, P.; Carrell, H. L.; Glusker, J. P., Determination

of the absolute configuration at the sulfonium center of S-adenosylmethionine. Correlation

with the absolute configuration of the diastereomeric S-carboxymethyl-(S)-methionine

salts. J Am Chem Soc 1977, 99 (22), 7292-7300.

34. Li, X.; Cournoyer, J. J.; Lin, C.; O'Connor, P. B., Use of 18O labels to monitor deamidation

during protein and peptide sample processing. J Am Soc Mass Spectrom 2008, 19 (6), 855-

64.

35. Xiao, G.; Bondarenko, P. V.; Jacob, J.; Chu, G. C.; Chelius, D., 18O labeling method for

identification and quantification of succinimide in proteins. Anal Chem 2007, 79 (7), 2714-

21.

36. Terashima, I.; Koga, A.; Nagai, H., Identification of deamidation and isomerization sites

on pharmaceutical recombinant antibody using H(2)(18)O. Anal Biochem 2007, 368 (1),

49-60.

Page 37: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

18

37. Ye, X.; Luke, B.; Andresson, T.; Blonder, J., 18O stable isotope labeling in MS-based

proteomics. Brief Funct Genomic Proteomic 2009, 8 (2), 136-44.

38. Liu, M.; Zhang, Z.; Zang, T.; Spahr, C.; Cheetham, J.; Ren, D.; Zhou, Z. S., Discovery of

undefined protein cross-linking chemistry: a comprehensive methodology utilizing 18O-

labeling and mass spectrometry. Anal Chem 2013, 85 (12), 5900-8.

39. Struys, E. A.; Jansen, E. E.; de Meer, K.; Jakobs, C., Determination of S-

adenosylmethionine and S-adenosylhomocysteine in plasma and cerebrospinal fluid by

stable-isotope dilution tandem mass spectrometry. Clin Chem 2000, 46 (10), 1650-6.

40. Wan, W.; Zhao, G.; Al-Saad, K.; Siems, W. F.; Zhou, Z. S., Rapid screening for S-

adenosylmethionine-dependent methylation products by enzyme-transferred isotope

patterns analysis. Rapid Commun Mass Spectrom 2004, 18 (3), 319-24.

41. Peters, W.; Willnow, S.; Duisken, M.; Kleine, H.; Macherey, T.; Duncan, K. E.; Litchfield,

D. W.; Luscher, B.; Weinhold, E., Enzymatic site-specific functionalization of protein

methyltransferase substrates with alkynes for click labeling. Angew Chem Int Ed Engl 2010,

49 (30), 5170-3.

42. Lee, B. W.; Sun, H. G.; Zang, T.; Kim, B. J.; Alfaro, J. F.; Zhou, Z. S., Enzyme-catalyzed

transfer of a ketone group from an S-adenosylmethionine analogue: a tool for the functional

analysis of methyltransferases. J Am Chem Soc 2010, 132 (11), 3642-3.

43. Luo, M., Current chemical biology approaches to interrogate protein methyltransferases.

ACS Chem Biol 2012, 7 (3), 443-63.

44. Dillon, S. C.; Zhang, X.; Trievel, R. C.; Cheng, X., The SET-domain protein superfamily:

protein lysine methyltransferases. Genome Biol 2005, 6 (8), 227.

45. Gui, S.; Wooderchak-Donahue, W. L.; Zang, T.; Chen, D.; Daly, M. P.; Zhou, Z. S.; Hevel,

J. M., Substrate-induced control of product formation by protein arginine

methyltransferase 1. Biochemistry 2013, 52 (1), 199-209.

46. Broom, A. D., Rational design of enzyme inhibitors: multisubstrate analogue inhibitors. J

Med Chem 1989, 32 (1), 2-7.

47. Le Calvez, P. B.; Scott, C. J.; Migaud, M. E., Multisubstrate adduct inhibitors: drug design

and biological tools. J Enzyme Inhib Med Chem 2009, 24 (6), 1291-318.

48. Coward, J. K.; Pegg, A. E., Specific multisubstrate adduct inhibitors of

aminopropyltransferases and their effect on polyamine biosynthesis in cultured cells. Adv

Enzyme Regul 1987, 26, 107-13.

Page 38: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

19

49. Suwal, S.; Pflum, M. K., Phosphorylation-dependent kinase-substrate cross-linking.

Angew Chem Int Ed Engl 2010, 49 (9), 1627-30.

50. Suwal, S.; Senevirathne, C.; Garre, S.; Pflum, M. K., Structural analysis of ATP analogues

compatible with kinase-catalyzed labeling. Bioconjug Chem 2012, 23 (12), 2386-91.

51. Gower, C. M.; Chang, M. E.; Maly, D. J., Bivalent inhibitors of protein kinases. Crit Rev

Biochem Mol Biol 2014, 49 (2), 102-15.

52. Brandvold, K. R.; Santos, S.; Breen, M. E.; Lachacz, E. J.; Steffey, M. E.; Soellner, M. B.,

Exquisitely Specific Bisubstrate Inhibitors of c-Src Kinase. ACS Chem Biol 2015.

53. Osborne, T.; Roska, R. L.; Rajski, S. R.; Thompson, P. R., In situ generation of a bisubstrate

analogue for protein arginine methyltransferase 1. J Am Chem Soc 2008, 130 (14), 4574-5.

54. Zhang, C.; Weller, R. L.; Thorson, J. S.; Rajski, S. R., Natural product diversification using

a non-natural cofactor analogue of S-adenosyl-L-methionine. J Am Chem Soc 2006, 128

(9), 2760-1.

Page 39: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

20

Chapter 2: Affinity Capturing Substrates via in situ Formation of Bisubstrate-Adducts

This chapter is based on a manuscript with the title of “Capturing Unknown Substrates via

in situ Formation of Tightly Bound Bisubstrate-adducts: S-Adenosyl-Vinthionine as a Functional

Probe for S-Adenosyl-Methionine-Dependent Methyltransferases” with the authors list: Wanlu Qu,

Kun Zhang, Michael Pablo, James Glick, Yuan Xiu, Nathaniel Kenton, R. I. Duclos, Jr, and

Zhaohui Sunny Zhou

Co-authors’ works in this chapter: Wanlu Qu: experimental design and performance, data

analysis, manuscript writing and revision; Kun Zhang, kinetics study; Michael Pablo, AdoVin

purification and revision during paper writing; James Glick, idea contributions to mass

spectrometric analysis; Yuan Xiu and Nathaniel Kenton, synthesis and characterization of

vinthionine; R. I. Duclos, Jr, synthesis and characterization of 2-amino-5-mercaptobenzoic acid;

Zhaohui Sunny Zhou, principal investigator.

Page 40: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

21

2.1 Introduction

2.1.1 S-Adenosyl-methionine Synthetase

AdoMet is the second most widely used enzyme substrate, following adenosine 5’-

triphosphate (ATP).1 One of enzymatic ways to synthesize AdoMet is transferring adenosyl group

from ATP to sulfur of methionine catalyzed by L-methionine S-adenosyltransferase (S-adenosyl-

methionine synthetase, MAT, EC 2.5.1.6), as shown in Scheme 2.1.

Scheme 2.1. MAT catalyzed AdoMet synthesis stoichiometrically from ATP and methionine.

Many analogues of methionine have behaved as possible substrates or inhibitors of MAT,2

for example, ethionine, a methionine analogue, is the inhibitor to the rat liver MAT. Many efforts

have been taken to synthesize more AdoMet analogues (as substrates of MTases), by using the

corresponding methionine analogues,3 even in cellular level.4

In my work, MAT was used to catalyze the reaction between vinthionine and ATP, and the

subsequent product, AdoVin forms a bisubstrate-adduct with the nucleophilic substrates of

MTases. There are two advantages of using methionine analogues to enzymatically synthesize

AdoMet analogues: firstly, methionine analogues are more stable than AdoMet analogues during

Page 41: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

22

synthesis and purification;5-7 and secondly, methionine analogues are readily absorbed by cellular

membranes and thus are accessible for in vivo studies, while AdoMet analogues are not.

We used MAT from archaeon Methanococcus jannaschii, and the gene encoding MAT

was identified as MJ1208, in which the majority of the sequence is highly diverse from those of

bacterial and eukaryal MAT proteins. With respect to catalytic activities, archaeon MAT shares

most of functional properties with bacterial or eukaryal MAT. For instance, Mg2+ and K+ are

essential to their high catalytic activity. In addition, all the MATs catalyze two-step reactions, that

is, AdoMet formation and tripolyphosphate (PPPi) cleavage and release. One significant difference

between archaeon MAT and bacterial or eukaryal MAT is that archaeon MAT is quite thermally

stable, and its functional properties were evaluated at 70 °C.8

Aside from more stability of vinthionine than AdoVin and accessibility of vinthionine for

in vivo studies, formation of AdoVin bisubstrate-adduct via enzyme MAT-MTases coupled

reactions is advantageous with another reaction: the detection of AdoVin and AdoVin adduct can

be facilitated by introduction of stable isotope labeling.

Scheme 2.2. Formation of isotope labeled bisubstrate-adduct via isotope labeled ATP.

Page 42: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

23

As shown in Scheme 2.2, isotope labeled ATP can be used to generate AdoVin with stable

isotope labeling catalyzed by MAT, and subsequently, AdoVin bisubstrate-adduct will be isotope

labeled due to the catalysis of MTases. Combining with mass spectrometry, isotope labeled

AdoVin and AdoVin bisubstrate-adduct will be observed with +15 mass shift, but sharing identical

LC retention time, ionization efficiency and fragmentation patterns.

One limitation of mass spectrometry is that unionized substrates or methylation products

cannot be detected in mass spectrometry, most commonly with small molecule substrates or

products, like catechol, the substrate of catechol O-MTase. It is more accessible to detect AdoVin

and its bisubstrate-adduct by mass spectrometry, because they are always positively charged at the

LC-MS conditions (mildly acidic).

Page 43: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

24

2.2 Results and Discussions

2.2.1 Enzymatic Synthesis of S-adenosyl-vinthionine

Synthesis of Vinthionine

Vinthionine was prepared by organic synthesis (Scheme 2.3), and was characterized by

using 1H-NMR and 13C-NMR. Purified vinthionine was dissolved in Milli Q water and subjected

to LC-MS for further verification. Figure 2.1 shows that vinthionine was observed as singly

charged (m/z 162.0), and the detailed fragmentation of vinthionine under CID was shown in Figure

2.2.

Scheme 2.3. Synthesis of vinthionine (vinyl homocysteine).

Page 44: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

25

Figure 2.1. Mass spectrum of vinthionine (expected m/z 162.05 for vinthionine M+H+, observed

m/z 162.07; mass difference 0.02 Da).

Figure 2.2. CID MS/MS spectrum of vinthionine from LC- ESI/MS. The +1 precursor ion for

MS/MS analysis was m/z 162.07.

Page 45: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

26

Enzymatic Synthesis of S-adenosyl-vinthionine

As shown in Scheme 2.4, AdoVin was synthesized stoichiometrically from ATP and

vinthionine catalyzed by L-methionine S-adenosyltransferase (AdoMet synthetase, MAT, EC

2.5.1.6). With similar structure with AdoMet, AdoVin shares similar characteristics with AdoMet.

First of all, AdoVin has the same UV absorbance with AdoMet at 260 nm (Figure 2.4), due to the

same UV active adenosyl group (ε260 nm = 15,400 M-1 cm-1).9 Another significant characteristic of

AdoVin is that the permanent positive charge from the sulfonium ion makes both AdoMet and

AdoVin very hydrophilic, and hardly retained on C18 column, even though AdoVin is a little more

hydrophobic than AdoMet replacing methyl group by vinyl group, as Figure 2.3 shown.

Scheme 2.4. Synthesis of S-adenosyl-vinthionine (AdoVin) catalyzed by MAT.

Page 46: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

27

Figure 2.3. Reverse-phase HPLC chromatogram (monitored at 260 nm) of AdoVin synthesis with

(top trace) or without (middle trace) MAT.

Figure 2.4. UV/Vis spectra of synthesized AdoVin (blue) and AdoMet (red).

Page 47: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

28

Instead of reverse phase HPLC, strong cation exchange (SCX) is a good method to separate

positively charged AdoVin from other species in the reaction mixture at pH 3.0, such as negatively

charged ATP and neutral vinthionine. Indeed, AdoVin binds with SCX column more strongly than

C18 column, and it retained much longer than ATP and vinthionine. As shown in Figure 2.3 and

Figure 2.5, AdoVin was produced only when MAT was provided, indicating that formation of

AdoVin is MAT-dependent.

Figure 2.5. Strong cation exchange (SCX) HPLC chromatogram (monitored at 260 nm) showing

the MAT-catalyzed synthesis of AdoVin and AdoMet; a) MAT-catalyzed AdoVin synthesis; b)

MAT-catalyzed AdoMet synthesis.

Page 48: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

29

Characterization of Enzymatic Synthesis of AdoVin by MS

Since all the compounds containing adenine group have the maximum absorbance at 260

nm in the reaction mixture, we need further verification of the newly found peak as AdoVin. Due

to the instability of AdoVin, similar with AdoMet,5-7 purification of AdoVin is not practical, thus

NMR is not the appropriate tool for the characterization. Instead, LC-MS was used to separate and

then verify the synthesized AdoVin in the reaction mixture. Linear ion trap mass spectrometer was

used to detect AdoVin synthesized from MAT catalyzed reactions. In order to maintain the

cleanness of MS, volatile buffer salts NH4HCO3 was used for AdoVin synthesis at pH 8.0. MAT

was precipitated by using 100% trichloroacetic acid (TCA) prior to the injection into LC-MS

system. AdoVin was observed as m/z 411.07 with its single positive charge (Figure 2.6-2.7). CID

MS/MS spectrum matches the theoretical prediction of AdoVin fragmentation, and adenosyl

fragment is the most abundant species (Figure 2.8).

Figure 2.6. Extracted ion chromatogram of m/z 411.0 (expected for AdoVin).

Page 49: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

30

Figure 2.7. Mass spectrum of MAT-catalyzed AdoVin synthesis (expected m/z 411.10 for AdoVin

M+, observed m/z 411.07; mass difference 0.03 Da).

Figure 2.8. CID MS/MS spectrum of AdoVin from LC-ESI/MS. The +1 precursor ion for MS/MS

analysis was m/z 411.07.

Page 50: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

31

2.2.2 in situ formation of AdoVin Adduct Catalyzed by Methyltransferases

Formation and Characterization of AdoVin-TNB Adduct

In order to test the activity of AdoVin, thiopurine methyltransferase (TPMT, EC 2.1.1.67)

was selected for two reasons: first, TPMT has a broad substrate specificity to aromatic thiols,10-13

which we anticipated would show high activity to the vinyl group of AdoVin, based on Michael

addition.14 Second, some of the aromatic thiol substrates and their methyl products or alkyl

products have strong UV absorbance above 260 nm, therefore, the expected adduct between

AdoVin and the aromatic thiols would have a signature UV absorbance.

Reduced Ellman’s reagent, 5-thio-2-nitrobenzoic acid (TNB) was used as the substrate to

test the activity of AdoVin catalyzed by TPMT.15 The maximum UV absorbance of TNB is at 325

nm at pH 2-3, while the UV absorbance of methyl-TNB shifts to 350 nm.16 Our theoretical

prediction of the UV spectrum for AdoVin-TNB is maximal at both 260 nm and the range 325-

350 nm (Scheme 2.5).

Page 51: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

32

Scheme 2.5. TPMT catalyzed methylation of TNB (top) and formation of bisubstrate-adduct

between AdoVin and TNB (bottom).

In our experiments, AdoVin was synthesized from vinthionine and ATP catalyzed by MAT

first, and then an AdoVin-TNB adduct was formed in situ, catalyzed by TPMT. A negative control

without MAT (AdoVin) was included in the investigation. As Figure 2.9 shows, there was a peak

after 1 hour incubation in the reaction mixture, while not in the negative control. Moreover, the

UV absorbance of the new peak matches the expected combination of both the UV active

chromophores, adenosyl group (260 nm)9 and alkyl-TNB (330 nm),16 and the ratio between 260

nm and 330 nm are very close to the estimated ratio of AdoVin-TNB adduct (Figure 2.10, Table

2.1). Therefore, the new peak was estimated as the AdoVin-TNB adduct.

Page 52: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

33

Figure 2.9. HPLC chromatogram (325 nm) of AdoVin-TNB reaction catalyzed by MAT-TPMT

(top), and AdoVin-TNB reaction mixture without MAT (bottom).

Page 53: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

34

Figure 2.10. UV/Vis absorbance spectra of adenosine (red), methylated TNB (blue) and AdoVin-

TNB adduct (black).

Page 54: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

35

Table 2.1. Calculation of extinction coefficient of the AdoVin-TNB Adduct.

Extinction Coefficient

(ε, M-1 cm-1)

ε 260 nm ε 350 nm

Adenosine 15,4009 0

Methyl-TNB 5,25416 10,83516

AdoVin-TNB Adduct, Calculated 20,654 10,835

Calculated Ratio (260 nm: 350 nm)

For the AdoVin-TNB Adduct

20,514/10,835 = 1.91

Observed Ratio (260 nm: 350 nm)

For the AdoVin-TNB Adduct

392/198 = 1.98

To further verify the enzymatically synthesized AdoVin-TNB adduct, LC-MS was used.

A species was observed with single positive charge (610.0 m/z). AdoVin-TNB was only observed

in reaction mixture, not in the negative control “No TPMT” in which the region around 7.1 min

(where the adduct eluted) was enlarged 50 times (Figure 2.11 and 2.12). This observation indicated

that AdoVin-TNB adduct formation is TPMT dependent, and the subsequent CID MS/MS

fragmentation of m/z 610.27 matches the theoretical prediction of AdoVin-TNB adduct (Figure

2.13).

Page 55: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

36

Figure 2.11. Extracted ion chromatogram of m/z 411.0 (expected AdoVin), and 610.0 (expected

AdoVin-TNB adduct) in AdoVin-TNB reaction (top), and AdoVin-TNB reaction without TPMT

(bottom).

Page 56: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

37

Figure 2.12. Mass spectrum of AdoVin-TNB adduct (expected m/z 610.14 for AdoVin-TNB

adduct M+, observed 610.27; mass difference 0.13 Da).

Figure 2.13. CID MS/MS spectrum of AdoVin-TNB adduct from LC-ESI/MS. The +1 precursor

ion for MS/MS analysis was m/z 610.07.

Page 57: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

38

2.2.3 Substrate Specificity toward AdoVin

When designing enzyme substrate analogues, the first priority is for enzyme specificity,

that is, whether AdoMet analogues have the similar substrate specificity as AdoMet. To this end,

we studied substrate specificity towards AdoVin by testing other known substrates of TPMT, and

the structures of the thiol substrates are included in Table 2.2. Using HPLC-UV and LC-MS, we

observed that all the tested known substrates reacted with AdoVin to form bisubstrate-adducts

catalyzed by TPMT. For example, bisubstrate-adducts between AdoVin and 2-Bromothiophenol

and 3-Bromothiophenol were observed in the MAT-TPMT coupled reactions, but not in the

negative control (Figure 2.14 and Figure 2.15, c and d in Table 2.2). In the meanwhile, no adduct

was observed between AdoVin and the non-substrate toward AdoMet, 4-nitrophenol (g in Table

2.2), indicating that 4-nitrophenol did not show activity to AdoVin, either (Figure 2.16). These

observations indicate that the substrate specificity of TPMT toward AdoVin and AdoMet mirrors

each other.

Table 2.2. Similar substrate specificity toward AdoVin and AdoMet

Page 58: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

39

Figure 2.14. Selected ion chromatography of m/z 599.0 (theoretical AdoVin-BrPhSH adduct, m/z

599.07) in AdoVin-2-Bromothiophenol (2BrPhSH, c) and AdoVin-3-Bromothiophenol (3BrPhSH,

d) reactions. The adducts between AdoVin and 2BrPhSH and 3BrPhSH were observed at 13.5 and

14.3 min, respectively.

Page 59: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

40

Figure 2.15. Extracted ion chromatogram of m/z 599.0 (theoretical AdoVin-BrPhSH adduct, m/z

599.07) in AdoVin-2BrPhSH reaction (top) and negative control “No 2BrPhSH” (bottom).

Page 60: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

41

Figure 2.16. HPLC chromatogram (325 nm) of in vitro AdoVin-Nitrophenol (g) reactions (top)

and negative control at 3 hours incubation (bottom). No adduct was not observed, indicating that

no reaction occurred between AdoVin and 4-nitrophenol (non-substrate of TPMT).

Page 61: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

42

2.2.4 Tight Binding between Bisubstrate-Adduct and Methyltransferases

Taking the concept of bisubstrate-adduct inhibition into consideration, a synergistic

interaction between the bisubstrate-adduct and MTases is expected. To investigate the binding

strength of the AdoVin adduct for methyltransferases, ultrafiltration was performed to separate

TPMT and other large proteins from small molecules (free ligands) in the reaction mixture by

molecular weight (MW of TPMT is 30,343; MWCO of filters is 10,000). Immobilized metal ion

affinity chromatography (IMAC) was also applied to isolate TPMT from other components, since

our recombinant TPMT contains a hexa-histidine tag.

After isolation of TPMT using both methods, TPMT and other large proteins were

precipitated using 100% TCA, and the supernatant was injected into LC-MS. Under both

ultrafiltration and IMAC conditions, AdoVin-TNB adduct was observed only in the enzyme

complex (Table 2.3 and Figure 2.17), indicating significantly strong binding between AdoVin-

TNB adduct and TPMT. While other small molecules were only observed in free ligands fraction,

but not in TPMT complex fraction. This suggests markedly weak binding or no interaction between

other small molecules and TPMT, including the non-natural substrates, vinthionine and AdoVin

(Table 2.3 and Figure 2.18). The adducts between AdoVin and other thiol substrates of TPMT, for

example, AdoVin-2-Bromothiophenol and AdoVin-3-Bromothiophenol, were also tested for the

tight binding event (Figure 2.19-2.20). All the AdoVin-thiols adducts were observed tightly bound

with TPMT, suggesting that the tight binding event is the common property of AdoVin adducts.

Page 62: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

43

Table 2.3. Main components observed in the fractions of isolated TPMT and free ligands

Isolated TPMT fraction Free ligands fraction

AdoVin-thiol bisubstrate-adduct

Vinthionine

ATP

TNB

AdoVin

Figure 2.17. HPLC chromatogram of isolated TPMT fraction (top) and free ligands fraction

(bottom) with UV detection at 325 nm.

Page 63: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

44

Figure 2.18. Extracted ion chromatogram of m/z 162.07 (theoretical vinthionine, m/z 162.05) and

610.14 (theoretical AdoVin-TNB adduct, m/z 610.27) in isolated TPMT fraction (top) and free

ligands fraction (bottom).

Page 64: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

45

Figure 2.19. Selected ion chromatogram of m/z 599.0 (theoretical AdoVin-BrPhSH adduct, m/z

599.07) for the binding assay between AdoVin-BrPhSH adduct and TPMT. The intensity of peaks

between 13 and 14 min in “free ligands b)” fraction is over 50 fold less than that of AdoVin-

2BrPhSH and AdoVin-3BrPhSH adducts in “TPMT complex a)”, indicating that the no AdoVin-

BrPhSH adduct was detected in “free ligands b)”.

Page 65: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

46

Figure 2.20. CID MS/MS spectra of AdoVin-2BrPhSH adduct (top) and AdoVin-3BrPhSH adduct

(bottom) from LC- ESI/MS. The +1 precursor ions for MS/MS analysis were m/z 598.91 and

598.97, respectively.

Page 66: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

47

2.2.5 Kinetics of Methyltransferases-catalyzed AdoVin Adduct Formation

To further investigate the mechanism of TPMT catalyzed AdoVin bisubstrate-adduct

formation, kinetic studies of TPMT catalyzed AdoVin-TNB adduct formation were performed,

and the concentration change of TNB with time were monitored at 411 nm at pH 8.0. As shown in

Figure 2.21, AdoVin-TNB adduct formation is both time-dependent (first-order kinetics) and

enzyme concentration dependent. This is consistent with the first order reaction mechanism,

involving a rapid initial binding of the thiol substrate and AdoVin with TPMT and then a

subsequent addition reaction between the thiol substrate and AdoVin. Better still, only a single

turnover formation of the bisubstrate-adduct was observed (Figure 2.22), suggesting that the

resulting adduct binds much more tightly than either substrate, and thus inhibits the enzyme from

further turnover. This observation is also consistent with the enhanced binding affinity by the

bisubstrate-adduct compared to the individual substrates.

For the data processing, the concentration change of TNB was fitted to the first order

equation: y = m1 + m2*e^(-m3x), and the calculated “m3” is the “turnover number”, kcat, for

TPMT catalyzed AdoVin-TNB, and the half time, t1/2 is thus calculated by using equation t1/2 =

0.693/ kcat. The fitting parameters were shown in Table 2.4.

Page 67: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

48

Figure 2.21. Absorbance change of TNB at 411 nm of AdoVin-TNB reactions catalyzed by TPMT

under various concentrations

Page 68: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

49

Figure 2.22. The overall concentration change of TNB at 411 nm at different concentration of

TPMT, indicating a single turnover of the reaction.

Page 69: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

50

Table 2.4. Fitting parameters of first order kinetics

5 µM TPMT 10 µM TPMT 20 µM TPMT

Fitting equation y = m1 + m2*e^(-m3*x)

m1 -0.0602 -0.111 -0.212

m2 0.0635 0.113 0.220

m3 0.490 0.298 0.201

R2 0.847 0.997 0.998

t1/2

(min) = 0.693/ kcat

1.4 2.3 3.4

average of t1/2

(min) 2.4

standard deviation ơ of t1/2

(min) 0.7

kcat

(min-1

) = m3 0.490 0.298 0.201

average of kcat

(min-1

) 0.33

standard deviation ơ of kcat

(min-1

) 0.01

Page 70: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

51

2.2.6 Discovery and Identification of Unknown Substrates of Methyltransferases

2.2.6.1 Discovery of Unknown Adduct

One of our goals is to identify unknown substrates of MTases; in particular, no endogenous

substrate for TPMT has been reported. To this end, AdoVin was incubated with the crude cell

lysate of the E. coli strains that expressed recombinant human TPMT, but no AdoVin adduct was

detected (Figure 2.23 and 2.26), which was not unexpected as no aromatic thiol metabolites have

been reported for E. coli.

As a positive control, TNB was added to the E. coli crude cell lysate, and the corresponding

adduct was detected only in the enzyme complex (see Figure 2.23 and 2.24), indicating tight

binding under physiological conditions as well. Furthermore, Figure 2.24 illustrates the significant

enrichment of the adducts from very complex cellular mixtures, thereby making it possible to

readily identify the unknown substrate.

Interestingly and unexpectedly, besides the AdoVin-TNB adduct (m/z 610.0), another

adduct (m/z 580.0 in Figure 2.23 and 2.26) was detected in the TPMT complex, but only when

TNB was added to cell lysate, suggesting this unknown peak was derived from TNB. Isotope

labeled AdoVin (+15 Da shift) was introduced to verify the unknown peak. Based on the mass

shift (+15 Da) and its UV-Vis spectrum (Figure 2.25), it could be inferred that the unknown

contains both AdoVin (260 nm) and thiol moieties (330 nm). Isotope labeling was used to verify

the unknown adduct. Furthermore, based on the MSMS fragmentation (Figure 2.27) and the mass

change between the unknown adduct and AdoVin-TNB adduct (-30 Da), we postulated that the

nitro-group was reduced to an amine, with the knowledge of the existence of at least four

nitroreductases with broad specificities in E. coli.17-22 Further verification will be discussed in the

next section.

Page 71: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

52

Figure 2.23. Selected ion chromatogram of AdoVin adducts (m/z 610.0 and 580.0) in isolated

TPMT fractions from crude cell lysates of ex vivo AdoVin-TNB and negative control (without

exogenous thiols).

Page 72: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

53

Figure 2.24. HPLC chromatogram (260 nm) of ex vivo AdoVin-TNB reaction at 1 hour (top),

isolated TPMT fraction (middle), and free ligands fraction (bottom), showing the adducts tightly

bound with TPMT complex in ex vivo AdoVin-TNB reactions.

Page 73: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

54

Figure 2.25. UV-Vis spectra of AdoVin-TNB adduct (black) and unknown adduct (red).

Figure 2.26. Extracted ion chromatogram (XIC) of m/z 610.0 and 580.0 in TPMT fraction of ex

vivo AdoVin-TNB (top) and ex vivo “No thiols” negative control (bottom), indicating that

unknown adduct m/z 580.0 was found, and also no AdoVin adduct was observed without

exogenous thiols.

Page 74: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

55

Figure 2.27. CID MS/MS spectra of AdoVin-TNB adduct, regular and isotope labeled unknown

adduct from LC-ESI/MS. The +1 precursor ions for MS/MS analysis were m/z 610.01, 580.08 and

595.08, respectively.

Page 75: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

56

2.2.6.2 Elucidation and Confirmation of Unknown Adduct

Synthesis of 2-Amino-5-Mercaptobenzoic Acid

We hypothesized that the nitro group of the AdoVin-TNB adduct was reduced to amine

group, and postulated the unknown thiol to be 2-amino-5-mercaptobenzoic acid (Scheme 2.6). In

order to confirm this hypothesis, authentic 2-amino-5-mercaptobenzoic acid (AMBA) was

synthesized, and the reaction between AMBA and AdoVin was investigated. First, the disulfide

form, 5,5’-dithio-bis-(2-aminobenzoic acid), was synthesized from the disulfide form of TNB

(5,5’-dithio-bis-(2-aminobenzoic acid), DTNB), and then AMBA was synthesized from its

disulfide form by the reduction of tris(2-carboxyethyl)phosphine (TCEP).

Scheme 2.6. Synthesis of 2-amino-5-mercaptobenzoic acid.

Page 76: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

57

in vitro Bisubstrate-adduct Formation between AdoVin and AMBA

Next, the reaction between authentic AMBA and AdoVin was performed under the

catalysis of TPMT (Scheme 2.7). It was expected that AMBA would react with AdoVin to form a

bisubstrate-adduct. The co-elution of the in vitro formed AdoVin-AMBA adduct and the ex vivo

unknown adduct indicates that they are the same compound (Figure 2.28), which was also verified

by overlying their UV spectra (Figure 2.29) and their mass spectra with isotope labeled AdoVin-

AMBA adduct (Figure 2.30). The CID MSMS fragmentation of unknown adduct was then

assigned as AdoVin-AMBA adduct, sharing similar fragmentation with AdoVin-TNB adduct

(Figure 2.31).

Scheme 2.7. in vitro bisubstrate-adduct formation between AdoVin and AMBA catalyzed by

thiopurine S-methyltransferase.

Page 77: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

58

Figure 2.28. HPLC chromatogram (350 nm) of authentic AdoVin-AMBA adduct (top), co-

injection of ex vivo AdoVin-TNB reaction and authentic AdoVin-AMBA adduct (middle), and ex

vivo AdoVin-TNB reaction (bottom).

Page 78: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

59

Figure 2.29. UV spectra of ex vivo AdoVin-AMBA adduct (black) and authentic AdoVin-AMBA

adduct standard (red).

Figure 2.30. Extracted ion chromatogram of m/z 580.08 (AdoVin-AMBA adduct) and 595.06

(isotope labeled) in isolated TPMT fraction (top) and free ligands fraction (bottom).

Page 79: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

60

Figure 2.31. CID MS/MS spectra of AdoVin-TNB adduct, regular and isotope labeled AdoVin-

AMBA from LC-ESI/MS. The +1 precursor ion for MS/MS analysis was m/z 610.01, 580.08 and

595.06, respectively.

Page 80: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

61

2.2.6.3 Confirmation of 2-Amino-5-Mercaptobenzoic Acid as TPMT Substrate

It is worth noting that this amino thiol (AMBA) had not been reported as a substrate of

TPMT, so next, methylation of AMBA was performed to characterize AMBA as the substrate of

TPMT (Scheme 2.8). A new peak was observed eluting after AMBA in C18 column, and only

occurred when TPMT was used (Figure 2.32). Together with its UV spectrum (Figure 2.33), the

new peak was confirmed as methylation product of AMBA. Thereby, AMBA was confirmed as a

substrate of methylation. Altogether, this serendipitous finding underscores the utility of our

approach in directly identifying enzyme substrates, even unknowns.

Scheme 2.8. Proposed methylation of 2-amino-5-mercaptobenzoic acid catalyzed by thiopurine

methyltransferase.

Page 81: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

62

Figure 2.32. HPLC chromatogram (350 nm) of TPMT catalyzed methylation of 2-amino-5-

mercaptobenzoic acid (top), negative control for AMBA methylation without TPMT (middle), and

co-injection of AMBA methylation and authentic AMBA standard (bottom).

Page 82: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

63

Figure 2.33. UV spectra of methylated AMBA (black) and AMBA (red). The spectra were

normalized at 260 nm (AMBA) and 270 nm (Me-AMBA).

Page 83: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

64

2.2.7 No Modification of Enzymes by AdoVin

One of the general concerns of AdoMet analogues is whether the analogue modifies

enzymes. MALDI-TOF/TOF was performed to monitor the potential background adduct

formation between AdoVin and proteins. As shown in Figure 2.34 and 2.35, no modification was

detected on either TPMT or MAT. While the vinyl sulfonium group is intrinsically reactive,

extensive solvation in aqueous solution renders low reactivity of highly charged sulfoniums. For

instance, no background reaction between aromatic thiols and AdoVin was observed in the absence

of TPMT, and moreover, even under ex vivo conditions where many metabolites exist, no adducts

were detected unless exogenous thiols were provided.

Page 84: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

65

Figure 2.34. MALDI mass spectra of native MAT (top) and treatment with AdoVin (bottom). The

molecular weight of native MAT is 48019.0. Under the instrument error range, no mass shift (+

411.1 Da) was observed, indicating that no modification with AdoVin was observed.

Page 85: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

66

Figure 2.35. MALDI mass spectra of native TPMT (top) and treatment with AdoVin (bottom). The

molecular weight of native TPMT is 30343.6. Under the instrument error range, no mass shift (+

411.1 Da) was observed, indicating that no modification with AdoVin was observed.

Page 86: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

67

2.3 Conclusions

In sum, our strategy can indeed capture and identify enzyme substrates, even unknowns.

Conversely, if a substrate or methylation product is known, the corresponding enzyme can be

identified as well. Applications in whole cells and organisms can also be envisioned, as AdoVin

can be synthesized in vivo when vinthionine is supplemented. Moreover, the formation of

bisubstrate-adducts may also have broad utility in facile generation of strong specific inhibitors

and structural elucidation of substrate-enzyme interactions. Lastly, our approach can be applied to

many enzymes in other classes, particularly those catalyzing group transfers.

Page 87: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

68

2.4 Experimental Section

2.4.1 General Procedures

All chemicals with reagent purity or above were purchased from Sigma and Fisher. A 100

mM Adenosine-13C10,15N5 5′-triphosphate sodium salt solution (+15 ATP) in 5 mM Tris buffer

was purchased from Sigma (catalog No. 645702). Immobilized metal ion affinity chromatography

was performed on HisTrap HP columns (GE Healthcare Life Sciences, catalog No. 17-52447-01),

and the HisPur Ni-NTA Spin Column with 0.2 mL resin bed (ThermoScientific Pierce, catalog No.

12393730), respectively. Ultrafiltration was carried out using filters with 30,000 molecular weight

cut off (MWCO). The filters were purchased from Fisher Scientific (EMD Millipore Amicon Ultra

0.5 mL, catalog No. UFC50VL96).

UV absorbances were measured on a Cary 50 Bio UV/VIS spectrophotometer. HPLC

chromatograms were acquired on an Agilent 1100 HPLC with Dynamax SD-200 pumps and a

Varian ProStar 330 photodiode array detector interfaced with the Varian ProStar Chromatography

Workstation software version 6.41. Mass spectral data were acquired on a Thermo LCQ Deca XP

Ion trap mass spectrometer in lined with an Agilent 1200 HPLC, and processed by using Xcalibar

Data System 2.0.7 (Thermo Fisher Scientific Inc. Waltham, MA). Graphics were constructed by

using the Kaleidagraph software package 4.1 (Synergy Software, Reading, PA).

Page 88: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

69

2.4.2 Expression and Purification of Enzymes

Recombinant Thiopurine S-Methyltransferase (TPMT)

Recombinant human TPMT was prepared as described previously.23 The human thiopurine

S-methyltransferase gene was ligated into pET28a vector (Novagen, Madison, WI) between NdeI

and EcoRI sites. The amino acid sequence of the N-terminal histidine tag prior to the original

starting methionine is MGSSHHHHHH SSGLVPRGSH. The recombinant plasmid, TPMT7A,

was transformed into Escherichia coli strain BL21 (DE3) (Novagen) for protein expression. A

starter culture of 10 mL in Luria-Bertani broth (LB) with 50 μg/mL kanamycin was initiated with

a single transformed colony and grown overnight at 37 °C in a shaker (50 rpm) to reach an OD600

reading around 0.6. The BL21 (DE3)/TPMT7A cells were then transferred to 1 L LB broth with

50 μg/mL kanamycin and grown aerobically at 37 °C in a shaker (50 rpm) to an OD600 reading of

0.6. Then, isopropyl-β-thiogalactopyranoside (IPTG) was added to a final concentration of 1 mM

to induce protein expression. Cells were harvested 8 h post induction by centrifugation (4500 g,

4 °C, and 20 minutes) and resuspended in a cold solution containing 20 mM potassium phosphate

and 0.5 M NaCl at pH 8.0. Cells were lysed via sonication on ice, and the cell debris and unbroken

cells were removed by centrifugation (8000 g, 4 °C, and 60 minutes).

The purification procedure was based on the protocol from the manufacturer. A 5 mL

HiTrap nickel-chelate column (GE Healthcare) was preconditioned with 25 mL MilliQ water, 5

mL 0.1 M Nickel Sulfate (filtered by 0.2 µm membrane), 25 mL MilliQ water, 30 mL binding

buffer (20 mM potassium phosphate, 0.5 M NaCl and 5 mM imidazole, pH 8.0, filtered by 0.2 µm

membrane). After column conditioning, the supernatants from TPMT lysate were loaded onto the

column, equilibrated with 50 mL binding buffer, and washed with 25 mL washing buffer (20 mM

potassium phosphate, 0.5 M NaCl and 50 mM imidazole, pH 8.0, filtered by 0.2 µm membrane).

Page 89: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

70

The TPMT protein was eluted with 25 mL elution buffer (20 mM potassium phosphate, 0.5 M

NaCl and 500 mM imidazole, pH 8.0, filtered by 0.2 µm membrane). Stripping buffer (25 mL, 20

mM potassium phosphate, 0.5 M NaCl and 500 mM EDTA, pH 8.0, filtered by 0.2 µm membrane)

was used to strip the remaining proteins on the column. The protein solution was dialyzed in 50

mM potassium phosphate at pH 8.0 containing 2 mM DTT. The purified TPMT solutions were

mixed with glycerol (10%, v/v) and stored at -80 °C.

Protein purity was analyzed by electrophoresis in 12% polyacrylamide gels in the presence

of sodium dodecyl sulfate (SDS-PAGE) and visualized by Coomassie blue staining. The molecular

weight of TPMT is calculated as 34401.3 Da, and the concentration of TPMT is determined by

UV absorbance at 280 nm, using the estimated extinction coefficient ε280 nm = 39,420 M-1cm-1.

Recombinant Methionine S-Adenosyltransferase (MAT)

The MJ1208 gene was amplified from M. jannaschii genomic DNA by polymerase chain

reaction using the primers MJ1208FWD (GCATATGAGAAACATAATTGTAAAAAAATTAG)

and MJ1208REV (GGATCCTTAGAATGTAGTTACTTTTCC). The NdeI and BamHI sites

introduced into the gene through the primers were used to clone the DNA into the pET-19b vector

(Novagen Inc.). E. coli BL21 (DE3) cells were transformed with plasmid (pMJ1208-1) for protein

expression.8 Ampicillin (100 mg/ml) was used as the antibiotic for MAT expression. The detailed

expression and purification procedure is similarly described in TPMT section. The molecular

weight of MAT is calculated as 50427.7, and the concentration of MAT is determined by UV

absorbance at 280 nm using the estimated extinction coefficient ε280 nm = 24,870 M-1cm-1.

Page 90: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

71

Recombinant E. coli 5'-methylthioadenosine/S-adenosyl-homocysteine nucleosidase

An EcoRI/NotI fragment from p5Xmtan (Cornell & Riscoe, 1998) containing the complete

E. coli MTA/AdoHcy nucleosidase gene (accession No. U24438) was ligated into EcoRI/NotI-

digested pPROEX HTa expression vector (Gibco BRL) and transformed into E. coli strain

TOP10F’. The expressed enzyme contains a 31-residue N-terminal tag consisting of a six-histidine

tag, a spacer sequence and an rTEV protease cleavage site prior to the native initiating methionine

of the nucleosidase.24 Ampicillin (100 mg/ml) was used as the antibiotic for 5'-

methylthioadenosine/S-adenosyl-homocysteine nucleosidase (MTAN) expression. The detailed

expression and purification procedure is similarly described in TPMT section. The molecular

weight of MTAN is calculated as 28157.0 Da, and the concentration of MTAN is determined by

UV absorbance at 280 nm using the estimated extinction coefficient ε280 nm = 11,920 M-1cm-1.

2.4.3 Protein Precipitation

100% (w/v) trichloroacetic acid (TCA) was prepared by dissolving 5 g TCA into 3.5 mL

Milli Q water. The TCA stock solution was stored at room temperature, protected from light. To

precipitate proteins in the reaction mixtures, 1 volume TCA stock solution was added to 4 volumes

of protein containing reaction mixtures, and incubated 10 minutes at 4 °C. The protein precipitation

was then spun down at 14K rpm for 5 minutes. The supernatant was saved for further analysis.

Page 91: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

72

2.4.4 Preparation of Vinthionine

Vinthionine. [CAS: 83768-87-0] Vinthionine was prepared as reported.25 To a 250 mL,

three-necked, round-bottom flask over a dry-ice/ethanol bath, DL-homocystine (1.01 g, 7.47 mmol)

was added. The sealed system was evacuated and then purged with anhydrous nitrogen several

times. Anhydrous ammonia was then condensed into the round-bottom flask until the volume of

liquid ammonia was approximately 50 mL. Small pieces of sodium metal were added slowly with

stirring until the solution remained blue for ten minutes. Ammonium chloride was added slowly

until the blue color faded, and the solution was allowed to warm to room temperature. After all

ammonia as evaporated, the flask was cooled to 0 °C, and 15 mL of anhydrous DMSO was added

to dissolve the residue. Dried acetylene gas was then bubbled though the solution for 4 hours,

during which the flask was allowed to return room temperature. The syrup was then neutralized to

pH near 7 with 1 N HCl. The mixture was cooled to -20 °C overnight to allow crystallization

before filtering and washing with cold ethanol. The light-yellow powder was dried under vacuum.

The product was used in the following procedures without further purification. 1H NMR

(D2O/K2CO3, 400 MHz): δ 6.33 (dd, 1H, J = 16.9 Hz, 10.3 Hz), 5.24 (d, 1H, J = 10.3 Hz), 5.16 (d,

1H, J = 16.9 Hz), 3.74 (t, 1H, J = 6.2 Hz), 2.77 (t, 2H, J = 7.3 Hz), 2.10 (m, 2H);13C NMR (D2O,

100 MHz): δ 174.45, 130.68, 112.70, 53.94, 30.17, 26.42.

Page 92: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

73

Figure 2.36. 1H-NMR spectrum of vinthionine (400 MHz, D2O).

Page 93: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

74

Figure 2.37. 13C-NMRspectrum of vinthionine (100 MHz, D2O).

Page 94: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

75

2.4.5 Enzymatic Synthesis of AdoVin

S-Adenosyl-Vinthionine. [83768-89-2] The reaction contained 50 mM potassium

phosphate (pH 8.0), 5 mM KCl, 2.5 mM MgCl2, 1 mM ATP and 500 μM vinthionine, and was

initiated with 50 μM MAT and incubated at 37 °C. The same solutions without MAT was

performed as the negative control “No MAT”. The same reaction containing methionine instead

of vinthionine was performed as the positive control “AdoMet synthesis”. About 150 μM AdoVin

was synthesized after 2 hours incubation.The concentration of AdoVin was determined using ε260

nm = 15,400 M-1 cm-1 based on the value for AdoMet.9

2.4.6 in vitro Formation of AdoVin Adduct in MAT-TPMT Coupled Reactions

AdoVin-TNB Adduct Formation

The reaction contained 50 mM potassium phosphate, pH 8.0, 5 mM KCl, 2.5 mM MgCl2,

1 mM ATP, 600 μM vinthionine, 480 μM 2-nitro-5-mercaptobenzoic acid (TNB), 2 mM TCEP,

83 μM TPMT and 1.75 μM MTAN. The reaction was initiated with 120 μM MAT and incubated

at 37 °C. The same solutions without MAT or TPMT were performed as negative controls, “No

MAT” and “No TPMT.” The concentration of TNB was determined using ε 411 nm = 13,600 M-

1 cm-1.26

AdoVin-Thiophenol Adduct Formation

The reaction contained 50 mM potassium phosphate, pH 8.0, 5 mM KCl, 2.5 mM MgCl2,

1 mM ATP, 900 μM vinthionine, 510 μM thiophenol, 2 mM TCEP, 120 μM TPMT, and 1.75 μM

MTAN. The reaction was initiated with 300 μM MAT and incubated at 37 °C. HPLC-UV and LC-

Page 95: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

76

MS were used to monitor the reactions. The same solutions without MAT or TPMT were

performed as negative controls, “No MAT” and “No TPMT.”

AdoVin-Bromothiophenol Adduct Formation

The reaction contained 50 mM potassium phosphate, pH 8.0, 10 mM KCl, 5 mM MgCl2,

2 mM ATP, 900 μM vinthionine, 410 μM 2-Bromothiophenol (2BrPhSH) or 3-Bromothiophenol

(3BrPhSH), 2 mM TCEP, 120 μM TPMT, and 1.75 μM MTAN. The reaction was initiated with

300 μM MAT and incubated at 37 °C. The same reactions without MAT, TPMT and 2BrPhSH

were performed as negative controls, “No MAT”, “No TPMT” and “No 2BrPhSH,” and the

negative controls contained both 2-Bromothiophenol and 3-Bromothiophenol.

AdoVin-4-Nitrothiophenol Adduct Formation

The reaction contained 50 mM potassium phosphate, pH 8.0, 5 mM KCl, 2.5 mM MgCl2,

1 mM ATP, 450 μM vinthionine, 400 μM 4-nitrothiophenol, 2 mM TCEP, 1.75 μM MTAN, and

74 μM TPMT. The reaction was initiated with 160 μM MAT and incubated at 37 °C. The same

reactions without MAT or TPMT were conducted as negative controls. The concentration of 4-

nitrothiophenol was determined using ε 411 nm = 13,650 M-1 cm-1.15

AdoVin-4-Nitrophenol Adduct Formation

The reaction contained 50 mM potassium phosphate, pH 8.0, 10 mM KCl, 5 mM MgCl2,

2 mM ATP, 900 μM vinthionine, 1.0 mM 4-nitrophenol, 2 mM TCEP, 1.75 μM MTAN, and 74

μM TPMT. The same reactions without MAT or TPMT were conducted as negative controls, “No

MAT” and “No TPMT.” The reaction was initiated with 182 μM MAT and incubated at 37 °C.

Page 96: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

77

2.4.7 Binding Assay

The binding assay was conducted at 4 °C using the HisPur Ni-NTA Spin Column (0.2 mL

resin bed, Thermo scientific). Storage buffer of column was removed by centrifuging column at

700 g for 2 min, and the column was equilibrated with 400 μL equilibration buffer (20 mM

potassium phosphate, 300 mM NaCl and 10 mM imidazole, pH 8.0). The reaction samples were

mixed with an equal volume of equilibration buffer and then loaded onto the columns. The column

was then mixed on an end-over-end mixer for 30 min at 4 °C. Then 3 x 400 μL washing buffer (50

mM potassium phosphate, 300 mM NaCl, and 25 mM imidazole, pH 8.0) was used to wash the

resin. TPMT and TPMT-adduct complex eluted with 3 x 200 μL elution buffer (50 mM potassium

phosphate, 300 mM NaCl, and 250 mM imidazole, pH 8.0).

Ultrafiltration was carried out using filters with 30,000 MWCO (0.5 mL). The reaction

sample (100 μL) was loaded to the membrane containing vials. Ammonium bicarbonate, pH 8.0

(400 μL 50 mM) was added to the retentate, and the diluted free ligands were filtered out again by

12,000 g for 5 min. The buffer exchange procedure was continued until the UV absorbance of the

flow through was 0. The retentate was removed for further analysis, as well as free ligands fraction

by the first filtration.

The protein containing fractions from both methods were monitored by measuring the UV

absorbance at 280 nm, and injected into HPLC after protein acidic precipitation.

Page 97: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

78

2.4.8 ex vivo AdoVin Adduct Formation Catalyzed by TPMT

Preparation of ex vivo AdoVin-TNB reactions

The concentration of TPMT in TPMT lysate was estimated as 336 μM by the TPMT band

on SDS-PAGE gel. The TPMT lysate was prepared as described in general procedure without the

purification. The reaction (200 μL) contained 50 mM potassium phosphate, pH 8.0, 50 mM KCl,

25 mM MgCl2, 10 mM ATP, 1 mM vinthionine, 489 μM TNB, 2 mM TCEP, 3.5 μM MTAN and

100 μL TPMT cell lysate. The reaction was initiated with 295 μM MAT and incubated at 37 °C.

To do the binding assay between ex vivo formed AdoVin-TNB adduct and TPMT, the ex

vivo TNB reaction sample was loaded onto the HisPur Ni-NTA Spin Column, and isolation of

TPMT fraction from TPMT lysates was described previously.

Preparation of isotope labeling AdoVin-TNB adduct

The reaction contained 50 mM potassium phosphate, pH 8.0, 5 mM KCl, 2.5 mM MgCl2,

1 mM Adenosine-13C10,15N5-5′-triphosphate sodium salt solution (isotope labeled ATP, Sigma-

Aldrich, catalog No. 645702), 600 μM vinthionine, 489 μM TNB, 2 mM TCEP, 1.75 μM MTAN,

and 70 μM TPMT. The reaction was initiated with 120 μM MAT and incubated at 37 °C.

Page 98: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

79

2.4.9 Elucidation and Confirmation of Unknown Adduct

Synthesis of 5,5'-dithio-bis-(2-aminobenzoic acid)

As reported,27-28 an 8 mL solution of 7.5 M ammonium hydroxide was degassed by

bubbling N2 through the magnetically stirred solution and was heated to 85-90 °C with an oil bath

at 100 °C. Ferrous sulfate heptahydrate (1.63 g, 5.86 mmol) was added to the basic solution while

heating, stirring, and maintaining a nitrogen atmosphere. Ellman’s reagent (5,5-dithio-bis-(2-

nitrobenzoic acid), TNB, Alfa Aesar A14331, 0.116 g, 0.293 mmol) in 1.5 mL of 7.5 M ammonium

hydroxide was added dropwise to the reaction mixture. After two hours, the oil bath was removed

and the reaction mixture was allowed to cool to room temperature. Decolorizing carbon (100 mg)

was added to the black reaction mixture, which was then filtered through filter paper. The yellow-

orange filtrate was collected and cooled to 5 °C. The filtrate was acidified with concentrated

phosphoric acid to pH 3. The aqueous mixture was extracted with 2-propanol/chloroform (25:75,

4 x 15 mL). The organic extracts were combined and back-washed with water (2 x 30 mL). The

organic phase was dried over sodium sulfate, the solvent removed by rotary evaporation, and

stored under vacuum overnight to give 5,5'-dithio-bis-(2-aminobenzoic acid) as a yellow powder

that was homogeneous by TLC Rf = 0.08 in 25:75 i-PrOH/CHCl3; m.p. 200°C; 1H-NMR

(CDCl3/DMSO-d6 95:5) δ 7.88 (d, J = 2.1 Hz, 2H), 7.22 (dd, J = 8.6, 2.1 Hz, 2H), 6.65 (d, J = 8.6

Hz, 2H). LC-MS ES-TOF (m/z)+ 337.0 [(M+H)+, calcd for C14H13N2O4S2: 337.0] and 319.0 [(M-

OH)+, calcd for acylium ion C14H11N2O3S2: 319.0].

Page 99: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

80

in vitro Bisubstrate-Adduct Formation between AdoVin and AMBA

The reaction contained 50 mM potassium phosphate, pH 8.0, 50 mM KCl, 25 mM MgCl2,

10 mM ATP, 1 mM vinthionine, 500 μM AMBA, 2 mM TCEP, 3.5 μM MTAN and 100 μM TPMT.

The reaction was initiated with 200 μM MAT and incubated at 37 °C.

To verify the adduct peak in AdoVin-AMBA reaction, the in vitro reaction mixture was

co-injected with ex vivo AdoVin-TNB reaction mixture. The ex vivo AdoVin-TNB reaction sample

was prepared as described previously.

2.4.10 Confirmation of 2-Amino-5-Mercaptobenzoic Acid as TPMT Substrate

The assays contained 50 mM potassium phosphate, pH 8.0, 1 mM AdoMet, 1 mM AMBA,

2 mM TCEP, and 3.5 μM MTAN. The reaction was initiated with 13.2 μM TPMT and incubated

at 37 °C. 10 μL reaction samples (at 0, 15, 30, 60 and 120 min) were analyzed by HPLC. The

eluting profiles were monitored by UV absorbance at both 260 and 350 nm.

2.4.11 Modification of MAT and TPMT by AdoVin

AdoVin was enzymatically synthesized in 50 mM ammonium bicarbonate, pH 8.0, MAT

was filtered out by using filters with 30,000 Da molecular weight cut off (MWCO). The AdoVin

solution was injected into HPLC with UV detector, and the concentration of AdoVin was

determined by UV absorbance at 260 nm.

The assay solution contained 50 mM ammonium bicarbonate (pH 8.0), 71.32 μM AdoVin

was incubated with 41.2 μM MAT and 42 μM TPMT, respectively. Samples of MAT and TPMT

Page 100: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

81

without treatment with AdoVin were prepared as control. All the samples were incubated at 37 °C

for 18 h and analyzed by MALDI-TOF/TOF.

2.4.12 Kinetics Analysis of TPMT-Catalyzed AdoVin-TNB Adduct Formation

The reaction contained 200 mM Tris buffer, pH 8.0, 50 mM KCl, 25 mM MgCl2, 10 mM

ATP, 1 mM vinthionine, 56.8 μM TNB, 2 mM TCEP, 3.5 μM MTAN, 100 μM MAT and variable

TPMT concentrations (0, 5.9, 10.7 and 21.5 µM).

AdoVin-TNB adduct formation was initiated with TPMT at various concentrations, and

incubated at 37 °C. The spectral changes were monitored continuously between 260-610 nm for

240 min. Three scanning cycles were set up with different scan time intervals: 1 min for the first

60 min, then 5 min between 60 and 120 min incubation, and finally increased to 30 min until 240

min. Kaleidagraph software package 4.1 (Synergy Software, Reading, PA) was used to analyze

the absorbance change at 411 nm.

2.4.13 HPLC Analysis of AdoVin and AdoVin reactions

Reverse Phase HPLC Separation of AdoVin

Aliquots of reaction mixture (10 μL) were removed and analyzed by HPLC, and elution

profiles were monitored by UV absorption at 260 nm. The separation of AdoVin was carried out

on a reverse-phase column (Apollo, C18, 5μ, 4.6 mm x 150 mm), using 0.1% aqueous

trifluoroacetic acid (TCA) (mobile phase A) and 0.1% TCA in acetonitrile (mobile phase B) at a

flow rate of 1 mL/min. The gradient program was initiated with 2% mobile phase B, followed by

Page 101: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

82

a linear increase to 10% mobile phase B over 8 min, then a return to 2% mobile phase B over 1

min, and finally a hold at 2% mobile phase B over 5 min.

Strong Cation Exchange HPLC Separation of AdoVin

Aliquots of reaction mixture (10 μL) were removed and analyzed by HPLC, and elution

profiles were monitored by UV absorption at 260 nm. The chromatography was performed on a

strong cation exchange (SCX) column (ProPac SCX-10 analytical, 4×250 mm) using 20%

Acetonitrile and 0.07% NH4OH inwater (mobile phase A,pH 3) and 50 mM (NH4)2SO4, 20%

Acetonitrile and 0.07% NH4OH in water; (mobile phase B, pH 3) at a flow rate of 1 mL/min. The

gradient program was initiated with 0% mobile phase B, followed by a linear increase to 50%

mobile phase B over 20 min, then a return to 0% mobile phase B over 1 min, and finally a hold at

0% mobile phase B over 9 min.

Reverse Phase HPLC Separation of AdoVin Adducts

All the samples were injected and analyzed by HPLC, elution profiles were monitored by

UV absorption at 260 nm and 350 nm, based on the absorbance for AdoMet and alkyl TNB. The

chromatography was performed on a reversed-phase column (Apollo, C18, 5μ, 4.6 mm x 150 mm)

using 0.1% aqueous TFA (mobile phase A) and 0.1% TFA in acetonitrile (mobile phase B) at a

flow rate of 1 mL/min. The gradient program was initiated with 2% mobile phase B, and increased

to 10% mobile phase B over 8 min, then increased to 30% mobile phase B over 12 min, then

increased to 70% mobile phase B over 15 min, followed by an increase to 90 % mobile phase B

over 5 min, then a return to 2% mobile phase B over 1 min, and finally a hold at 2% mobile phase

B over 5 min.

Page 102: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

83

Reverse Phase HPLC Separation of methylated AMBA

Aliquots (10 μL) of reactions and negative controls were injected and analyzed by HPLC,

elution profiles were monitored by UV absorption at 260 nm and 350 nm, based on the absorbance

for AdoMet and alkyl TNB. The chromatography was performed on a reversed-phase column

(Apollo, C18, 5μ, 4.6 mm x 150 mm) using 0.1% aqueous formic acid (mobile phase A) and 0.1%

formic acid in acetonitrile (mobile phase B) at a flow rate of 1 mL/min. Same chromatography

gradient program was used in separation of AdoVin adducts.

2.4.14 Mass spectrometry

All the analysis samples were exchanged into 50 mM ammonium bicarbonate, pH 8.0.

TCA (100% v/v) was used to precipitate the proteins in the samples, and the supernatants were

saved for further analysis by mass spectrometry. Reaction and negative control samples (15 μL of

each sample) were taken for HPLC-MS analysis. Free ligands fraction after ion metal affinity

chromatography (50 μL) were taken for HPLC-MS analysis.

LCQ Ion Trap mass spectrometer was used in-line with HPLC system (Agilent 1100) for

the identification of AdoVin and AdoVin adducts, respectively. The chromatography was

performed on a reversed-phase column (Apollo, C18, 5μ, 4.6 mm x 150 mm) using 0.1% aqueous

formic acid (mobile phase A) and 0.1% formic acid in acetonitrile (mobile phase B) at a flow rate

of 1 mL/min. The gradient program was initiated with 2% mobile phase B, and increased to 10%

mobile phase B over 8 min, then increased to 30% mobile phase B over 12 min, then increased to

70% mobile phase B over 15 min, followed by an increase to 90 % mobile phase B over 5 min,

then a return to 2% mobile phase B over 1 min, and finally a hold at 2% mobile phase B over 5

Page 103: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

84

min. LCQ was operated with a full scan and data-independent MSMS scan of the top ten most

abundant ions. The spray voltage was 2.3 kV, and the capillary temperature was 285 °C. Data were

processed using the Xcalibar Data System 2.0 (Thermo Fisher, Waltham, MA).

2.4.15 MALDI-TOF/TOF Analysis

Protein samples was diluted to 50 µM using a solution of acetonitrile/water/TFA (v:v:v:,

50:50:0.5). Protein samples (0.5 µL) were spotted on a clean standard 192 well stainless steel

MALDI sample plate followed by the addition of sinapinic acid matrix solution (0.5 µL, 10

mg/mL in a mixture of acetonitrile/water/TFA, v:v:v:, 50:50:0.5). The resulting mixtures were

air-dried and analyzed using an AB 5800 MALDI-TOF/TOF Proteomics Analyzer (Applied

Biosystems Framingham, MA). MS spectra were acquired in linear positive mode. Lysozyme

([M + H]+: 14,308 Da) and BSA ([M + H]+: 66,464 Da) were used for external calibration.

Software Data Explorer 4.6 was used to analyze the data.

Page 104: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

85

2.5 References

1. Cantoni, G. L., Biological methylation: selected aspects. Annu Rev Biochem 1975, 44, 435-

51.

2. Kim, H. J.; Balcezak, T. J.; Nathin, S. J.; McMullen, H. F.; Hansen, D. E., The use of a

spectrophotometric assay to study the interaction of S-adenosylmethionine synthetase with

methionine analogues. Anal Biochem 1992, 207 (1), 68-72.

3. Wang, R.; Zheng, W.; Luo, M., A sensitive mass spectrum assay to characterize engineered

methionine adenosyltransferases with S-alkyl methionine analogues as substrates. Anal

Biochem 2014, 450, 11-9.

4. Wang, R.; Islam, K.; Liu, Y.; Zheng, W.; Tang, H.; Lailler, N.; Blum, G.; Deng, H.; Luo,

M., Profiling genome-wide chromatin methylation with engineered posttranslation

apparatus within living cells. J Am Chem Soc 2013, 135 (3), 1048-56.

5. Wu, S. E.; Huskey, W. P.; Borchardt, R. T.; Schowen, R. L., Chiral instability at sulfur of

S-adenosylmethionine. Biochemistry 1983, 22 (12), 2828-32.

6. Hoffman, J. L., Chromatographic analysis of the chiral and covalent instability of S-

adenosyl-L-methionine. Biochemistry 1986, 25 (15), 4444-9.

7. Iwig, D. F.; Booker, S. J., Insight into the polar reactivity of the onium chalcogen analogues

of S-adenosyl-L-methionine. Biochemistry 2004, 43 (42), 13496-509.

8. Graham, D. E.; Bock, C. L.; Schalk-Hihi, C.; Lu, Z. J.; Markham, G. D., Identification of

a highly diverged class of S-adenosylmethionine synthetases in the archaea. J Biol Chem

2000, 275 (6), 4055-9.

9. Farrar, C.; Clarke, S., Altered levels of S-adenosylmethionine and S-

adenosylhomocysteine in the brains of L-isoaspartyl (D-Aspartyl) O-methyltransferase-

deficient mice. J Biol Chem 2002, 277 (31), 27856-63.

10. Wu, H.; Horton, J. R.; Battaile, K.; Allali-Hassani, A.; Martin, F.; Zeng, H.; Loppnau, P.;

Vedadi, M.; Bochkarev, A.; Plotnikov, A. N.; Cheng, X., Structural basis of allele variation

of human thiopurine-S-methyltransferase. Proteins 2007, 67 (1), 198-208.

11. Ames, M. M.; Selassie, C. D.; Woodson, L. C.; Van Loon, J. A.; Hansch, C.; Weinshilboum,

R. M., Thiopurine methyltransferase: structure-activity relationships for benzoic acid

inhibitors and thiophenol substrates. J Med Chem 1986, 29 (3), 354-8.

Page 105: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

86

12. Krynetski, E. Y.; Krynetskaia, N. F.; Yanishevski, Y.; Evans, W. E., Methylation of

mercaptopurine, thioguanine, and their nucleotide metabolites by heterologously expressed

human thiopurine S-methyltransferase. Mol Pharmacol 1995, 47 (6), 1141-7.

13. Woodson, L. C.; Weinshilboum, R. M., Human kidney thiopurine methyltransferase.

Purification and biochemical properties. Biochem Pharmacol 1983, 32 (5), 819-26.

14. Carey, F. A.; Sundberg, R. J., Advanced organic chemistry. 4th ed.; Kluwer

Academic/Plenum Pub.: New York, 2000; p 2 volumes.

15. Eyer, P.; Worek, F.; Kiderlen, D.; Sinko, G.; Stuglin, A.; Simeon-Rudolf, V.; Reiner, E.,

Molar absorption coefficients for the reduced Ellman reagent: reassessment. Anal Biochem

2003, 312 (2), 224-7.

16. Cannon, L. M.; Butler, F. N.; Wan, W.; Zhou, Z. S., A stereospecific colorimetric assay

for (S,S)-adenosylmethionine quantification based on thiopurine methyltransferase-

catalyzed thiol methylation. Anal Biochem 2002, 308 (2), 358-63.

17. Spain, J. C., Biodegradation of nitroaromatic compounds. Annu Rev Microbiol 1995, 49,

523-55.

18. Meyer, D.; Witholt, B.; Schmid, A., Suitability of recombinant Escherichia coli and

Pseudomonas putida strains for selective biotransformation of m-nitrotoluene by xylene

monooxygenase. Appl Environ Microbiol 2005, 71 (11), 6624-32.

19. Race, P. R.; Lovering, A. L.; Green, R. M.; Ossor, A.; White, S. A.; Searle, P. F.; Wrighton,

C. J.; Hyde, E. I., Structural and mechanistic studies of Escherichia coli nitroreductase with

the antibiotic nitrofurazone. Reversed binding orientations in different redox states of the

enzyme. J Biol Chem 2005, 280 (14), 13256-64.

20. Roldan, M. D.; Perez-Reinado, E.; Castillo, F.; Moreno-Vivian, C., Reduction of

polynitroaromatic compounds: the bacterial nitroreductases. FEMS Microbiol Rev 2008,

32 (3), 474-500.

21. Christofferson, A.; Wilkie, J., Mechanism of CB1954 reduction by Escherichia coli

nitroreductase. Biochem Soc Trans 2009, 37 (Pt 2), 413-8.

22. Mercier, C.; Chalansonnet, V.; Orenga, S.; Gilbert, C., Characteristics of major Escherichia

coli reductases involved in aerobic nitro and azo reduction. J Appl Microbiol 2013, 115 (4),

1012-22.

Page 106: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

87

23. Peng, Y.; Feng, Q.; Wilk, D.; Adjei, A. A.; Salavaggione, O. E.; Weinshilboum, R. M.;

Yee, V. C., Structural basis of substrate recognition in thiopurine s-methyltransferase.

Biochemistry 2008, 47 (23), 6216-25.

24. Lee, J. E.; Cornell, K. A.; Riscoe, M. K.; Howell, P. L., Expression, purification,

crystallization and preliminary X-ray analysis of Escherichia coli 5'-

methylthioadenosine/S-adenosylhomocysteine nucleosidase. Acta Crystallogr D Biol

Crystallogr 2001, 57 (Pt 1), 150-2.

25. Leopold, W. R.; Miller, J. A.; Miller, E. C., S--vinyl homocysteine, an analog of ethionine

that is highly mutagenic for S. typhimurium TA100. Biochem Biophys Res Commun 1979,

88 (2), 395-401.

26. Hanes, J. W.; Kraft, C. E.; Begley, T. P., An assay for thiaminase I in complex biological

samples. Anal Biochem 2007, 368 (1), 33-8.

27. Stajer, G.; Kottke, K.; Pohloudek-Fabini, R., [Synthesis of 2,6-dimercapto-3-aryl-4-

quinazolones and various derivatives]. Pharmazie 1973, 28 (7), 433-7.

28. Wojahn, H.; Lerch, I., [Aromatic disulfide compounds with tuberculostatic effect].

Arzneimittelforschung 1952, 2 (10), 455-60.

Page 107: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

88

Chapter 3: Identification of Substrates by Native Mass Spectrometry

This chapter is based on a manuscript with the title of “Identifying Substrates of Thiopurine

Methyltransferase via Enzyme-Ligand Complex Using Native Mass Spectrometry” with the author

list: Jing Yan, Wanlu Qu, Kalli Catcott, Vicki H. Wysocki, and Zhaohui Sunny Zhou

Co-authors’ works in this chapter: Jing Yan and Wanlu Qu contributed equally; Jing Yan,

mass spectrometric analysis; Wanlu Qu, experimental design and performance on AdoVin

reactions; Kalli Catcott, cell lysates preparation, manuscript writing and idea contributions; Vicki

H. Wysocki, idea contributions; Zhaohui Sunny Zhou, principal investigator.

Page 108: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

89

3.1 Introduction

3.1.1 Native Mass Spectrometry

Understanding proteins and conformation of protein networks is of high importance.

However, the characterization of proteins and native protein networks, including protein-protein

interactions and protein-small molecule interactions, remains challenging, despite the routine

determination of protein primary sequence. Recently, the application of soft ionization methods in

mass spectrometry (native MS) allows the efficient transfer of intact large proteins into the gas

phase, making it possible to characterize intact proteins with higher-order structure and detect

native protein networks.1

Native protein mass spectrometry was termed by Heck and van den Heuvel as a powerful

technique to characterize proteins in their native functional form, and analyze large intact protein

complexes, even non-covalent complexes.2-4 Currently, native MS with the soft ionization method,

nano-ESI, is applicable for analyzing protein complexes with masses up to 18 MDa.3 Furthermore,

the development of tandem MS followed by native MS has broadened the utility of MS in the

characterization of protein structures and protein complexes, especially those with unknowns.

Page 109: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

90

3.1.2 Tandem Mass Spectrometry and Fragmentation in Tandem MS

Tandem mass spectrometry, also known as MS/MS or MS2, is a common technique for

generating fragments that can contribute to the structural analysis of precursor ions. There are

mainly three steps in tandem mass spectrometry: 1) selection of precursor ions from the first stage

mass analyzer; 2) fragmentation of selected precursor ions; 3) analysis of the fragmentation ions

by a second stage mass analyzer. Multiple fragmentation can be performed as MSn.5 Additionally,

the precursor ions can be selected and isolated by both data-dependent and precursor ion targeting

modes.

There are two commonly-used fragmentation techniques in tandem mass spectrometry:

collision-induced dissociation (CID, also known as collisionally activated dissociation) and

electron transfer dissociation (ETD). In our work, another newly developed fragmentation

technique has been applied, that is, surface induced dissociation (SID).

CID involves collision of the selected precursor ions with inert gas ions, i.e., helium or

nitrogen, resulting in a vibrational excitation in the precursor ions. Part of the kinetic energy of the

precursor ions can be converted into internal energy that generates corresponding product ions.

CID is commonly applied to break the amide bonds of peptide backbone.6 The limitation of CID

is exemplified in the detection of protein posttranslational modifications (PTMs), because most

modifications bonds are weaker than peptide bonds, and thus are cleaved first under CID

fragmentation.7

ETD is a newly developed fragmentation technique that transfers electrons to positively

charged proteins or peptides, which turns proteins or peptides into radicals, thus generating

fragmentation ions.6 Unlike CID, ETD cleaves Cα-N bonds along the peptide backbones, while

Page 110: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

91

leaving the side chains and their modifications intact. Therefore, ETD is widely applicable in the

discovery and localization of PTMs.5

In CID, the activation occurs with internal energy via many low-energy collisions with

inert gas atoms. Differently, in SID, the fragmentation is caused by the collision of precursor ions

with a surface under high vacuum in a much shorter time.8 Therefore, SID is more effective and

efficient for detecting protein folding/unfolding transition states than CID, in which significant

structural rearrangement may occur and lead to unfolded monomers. Currently, the combination

of CID and SID is applied to investigate the structures and the dissociation pathways of protein

complexes.

3.1.3 Ion Mobility Spectrometry with Mass Spectrometry

Ion mobility spectrometry (IMS)9 is used to separate ions according to their mobilities in

gas phase,10-11 and it has been highlighted as a potential tool to elucidate the structures of proteins.12

The application of a unique gas-phase electrophoretic mobility molecular analyzer (GEMMA) has

extended the measurement of large protein complexes up to the megadalton range.13-16

Separately, mass spectrometry has been applied in many fields, such as proteomics,

nanomaterials, and medicinal screening, because of its advantages: easy operation, high sensitivity,

high throughput, and high resolution. Ion mobility separates ions based on their size and shape,

and MS identifies the ions by their mass-to-charge ratio (m/z). Currently, the combination of Ion

mobility spectrometry and mass spectrometry is an analytical technique that is widely applied on

structural biology with more sensitivity and greater peak capacity than mass spectrometry.17-19

Page 111: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

92

3.1.4 Identification of Enzyme Substrates

Identification of substrates of enzymes is the essential step to fully understand the

functional activities of enzymes. Previous efforts to identify the substrate specificity of enzymes

have included broad substrate screening, computer-aided substrate screening and substrate-

trapping screening.

High-throughput Screening

High-throughput screening on a broad range of substrates has been developed to identify

substrates of enzymes, in which the substrate binding domains are not conserved.20-21 For example,

Gcn5-related N-acetyltransferases (GNATs), which catalyze the transfer of an acetyl group from

acetyl coenzyme A (AcCoA) to a substrate.22 Even though the structure elucidation for GNATs

has been established, the identification of its substrates remains challenging, due to the flexible

substrate binding domains and a wide variety of substrate specificity. This method is a potential

way to identify substrate specificity of enzymes, but cannot be applied to enzymes with unknown

structures.

Computer-Aided Substrate Screening (CASS)

Computer-Aided Drug Design (CADD) has been developed rapidly in the design of protein

inhibitors.23 Based on the similar mechanism between protein-inhibitor binding and enzyme-

substrate interaction, a computer-aided substrate screening (CASS) using restricted molecular

docking was designed.24-25 Structure-based CASS is a reliable and highly accurate method for

identifying the substrate specificity of enzymes. Compared to experimental screening, CASS is

less laborious, less time consuming and less costly. However, the limitation of this method is that

molecular docking may not be appropriate for prediction of the substrate-enzyme interaction, since

Page 112: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

93

the substrate-enzyme binding may undergo energetically unfavorable intermediate that cannot be

predicted by molecular docking.26 Also, the method is only applicable to enzymes with known

structures.

Substrate-Trapping System

Substrate-trapping system relies on the protein mutants of its active sites. Due to the active

site mutations, the binding affinity between protein and its substrates is retained, or even enhanced,

but their catalytic activity is reduced. Therefore, stable enzyme-substrate intermediates can be

formed in the catalytically inactive sites of enzymes.27-28 This method enables the isolation and

identification of both in vitro and in vivo enzyme substrates.27, 29 However, the application of this

strategy is restricted to the abundance of substrates30 and conserved enzymes structures.28, 30

3.1.5 Identification of Enzyme Substrates via Enzyme-Substrate Complex

In our approach, AdoVin is an efficient “substrate-trapper” to identify substrates of MTases,

independent of the structures of enzymes. Previously, the TPMT-adduct complex was initially

isolated from the reaction mixture and then injected into HPLC and LC-MS for analysis after

protein acidic precipitation. Herein, we developed a new method to identify substrates of enzymes

via enzyme-substrate complex without any isolation of enzymes. In our method, native MS was

used to separate proteins and protein complexes by their mass to charge ratio (m/z), followed by

IMS, which separates proteins and protein complexes based on their size and shape.17

Subsequently, both CID and SID were used to dissociate the binding substrates from enzyme-

substrate complex, and thus the released substrates could be identified by the tandem MS

fragmentation (Scheme 3.1). Moreover, the binding affinity between enzymes and substrate was

Page 113: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

94

investigated under various collision energies of CID and SID. Using this method, TPMT complex

with AdoVin adduct was observed even in crude E. coli cell lysates. Altogether, our method

provides, for the first time, the observation of MTase-substrate binding complex in native MS.

Scheme 3.1. Detection of enzyme-ligand complex by using native mass spectrometry and

subsequent structural elucidation by tandem mass spectrometry (CID or SID).

Page 114: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

95

3.2 Results and Discussions

3.2.1 TPMT in Gas Phase

Two charge state distributions of TPMT were observed in gas phase, folded and unfolded.

As shown in Figure 3.1, TPMT with charge states from +9 to +13 was folded with salts attached,

so these peaks are broad, while TPMT with larger charge state, +14 to +16, was unfolded with

fewer salt attached, providing narrow peaks.

The structure of TPMT is not stable in gas phase. Two major peaks were observed at each

charge state (Figure 3.1). One was calculated to be 30212.6, 131.0 less than the theoretical mass

of TPMT (30343.6). The other one was 30596.0, 253.0 more than theoretical mass.

CID was then used to remove the salts attached to TPMT (+9 TPMT). Interestingly, after

targeting the broad +9 peak as a precursor ion in CID, the salts were dissociated from the +9 TPMT

peak and covalent bond cleavage was observed on parts of them (Figure 3.2). The mass of protein

was then calculated as 30214.0, according to the remaining +9 precursor ions. This observation

indicated that the TPMT sequence is one methionine less than theoretical.

Page 115: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

96

Figure 3.1. Mass spectrum of TPMT (top) and zoomed in mass spectrum of TPMT (bottom).

Page 116: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

97

Figure 3.2. Tandem mass spectrum of +9 precursor ions under CID.

Page 117: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

98

3.2.2 Detection of Enzyme-Substrate Complex

As described in Chapter Two, AdoVin reacted in situ with TNB in the presence of

thiopurine methyltransferase (TPMT), and the resulting bisubstrate-adduct tightly bound with

TPMT. In order to develop a method to identify substrate of enzyme via enzyme-substrate complex,

native MS was used to monitor the interaction between AdoVin adduct and TPMT in gas phase.

In these experiments, isotope labeled AdoVin-TNB was used to introduce an isotope

doublet pattern, and the ratio between regular and isotopic AdoVin-TNB adduct was 1:1. Both

TPMT and TPMT with AdoVin-TNB adduct were observed as different charge states (Figure 3.3).

Using the most abundant peaks “+10 TPMT and +10 TPMT-ligand”, the deconvoluted masses of

TPMT was calculated as 30214.1, and the TPMT-ligand was calculated as 30832.1. The difference

between TPMT and TPMT-ligand is 618.0, which exactly matches the mass average between

AdoVin-TNB (610.0) and isotope labeled AdoVin-TNB adduct (625.0). This result indicates that

AdoVin-TNB was formed, and the TPMT complex with AdoVin-TNB adduct can be observed in

the native MS. The two adduct forms were also clearly observed in the zoomed in MS spectrum

(Figure 3.3). The deconvoluted masses of the peaks are labeled, 30846.1 and 30831.6, respectively.

Compared to the mass of TPMT (30214.1), the mass difference (Δm) between the species and

TPMT are 609.8 and 624.3, close to the theoretical mass of AdoVin-TNB adduct (610.0) and its

isotopic form (625.0), indicating that AdoVin-TNB adduct is also bound to TPMT in the gas phase

and the isotope shift (15 Da) can be distinguished. Therefore, isotope labeled AdoVin adduct can

be applied to the verification of unknown enzyme-ligand complex in the future.

Page 118: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

99

Figure 3.3. Mass spectrum of reaction mixture of TPMT catalyzed AdoVin-TNB adduct formation

(top), and zoomed in mass spectrum of + 10 charged TPMT complex with AdoVin-TNB adduct.

Isotopic AdoVin-TNB adduct was used for verification (bottom).

Page 119: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

100

Next, to investigate the fragmentation of TPMT-ligand complex, various collision energies

were tested for both CID and SID. Apoenzyme TPMT, TPMT-AdoVin-TNB complex, and

dissociated AdoVin-TNB adduct were all observed. More AdoVin-TNB adduct dissociated from

TPMT with higher collision energies in both CID and SID (Figure 3.4 and 3.5). Moreover, higher

collision energies cause more fragmentation of AdoVin-TNB adduct (Figure 3.4 and 3.5), which

is consistent with the structure of AdoVin-TNB determined in chapter two. It worth noting that the

isotope labeling introduced isotopic doublet fragmentations, and this method could be extended to

enzyme-unknown ligand system, in which the fragmentations is the main evidence to identify

unknown ligand, and even better with the introduction of isotopic patterns.

In theory, CID could generate more fragmentation than SID at equivalent energies for

enzyme-ligand complexes, due to different ion dissociation mechanism (discussed in the

introduction), however, for TPMT-AdoVin-TNB complex, no significant difference in

fragmentation was observed between CID and SID (Figure 3.4 and 3.5). This observation is

explainable, since TPMT-AdoVin-TNB complex is not a real three component complex. Instead,

AdoVin and TNB was covalently bound and dissociated as one part in both CID and SID.

Page 120: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

101

Figure 3.4. Tandem mass spectra of +10 charged TPMT-ligand complex in AdoVin-TNB reaction

mixture under different CID energy (top). Zoomed in tandem mass spectra of +10 charged TPMT-

ligand complex in AdoVin-TNB reaction mixture under various CID energy (bottom).

Page 121: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

102

Figure 3.5. Tandem mass spectra of +10 charged TPMT-ligand complex in AdoVin-TNB reaction

mixture under different SID energy (top). Zoomed in tandem mass spectra of +10 charged TPMT-

ligand complex in AdoVin-TNB reaction mixture under various SID energy (bottom).

Page 122: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

103

3.2.3 Detection of Enzyme-Substrate Complex in Crude Biological Samples

In order to test the ability of native MS to detect enzyme-ligand complex in more

complicated cellular samples, over-expressed TPMT lysates (~50% total protein by weight) were

used to generate AdoVin-TNB adducts in situ with exogenous MAT. TPMT-ligand complexes

were observed (Figure 3.6) and isolated for both CID and SID fragmentation. Similar to the results

of in vitro samples, apoenzyme TPMT, TPMT-AdoVin-AMBA complex and AdoVin-AMBA

adduct were observed under both CID and SID fragmentation (Figure 3.7). However, the

significant difference is that AdoVin-AMBA adduct (2-amino-5-mercaptobenzoic acid, AMBA)

was the major adduct, rather than AdoVin-TNB adduct. This is likely due to the reduction of TNB

by any of four nitroreductases present in E. coli, as discussed in chapter two.

Figure 3.6. Mass spectrum of ex vivo reaction mixture of TPMT catalyzed AdoVin-TNB adduct

formation.

Page 123: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

104

Figure 3.7. Tandem mass spectra of +10 charged TPMT-ligand complex in ex vivo AdoVin-TNB

reaction by CID (500 eV) and SID (500 eV).

The TPMT complex with AdoVin-AMBA adduct was readily detected in ex vivo level with

the existence of over 50% TPMT. In typical biological system, most enzymes do not exist at such

high concentrations. Therefore, the possibility to detect enzyme-ligand complex in more

complicated samples needs to be explored. Different from previous experiments, TPMT was

expressed without any further induction (in the studies described above, E. coli cells were induced

with IPTG to express TPMT), and the cells were then lysed as the source of TPMT in AdoVin-

TNB reactions without further purification.

Page 124: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

105

We were happy to see, as shown in Figure 3.8, TPMT-adduct complex was observed in

native MS with lower content of TPMT (under 5% of total protein by weight). AdoVin-AMBA

adduct was also the major adduct, and readily identified under both CID and SID fragmentation

(Figure 3.9). Compared to the previous methods, no purification of TPMT was needed in this

convenient and streamlined method. Moreover, native MS provides a more sensitive detection for

TPMT-adduct complex and identification of AdoVin adduct. For instance, only 1 pmole TPMT-

adduct complex was needed, and the detection limit is much lower than that of previous detection

method, in which at least 10 pmole of AdoVin adduct was required.

Figure 3.8. Mass spectrum of ex vivo reaction mixture of TPMT catalyzed AdoVin-TNB adduct

formation in TPMT lysate from non-induced E. coli cells. AdoVin was enzymatically synthesized

by MAT, and AdoVin-TNB adduct was formed in situ.

Page 125: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

106

Figure 3.9. Tandem mass spectrum of +10 charged TPMT-ligand complex in ex vivo AdoVin-

TNB reaction with non-induced TPMT lysate by CID (500 eV).

Page 126: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

107

More importantly, prior to MS analysis, all the samples were buffer exchanged by

ultrafiltration, and the small molecules under 30,000 Da were removed by ultrafiltration, and

therefore the fact that TPMT-AdoVin complex was not observed in the retentate suggests no

modification on TPMT by AdoVin, consistent with the results reported in Chapter 2.

Using the example of tight binding AdoVin adduct with TPMT, this method has been

proven as an efficient way to detect enzyme-ligand complex by native MS and identify the ligands

by subsequent tandem MS. It is important to mention that TPMT complex with AdoMet, the

natural methyl donor of MTases, and AdoHcy, a natural product inhibitor to MTases, were also

observed using Native MS (Figure 3.10 and 3.13). Subsequently, both TPMT-AdoMet and TPMT-

AdoHcy complexes were detectable in low CID (300 eV) (Figure 3.10-3.13). In contrast to the

AdoVin adducts, with low energy SID (500 eV) and high energy CID (1000 eV), AdoMet and

AdoHcy (Kd = 0.75 µM)31 were readily dissociated from TPMT (Figure 3.12 and 3.14), due to

their weaker binding to TPMT than AdoVin-TNB or AdoVin-AMBA adduct. These results further

indicate that this method could be used to discover enzyme substrates and identify those substrates

with tandem MS fragmentation (Figure 3.12 and 3.15).

Overall, the tight binding event of AdoVin-TNB with TPMT was determined by using

native MS and the tandem MS with CID and SID, without any separation of TPMT from the

reaction mixture. Moreover, the observation of TPMT-AdoMet and TPMT-AdoHcy complex also

supports that this method is efficient and sufficient for enzyme-ligand screening, and drug

candidate development.

Page 127: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

108

Figure 3.10. Mass spectra of TPMT-AdoMet complex.

Page 128: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

109

Figure 3.11. Tandem mass spectrum of +10 peak (TPMT-AdoMet) dissociated by low energy CID.

The calculated mass of TPMT-AdoMet complex is 30955.8 Da, and the calculated mass of TPMT

is 30557.9 Da. The difference is 397.9, close to the mass of AdoMet (399.0 Da).

Page 129: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

110

Figure 3.12. Tandem mass spectra of +10 peak (TPMT-AdoMet) for high energy CID and low

energy SID.

Page 130: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

111

Figure 3.13. Mass spectra of in vitro AdoVin-TNB reaction (top) and in vitro AdoVin-TNB

reaction without MTAN (bottom). 5'-methylthioadenosine/S-adenosyl-homocysteine nucleosidase

(MTAN) is used to hydrolyze AdoHcy produced in methylation reactions.

Page 131: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

112

Figure 3.14. Tandem mass spectra of +10 peak (TPMT-AdoHcy) under various CID energies.

Page 132: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

113

Figure 3.15. Tandem mass spectra of AdoHcy under various CID energies.

Page 133: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

114

3.2.4 Ion Mobility and Mobiligram

Ion mobility cell was assembled after native MS and CID/SID dissociation. The

conformation information and drifting time of enzymes and enzyme-ligand complexes can be

collected. Enzymes and enzyme-ligand complexes can be separated by two dimension separation:

mass to charge in mass spectrometry and drift time (size and shape) in ion mobility. Therefore, in

these experiments, ion mobility was also used to monitor the conformation change of TPMT when

binding with AdoVin-TNB and AdoVin-AMBA adduct.

In a mobiligram, the higher intensity of peaks gives a lighter color. In general, the folded

proteins have narrower drift time than their unfolded forms, and the lower charge state of the

precursor helps to maintain the folded structure of TPMT. Precursors with lower charge states

require higher collision energy for fragmentation. As shown in Figure 3.16, high charge states of

TPMT and TPMT-AdoVin-TNB complex (+9 and +10) were unfolded, while TPMT and TPMT-

AdoVin-TNB complex with lower charge states (lower than +9) are more folded. Due to the

unfolded structure of TPMT-AdoVin-TNB complex, AdoVin-TNB adduct was able to dissociate

from TPMT under low dissociation energy (300 eV), as discussed in previous section.

Page 134: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

115

Figure 3.16. Mobiligram of reaction mixture of in vitro AdoVin-TNB adduct formation catalyzed

by thiopurine methyltransferase.

Next, we investigated the different roles of CID and SID on protein conformation change

by ion mobility. Compared to CID fragmentation, SID produces more folded (narrower drift time

range) TPMT fragments (Figure 3.17 and 3.18). This observation supports that SID is more

advantageous to detect the transition state of protein structure change than CID.

Although, ion mobility did not provide as much information as native mass spectrometry

with our goal to identify substrates of enzyme, we still believe that the native mass spectrometry

combined with ion mobility spectrometry can be a powerful tool for exploring structural changes

of unknown enzyme-ligand complexes.

Page 135: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

116

Figure 3.17. Mobiligram of the fragmentation of +10 TPMT-AdoVin-TNB complex under CID

(500 eV).

Page 136: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

117

Figure 3.18. Mobiligram of the fragmentation of +10 TPMT-AdoVin-TNB complex under SID

(500 eV).

Page 137: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

118

3.2.5 Detection of Potential Modification on MTases by AdoVin

AdoVin has been tested with other types of MTases, including catechol O-MTase

(COMT),32-33 protein L-isoaspartate O-MTases (PIMT),34-35 protein arginine N-MTase 1

(PRMT1),36-37 protein N-terminal MTases (NMT).38 However, it is very disappointing that none

of the expected AdoVin bisubstrate-adducts were observed under the catalysis of MTases, nor the

degradation products of AdoVin adducts. Despite the possibility in organic chemistry that AdoVin

(vinyl sulfonium ion) may only show activity to –SH group even under the efficient enzyme

catalysis, these results can be connected to the general concern of AdoMet analogue as discussed

previously: is there any modification of these MTases by AdoVin that inhibit further bisubstrate-

adduct formation? Therefore, potential AdoVin modification of these MTases can be studied into

two directions in the future: 1) non-covalent inhibition by AdoVin; 2) covalent modification on

MTases by AdoVin, since AdoVin may react with the cysteine side chains exposed in the active

sites of enzymes.

Based on the success of native mass spectrometry in the identification of TPMT-substrate

complex, we believe that native mass spectrometry combined with various tandem MS techniques,

such as CID and SID, can be used to identify both non-covalent and covalent modifications on

MTases by AdoVin. Additionally, possible confirmation changes of modified MTases can be

identified by ion mobility spectrometry.

Page 138: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

119

3.3 Conclusions

In summary, a new workflow was developed for analyzing enzyme-substrate complexes,

as well as substrate identification, using native mass spectrometry and ion mobility spectrometry.

Tandem MS with both collision induced dissociation (CID) and surface induced dissociation (SID)

fragmentation was applied to identify substrates bound to enzymes. Using our approach, TPMT

complex with AdoVin bisubstrate-adduct was detectable even in crude E. coli cell lysates. The

combination of ion mobility and native mass spectrometry is considered as a powerful tool to

identify substrates of enzyme and explore structural information of unknown enzyme-ligand

complexes.

Page 139: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

120

3.4 Experimental Section

3.4.1 General Procedures

All chemicals with reagent purity or above were purchased from Sigma and Fisher.

Ultrafiltration was carried out using filters with 30,000 molecular weight cut off (MWCO). The

filters were purchased from Fisher Scientific (EMD Millipore Amicon Ultra 0.5 mL, catalog No.

UFC50VL96).

Prior to analysis by MS, all samples were buffer exchanged into 20 mM ammonium acetate

(pH 8.0) using at least 10 cycles of concentration and dilution in a centrifuge, ultrafiltration

concentrator. Samples were then frozen at -80 °C and thawed immediately prior to analysis.

Further sample dilution was done in 20 mM ammonium acetate, pH 8.0. Direct infusion (2 μL)

was done for each MS analysis.

3.4.2 HPLC Analysis

All AdoVin adducts were separated by HPLC, elution profiles were monitored by UV

absorption at 260 nm and 350 nm. The chromatography was performed on a reversed-phase

column (Apollo, C18, 5μ, 4.6 mm x 150 mm) using 0.1% aqueous formic acid (mobile phase A)

and 0.1% formic acid in acetonitrile (mobile phase B) at a flow rate of 1 mL/min. The gradient

program was initiated with 2% mobile phase B, and increased to 10% mobile phase B over 8 min,

then increased to 30% mobile phase B over 12 min, then increased to 70% mobile phase B over

15 min, followed by an increase to 90 % mobile phase B over 5 min, then a return to 2% mobile

phase B over 1 min, and finally a hold at 2% mobile phase B over 5 min.

Page 140: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

121

3.4.3 Mass Spectrometry

Native mass Spectrometry

The nanoelectrospray experiment was performed on Synapt G2S HDMS (Waters corp.,

Wilmslow, UK) with a customized surface-induced dissociation (SID) device installed before the

ion mobility (IM) cell as previously described.39 Scheme 3.2 shows the travelling-wave ion guide

(TWIG) region in the Waters Synapt G2S HDMS. The arrows indicate the ion trajectory in SID-

IM mode. In SID-IM experiments, precursor ions are released in packets from the trap TWIG at

the beginning of each separation cycle, and are subsequently dissociated by surface collision. The

SID product ions then undergo separation in the ion mobility cell and are transferred into the TOF

analyzer for detection. For CID-IM experiments, the potentials on the lenses are adjusted to allow

a “fly-through” transmission. The precursor ions are dissociated by collisions with argon gas in

the trap TWIG and the product ions are released into the IM cell for mobility separation without

surface collision in the SID region. A delay time of 450 μs in the front of IM cell was used to

ensure all the ions started the separation at the same time point.

Scheme 3.2. Workflow of the Q-IM-TOF instrument. The arrow shows the ion trajectory.

Each sample was filled into a glass capillary pulled using a Sutter Instruments P-97

micropipette puller (Novato, CA) and ionized with a nanoelectrospray source at a voltage of 1.2-

1.5 kV. The sampling cone voltage was set to 20 V and the source offset voltage was set to 20 V.

Page 141: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

122

Other instrument conditions were 5e-3 mbar for the source pressure, 2.0 mL/min gas flow rate to

the trap cell, 120 mL/min gas flow to the helium cell and 60 mL/min gas flow to the ion mobility

cell.

Tandem Mass Spectrometry

Tandem mass spectrometry experiments were performed via dissociation the selected ions

with CID and SID. CID experiment was conducted under trap gas flow rate of 4.0 mL/min and

SID was conducted under trap gas flow rate of 2.0 mL/min. The acceleration voltage in CID and

SID was obtained as described previously.39-40 Briefly, a custom SID device was inserted before

the ion mobility cell of the instrument. The direct current voltages on the SID lenses can be tuned

either to allow a fly-through of the ions for CID experiments or to direct the ions onto the surface

for collision. The current instrument setup is able to perform CID or SID experiments after

quadrupole m/z selection and separate the CID/SID products in the ion mobility cell. The

acceleration voltage in CID is defined by the “Trap CE” setting, which is the potential difference

between the direct current offsets of the quadrupole and the trap traveling wave ion guide before

the ion mobility cell. The acceleration voltage in SID is defined by the potential difference between

the direct current offset of the trap traveling wave ion guide and the surface, which can be adjusted

using the “Trap bias” setting. The acceleration voltage multiplied by the charge state of the selected

ion provides collision energy in eV.

Page 142: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

123

3.4.4 Preparation of in vitro Enzyme-Substrate Complex

Preparation of in vitro Complex between TPMT and AdoVin-TNB Adduct

The reaction contained 50 mM potassium phosphate, pH 8.0, 50 mM KCl, 25 mM MgCl2,

10 mM ATP, 1 mM vinthionine, 500 μM TNB, 2 mM TCEP, 3.5 μM MTAN and 100 μM TPMT.

The reaction was initiated with 200 μM MAT and incubated at 37 °C for 3 hours.

Preparation of in vitro Complex between TPMT and AdoVin-AMBA Adduct

The reaction contained 50 mM potassium phosphate, pH 8.0, 50 mM KCl, 25 mM MgCl2,

10 mM ATP, 1 mM vinthionine, 500 μM AMBA, 2 mM TCEP, 3.5 μM MTAN, and 100 μM

TPMT. The reaction was initiated with 200 μM MAT and incubated at 37 °C for 3 hours.

3.4.5 Preparation of Enzyme-Substrate Complex in Crude Biological Samples

E. coli cell lysates with and without IPTG induction were prepared as described in general

procedure without purification. The concentration of TPMT was estimated as 336 μM in induced

lysate, and about 4.9 μM in unstimulated lysate.

The reaction (200 μL) contained 50 mM potassium phosphate, pH 8.0, 50 mM KCl, 25

mM MgCl2, 20 mM ATP, 1 mM vinthionine, 500 μM TNB, 2 mM TCEP, 3.5 μM MTAN and 100

μL TPMT cell lysate. The reaction was initiated with 200 μM MAT and incubated at 37 °C for 3

hours.

Page 143: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

124

3.5 References

1. Fenn, J. B.; Mann, M.; Meng, C. K.; Wong, S. F.; Whitehouse, C. M., Electrospray

ionization for mass spectrometry of large biomolecules. Science 1989, 246 (4926), 64-71.

2. Heck, A. J., Native mass spectrometry: a bridge between interactomics and structural

biology. Nat Methods 2008, 5 (11), 927-33.

3. Snijder, J.; Rose, R. J.; Veesler, D.; Johnson, J. E.; Heck, A. J., Studying 18 MDa virus

assemblies with native mass spectrometry. Angew Chem Int Ed Engl 2013, 52 (14), 4020-

3.

4. Rajabi, K.; Ashcroft, A. E.; Radford, S. E., Mass Spectrometric Methods to Analyse the

Structural Organization of Macromolecular Complexes. Methods 2015.

5. Hoffmann, E. d.; Charette, J. J.; Stroobant, V., Mass spectrometry : principles and

applications. Wiley ;Masson: Chichester ; New York Paris, 1996; p xii, 340 pages.

6. Molina, H.; Matthiesen, R.; Kandasamy, K.; Pandey, A., Comprehensive comparison of

collision induced dissociation and electron transfer dissociation. Anal Chem 2008, 80 (13),

4825-35.

7. Yates, J. R.; Ruse, C. I.; Nakorchevsky, A., Proteomics by mass spectrometry: approaches,

advances, and applications. Annu Rev Biomed Eng 2009, 11, 49-79.

8. Jones, C. M.; Beardsley, R. L.; Galhena, A. S.; Dagan, S.; Cheng, G.; Wysocki, V. H.,

Symmetrical gas-phase dissociation of noncovalent protein complexes via surface

collisions. J Am Chem Soc 2006, 128 (47), 15044-5.

9. von Helden, G.; Wyttenbach, T.; Bowers, M. T., Conformation of macromolecules in the

gas phase: use of matrix-assisted laser desorption methods in ion chromatography. Science

1995, 267 (5203), 1483-5.

10. Badman, E. R.; Hoaglund-Hyzer, C. S.; Clemmer, D. E., Monitoring structural changes of

proteins in an ion trap over approximately 10-200 ms: unfolding transitions in cytochrome

c ions. Anal Chem 2001, 73 (24), 6000-7.

11. Koeniger, S. L.; Merenbloom, S. I.; Clemmer, D. E., Evidence for many resolvable

structures within conformation types of electrosprayed ubiquitin ions. J Phys Chem B 2006,

110 (13), 7017-21.

12. Loo, J. A., Studying noncovalent protein complexes by electrospray ionization mass

spectrometry. Mass Spectrom Rev 1997, 16 (1), 1-23.

Page 144: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

125

13. Loo, J. A.; Berhane, B.; Kaddis, C. S.; Wooding, K. M.; Xie, Y.; Kaufman, S. L.;

Chernushevich, I. V., Electrospray ionization mass spectrometry and ion mobility analysis

of the 20S proteasome complex. J Am Soc Mass Spectrom 2005, 16 (7), 998-1008.

14. Hogan, C. J., Jr.; Kettleson, E. M.; Ramaswami, B.; Chen, D. R.; Biswas, P., Charge

reduced electrospray size spectrometry of mega- and gigadalton complexes: whole viruses

and virus fragments. Anal Chem 2006, 78 (3), 844-52.

15. Kaufman, S. L.; Skogen, J. W.; Dorman, F. D.; Zarrin, F.; Lewis, K. C., Macromolecule

analysis based on electrophoretic mobility in air: globular proteins. Anal Chem 1996, 68

(11), 1895-904.

16. Bacher, G.; Szymanski, W. W.; Kaufman, S. L.; Zollner, P.; Blaas, D.; Allmaier, G.,

Charge-reduced nano electrospray ionization combined with differential mobility analysis

of peptides, proteins, glycoproteins, noncovalent protein complexes and viruses. J Mass

Spectrom 2001, 36 (9), 1038-52.

17. Kaddis, C. S.; Loo, J. A., Native protein MS and ion mobility large flying proteins with

ESI. Anal Chem 2007, 79 (5), 1778-84.

18. Fenn, L. S.; McLean, J. A., Biomolecular structural separations by ion mobility-mass

spectrometry. Anal Bioanal Chem 2008, 391 (3), 905-9.

19. Ruotolo, B. T.; Benesch, J. L.; Sandercock, A. M.; Hyung, S. J.; Robinson, C. V., Ion

mobility-mass spectrometry analysis of large protein complexes. Nat Protoc 2008, 3 (7),

1139-52.

20. Kuhn, M. L.; Majorek, K. A.; Minor, W.; Anderson, W. F., Broad-substrate screen as a

tool to identify substrates for bacterial Gcn5-related N-acetyltransferases with unknown

substrate specificity. Protein Sci 2013, 22 (2), 222-30.

21. Mishra, A. K.; Lambright, D. G., High-Throughput Assay for Profiling the Substrate

Specificity of Rab GTPase-Activating Proteins. Methods Mol Biol 2015, 1298, 47-60.

22. Vetting, M. W.; LP, S. d. C.; Yu, M.; Hegde, S. S.; Magnet, S.; Roderick, S. L.; Blanchard,

J. S., Structure and functions of the GNAT superfamily of acetyltransferases. Arch

Biochem Biophys 2005, 433 (1), 212-26.

23. Xu, T.; Zhang, L.; Wang, X.; Wei, D.; Li, T., Structure-based substrate screening for an

enzyme. BMC Bioinformatics 2009, 10, 257.

24. Warren, G. L.; Andrews, C. W.; Capelli, A. M.; Clarke, B.; LaLonde, J.; Lambert, M. H.;

Lindvall, M.; Nevins, N.; Semus, S. F.; Senger, S.; Tedesco, G.; Wall, I. D.; Woolven, J.

Page 145: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

126

M.; Peishoff, C. E.; Head, M. S., A critical assessment of docking programs and scoring

functions. J Med Chem 2006, 49 (20), 5912-31.

25. Mohan, V.; Gibbs, A. C.; Cummings, M. D.; Jaeger, E. P.; DesJarlais, R. L., Docking:

successes and challenges. Curr Pharm Des 2005, 11 (3), 323-33.

26. Mulholland, A. J., Modelling enzyme reaction mechanisms, specificity and catalysis. Drug

Discov Today 2005, 10 (20), 1393-402.

27. Zhang, Y. L.; Yao, Z. J.; Sarmiento, M.; Wu, L.; Burke, T. R., Jr.; Zhang, Z. Y.,

Thermodynamic study of ligand binding to protein-tyrosine phosphatase 1B and its

substrate-trapping mutants. J Biol Chem 2000, 275 (44), 34205-12.

28. Flint, A. J.; Tiganis, T.; Barford, D.; Tonks, N. K., Development of "substrate-trapping"

mutants to identify physiological substrates of protein tyrosine phosphatases. Proc Natl

Acad Sci U S A 1997, 94 (5), 1680-5.

29. Zhang, W.; Savelieva, K. V.; Tran, D. T.; Pogorelov, V. M.; Cullinan, E. B.; Baker, K. B.;

Platt, K. A.; Hu, S.; Rajan, I.; Xu, N.; Lanthorn, T. H., Characterization of PTPRG in

knockdown and phosphatase-inactive mutant mice and substrate trapping analysis of

PTPRG in mammalian cells. PLoS One 2012, 7 (9), e45500.

30. Fukada, M.; Kawachi, H.; Fujikawa, A.; Noda, M., Yeast substrate-trapping system for

isolating substrates of protein tyrosine phosphatases: Isolation of substrates for protein

tyrosine phosphatase receptor type z. Methods 2005, 35 (1), 54-63.

31. Woodson, L. C.; Weinshilboum, R. M., Human kidney thiopurine methyltransferase.

Purification and biochemical properties. Biochem Pharmacol 1983, 32 (5), 819-26.

32. Mannisto, P. T.; Kaakkola, S., Catechol-O-methyltransferase (COMT): biochemistry,

molecular biology, pharmacology, and clinical efficacy of the new selective COMT

inhibitors. Pharmacol Rev 1999, 51 (4), 593-628.

33. Axelrod, J.; Tomchick, R., Enzymatic O-methylation of epinephrine and other catechols. J

Biol Chem 1958, 233 (3), 702-5.

34. Farrar, C.; Clarke, S., Altered levels of S-adenosylmethionine and S-

adenosylhomocysteine in the brains of L-isoaspartyl (D-Aspartyl) O-methyltransferase-

deficient mice. J Biol Chem 2002, 277 (31), 27856-63.

35. Alfaro, J. F.; Gillies, L. A.; Sun, H. G.; Dai, S.; Zang, T.; Klaene, J. J.; Kim, B. J.;

Lowenson, J. D.; Clarke, S. G.; Karger, B. L.; Zhou, Z. S., Chemo-enzymatic detection of

Page 146: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

127

protein isoaspartate using protein isoaspartate methyltransferase and hydrazine trapping.

Anal Chem 2008, 80 (10), 3882-9.

36. Gui, S.; Wooderchak, W. L.; Daly, M. P.; Porter, P. J.; Johnson, S. J.; Hevel, J. M.,

Investigation of the molecular origins of protein-arginine methyltransferase I (PRMT1)

product specificity reveals a role for two conserved methionine residues. J Biol Chem 2011,

286 (33), 29118-26.

37. Gui, S.; Wooderchak-Donahue, W. L.; Zang, T.; Chen, D.; Daly, M. P.; Zhou, Z. S.; Hevel,

J. M., Substrate-induced control of product formation by protein arginine

methyltransferase 1. Biochemistry 2013, 52 (1), 199-209.

38. Webb, K. J.; Lipson, R. S.; Al-Hadid, Q.; Whitelegge, J. P.; Clarke, S. G., Identification of

protein N-terminal methyltransferases in yeast and humans. Biochemistry 2010, 49 (25),

5225-35.

39. Zhou, M.; Dagan, S.; Wysocki, V. H., Protein subunits released by surface collisions of

noncovalent complexes: nativelike compact structures revealed by ion mobility mass

spectrometry. Angew Chem Int Ed Engl 2012, 51 (18), 4336-9.

40. Zhou, M.; Jones, C. M.; Wysocki, V. H., Dissecting the large noncovalent protein complex

GroEL with surface-induced dissociation and ion mobility-mass spectrometry. Anal Chem

2013, 85 (17), 8262-7.

Page 147: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

128

Chapter 4: Conclusions and Future Directions

In these two projects, S-adenosyl-vinthionine (AdoVin) serves as a new S-adenosyl-

methionine (AdoMet) analogue, which forms a bisubstrate-adduct in situ with nucleophilic

substrates of methyltransferases. Due to the tight binding between the AdoVin bisubstrate-adduct

and TPMT, TPMT and the thiol substrate were enriched and the TPMT-bisubstrate-adduct

complexes were characterized by using HPLC-UV and LC-MS. Chapter 2 discussed the progress

of the functional activity of AdoVin working with TPMT and its thiol substrates. Moreover, a

previously unknown substrate of TPMT, 2-amino-5-mercaptobenzoic acid (AMBA), has been

discovered and identified. More details about future research on the application of AdoVin will be

discussed in this chapter.

An additional improvement in this methodology is the ability to observe the adduct-enzyme

complex in a crude sample by native mass spectrometry without any purification step. The details

have been covered in Chapter 3 and the future directions of substrate discovery and identification

will be discussed in this chapter.

Page 148: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

129

4.1. AdoVin Bisubstrate-adduct Formation in Whole Cells

The data reported previously were from either in vitro or ex vivo studies by ruling out the

complexity existing within the living cells. It is possible to expand the utility of AdoVin adducts

by employing this technique in whole cells, which may yield new insights into MTases not

reflected by the results of in vitro or ex vivo experiments. There is also potential to use AdoVin as

a probe to fish out unknown endogenous substrates of MTases, and even unknown MTases by

known substrates. However, a more complicated environment presents more challenges, i.e.

specificity of AdoMet synthetase in certain cells, competition between vinthionine and methionine,

as well as competition between AdoVin and AdoMet (Scheme 4.1).

Scheme 4.1. in vivo methylation and bisubstrate-adduct formation with AdoVin starting from

methionine and vinthionine, respectively.

Page 149: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

130

Though previous research reported that vinthionine can be utilized by E. coli MAT,2 the

specificity of endogenous MAT in E. coli strains as well as other cell lines needs to be addressed,

because the activity of MAT using vinthionine may not be sufficient for effective labeling.

Additionally, vinthionine may not be able to compete with methionine in a cellular context. To

address these issues, MAT enzyme can be engineered to obtain desired activities and selectivities,

and then expressed in E. coli and other cells of interest.3

4.2. Affinity Capturing Unknown Methyltransferases

Another potentially powerful application of our approach is identifying unknown MTases

using the known substrates or methylation products. One target for this approach is human thiol

S-methyltransferase (TMT, EC 2.1.1.9).4-5 TMT is a membrane-bound enzyme that catalyzes the

S-methylation of aliphatic sulfhydryl groups and plays a key role in the metabolism of many

sulfhydryl drugs, including antihypertensive drug captopril, D-penicillamine, and N-acetylcysteine

through S-methylation (Scheme 4.2).6-7 However, the pharmacokinetics of each drug could vary

depending on the individual TMT isoform, and genetic polymorphism appears to be a determining

factor for the variation of activities.8-9 There are two reasons to choose human TMT as the target:

1) S-MTases are the obvious targets for expanding the study of AdoVin activity, given the

hypothesis that AdoVin shows good activity towards -SH group (Michael addition); 2) even

though the function of human TMT has been studied, its gene and sequence remain undetermined,

due to the difficulty of isolating and purifying a membrane-bound enzyme.

AdoVin has shown the activity TPMT, and we believe that AdoVin bisubstrate-adduct may

be formed and bind tightly with TMT. The possible persistent interaction between the bisubstrate-

Page 150: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

131

adduct and TMT would make AdoVin an efficient probe to affinity enrich and isolate TMT.

Modifying AdoVin or the nucleophilic substrates with affinity tags, such as biotin tag would

facilitate this enrichment. Thus, the isolated enzyme may be sequenced by standard peptide

mapping and protein mass spectrometry.

Scheme 4.2. Proposed AdoVin bisubstrate-adduct formation catalyzed by TMT and proposed

isolation of TMT catalyzed by affinity tag (red) on AdoVin bisubstrate-adduct.

Page 151: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

132

4.3. Substrate Identification for Other MTases using Native Mass Spectrometry via

Enhanced Binding without Formation of Covalent Adducts

The elucidation of methyltransferase substrate specificity is hampered by the transient

interaction (weak binding) between the nucleophilic substrates and the enzymes, in other words,

the binary complex between MTase and substrate as well as the tertiary complex between MTase,

substrate, and AdoMet, may not be persistent enough for the detection via Mass Spectrometry.

Therefore, tighter binding is required to establish the direction connection between substrates and

enzymes. Another method for creating direct connection between the substrates and MTases is to

strengthen the binding between nucleophilic substrates and MTases by AdoVin (Scheme 4.3).

“Tethering” to AdoVin allows the enhanced interaction between the nucleophilic substrates and

MTases to be observed via Mass Spectrometry. This hypothesis expands another possible utility

of AdoVin for substrate identification of MTases. Additionally, similar with the “substrate-

trapping” system,10-11 the enhanced binding between substrate and enzyme is also perfect to

investigate the transition state of enzyme catalyzed reactions.

Scheme 4.3. Substrate binding in methylation using AdoMet (top) and proposed binding between

nucleophilic substrates and methyltransferases enhanced by AdoVin (bottom).

Page 152: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

133

4.4. Facile Generation of Bisubstrate-adduct for Structural Investigation

It is possible that AdoVin only shows activity to S-MTases via addition reaction with the

thiol group (-SH). For other type of MTases, a potentially useful methodology would be to

derivatize the nucleophilic groups, such as amine (–NH2) and hydroxyl (–OH), to thiols (-SH). As

shown in Scheme 4.4, the modified substrates are expected to react with AdoVin to form tightly

binding bisubstrate-adducts catalyzed by their corresponding MTases (O-MTases, N-MTases or

others), for example, though lysine (-NH2) may not be active to AdoVin, its corresponding lysyl

thiol analog (-SH) could react with AdoVin and form bisubstrate adduct catalyzed by protein lysine

N-MTases (PKMTs) (Scheme 4.4 b). Thusly, AdoVin can be used to generate bisubstrate-adduct

inhibitors for other type of MTases via the change of various MTases into putative S-MTases.

One utility of the multisubstrate adduct inhibitor approach is to provide essential structural

knowledge of enzyme via enzyme co-crystallization.12 It is expected that co-crystallization of

MTases with AdoVin bisubstrate-adducts could elucidate structural information of enzyme-

substrate interactions. Additionally, using this methodology, AdoVin could contribute to the

generation of specific and strong bisubstrate-adduct inhibitors for many MTases, facilitating the

structural investigation of MTases.

Page 153: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

134

Scheme 4.4. (a) Potential generation of bisubstrate-adduct inhibitors by derivatizing other

nucleophilic substrates (such as -NH2 and -OH) to thiol substrates (-SH); (b) modification of –NH2

group to –SH in lysine for protein lysine N-MTases.

Page 154: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

135

4.5. References

1. Wan, W.; Zhao, G.; Al-Saad, K.; Siems, W. F.; Zhou, Z. S., Rapid screening for S-

adenosylmethionine-dependent methylation products by enzyme-transferred isotope

patterns analysis. Rapid Commun Mass Spectrom 2004, 18 (3), 319-24.

2. Kim, H. J.; Balcezak, T. J.; Nathin, S. J.; McMullen, H. F.; Hansen, D. E., The use of a

spectrophotometric assay to study the interaction of S-adenosylmethionine synthetase with

methionine analogues. Anal Biochem 1992, 207 (1), 68-72.

3. Wang, R.; Zheng, W.; Luo, M., A sensitive mass spectrum assay to characterize engineered

methionine adenosyltransferases with S-alkyl methionine analogues as substrates. Anal

Biochem 2014, 450, 11-9.

4. Glauser, T. A.; Kerremans, A. L.; Weinshilboum, R. M., Human hepatic microsomal thiol

methyltransferase. Assay conditions, biochemical properties, and correlation studies. Drug

Metab Dispos 1992, 20 (2), 247-55.

5. Glauser, T. A.; Nelson, A. N.; Zembower, D. E.; Lipsky, J. J.; Weinshilboum, R. M.,

Diethyldithiocarbamate S-methylation: evidence for catalysis by human liver thiol

methyltransferase and thiopurine methyltransferase. J Pharmacol Exp Ther 1993, 266 (1),

23-32.

6. Keith, R. A.; Jardine, I.; Kerremans, A.; Weinshilboum, R. M., Human erythrocyte

membrane thiol methyltransferase. S-methylation of captopril, N-acetylcysteine, and 7

alpha-thio-spirolactone. Drug Metab Dispos 1984, 12 (6), 717-24.

7. Kazui, M.; Hagihara, K.; Izumi, T.; Ikeda, T.; Kurihara, A., Hepatic microsomal thiol

methyltransferase is involved in stereoselective methylation of pharmacologically active

metabolite of prasugrel. Drug Metab Dispos 2014, 42 (7), 1138-45.

8. Roediger, W. E.; Babidge, W. J., Thiol methyltransferase activity in inflammatory bowel

disease. Gut 2000, 47 (2), 206-10.

9. Price, R. A.; Keith, R. A.; Spielman, R. S.; Weinshilboum, R. M., Major gene

polymorphism for human erythrocyte (RBC) thiol methyltransferase (TMT). Genet

Epidemiol 1989, 6 (6), 651-62.

10. Flint, A. J.; Tiganis, T.; Barford, D.; Tonks, N. K., Development of "substrate-trapping"

mutants to identify physiological substrates of protein tyrosine phosphatases. Proc Natl

Acad Sci U S A 1997, 94 (5), 1680-5.

Page 155: Identifying unknown substrates of S-adenosyl-methionine ...rx915f228/... · dissociation (CID) and surface induced dissociation (SID), in a single run. To the best of our knowledge,

136

11. Zhang, Y. L.; Yao, Z. J.; Sarmiento, M.; Wu, L.; Burke, T. R., Jr.; Zhang, Z. Y.,

Thermodynamic study of ligand binding to protein-tyrosine phosphatase 1B and its

substrate-trapping mutants. J Biol Chem 2000, 275 (44), 34205-12.

12. Le Calvez, P. B.; Scott, C. J.; Migaud, M. E., Multisubstrate adduct inhibitors: drug design

and biological tools. J Enzyme Inhib Med Chem 2009, 24 (6), 1291-318.