PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 5/08 KIProj/AccaPresICIRCIDateDuehdd THE SYNTHESIS AND EVALUATION OF DIAzo FUNCTIONALIZED SOLID SUPPORTS AND THEIR APPLICATION TOWARD THE IDENTIFICATION OF PHOSPHORYLATED PROTEINS By Samantha M. Frawley Cass A DISSERTATION Submitted to Michigan State University In partial fulfillment of the requirements For the degree of DOCTOR OF PHILOSOPHY Chemistry 2009 In) SI ABSTRACT THE SYNTHESIS AND EVALUATION OF DIAZO FUNCTIONALIZED SOLID SUPPORTS AND THEIR APPLICATION TOWARD THE IDENTIFICATION OF PHOSPHORYLATED PROTEINS BY Samantha M. Frawley Cass Many cellular processes are regulated by the reversible process of protein phosphorylation/dcphosphorylation on serine, threonine and tyrosine residues. Deregulation of the signal transduction cascade upsets this well-balanced system and is implicated in diseases such as cancer, type II diabetes, cystic fibrosis, and Alzheimer’s disease. Even though the human genome maps out the structure and sequence of our genes, it does not provide any insight into post-translational modifications, such as phosphorylation, which regulate cell function. In order to understand cellular regulation on a molecular level, post-translational modifications need to be identified and characterized. Unfortunately, proteomic techniques relevant to the elucidation of signal transduction pathways have lagged in development in comparison to genomic technologies. Despite recent advancements in mass spectrometry techniques, characterization of protein phosphorylation is often hampered by their low-abundance and signal suppression. Enrichment of phosphorylated peptides/proteins prior to their introduction into a mass spectrometer can therefore significantly facilitate phosphoprotein analysis. Here, we describe a method for the enrichment of phosphorylated serinelthreonine and tyrosine containing substrates using a II P .4 F a div £5.60” I 44. ‘ ”Pip v 4| unique solid phase enrichment technique. The method takes advantage of diazo-functionalized solid phase resins, which react with the phosphate moiety allowing for non-phosphorylated peptides to be efficiently removed by filtration. Liberation of the phosphorylated substrates from the resin using aqueous acid provides the products, which can be readily analyzed and sequenced by tandem mass spectrometry. The technique described herein has successfully enriched the phosphorylated substrates from B-casein and ovalbumin digests, in low- picomolar quantities and is found to be competitive with the current commercially available enrichment methods. Further application of this phosphopeptide enrichment technique towards a purified cellular extract will also be discussed. Copyright by SAMANTHA M. F RAWLEY CASS 2009 I dedicate this dissertation to my parents. To my mother for being an endless supply of faith and encouragement and to my father (and fellow chemist); this work is proof that the apple did not fall far from the tree... iv eh... :PA ‘. u I u 1 .‘II If.“ ACKNOWLEDGEMENTS There are many people that have contributed to the completion of this dissertation. I would like to take this Opportunity to say thank you to all those involved with my project and/or development of my graduate career. First and foremost, I would like to thank my advisor Dr. Jetze J. Tepe. Under his guidance I believe that I have grown to become an independent researcher. He motivated and encouraged me to continue to think critically about my own project and I don’t believe I would have grown as a researcher the way I have without him as my advisor. I would also like to thank Dr. Gavin Reid for all of his guidance in the biological mass spectrometry realm. I have learned a great deal from him these past few years and I truly appreciate all the time and effort he spent with me to help improve as a researcher. Gavin is very vocal about exactly what he thinks and I appreciated all of his “this is how it is” comments because by knowing how he felt, I could take his advice to improve in my writing, researching, thought process, etc. The rest of my guidance committee all deserves my thanks as well. Dr. Greg Baker, I am not sure if you remember this, but the first semester here I went to talk to you when l was not sure if I was going to stay because my grades were not the best. I remember you saying- “Just keep reading and working hard. I know you can make it." The faith you had in me so early on in my graduate vi career, I will never forget. I also would like thank Dr. Babak Borhan for his insight into many different questions I have had over the years concerning both my research project and graduate classes. Babak, whenever I have had a question for you, instead of just answering it yourself, you helped lead me to answering it for myself; that’s what academics are all about. Dr. Pinnavaia, l have Ieamed a lot about materials science as a result of our ongoing collaborations and I appreciate all Of the time that you have taken to help improve both the materials that l have used and my knowledge of them. I really want to take the time to thank all of my Iabmates; you guys have been my support system for the past six years. I feel very privileged that I worked with the people I did. I appreciate all of the camaraderie and teamwork with all of my “bio labmates”. Teri, I am so glad that you were there to train me on all the ins and outs of the bio lab. You are a terrific teacher; you don’t hover, which I like, but you were always there to answer any and all questions that I had. Daljinder, thank you for your advice and support (both research and personal), I appreciate it more than your know. Thu aka ‘the bio-lab organizer’, you have worked so hard to make it easier on all of the bio-lab members to find and use whatever it was we were looking for with your organization skills! Thank you for your years of friendship, conversations and of course egg rolls. I will miss all of the above. Amanda, we have had many experiences over the last few years; from ASMS to mini meetings. You have been a great friend as well as a research associate. We have spent endless hours bouncing ideas off one another and you are always there to help whenever I need it! Thank you for vii pl .F J19. I being there. I can’t imagine what my graduate school experience would have been without you! Additionally, I would like to thank all past and present Tepe group members, in particular, Chris and Jason. They have always been there for me when I needed them and I am privileged to call them my friends. We have all shared some memorable moments here in graduate school. I won’t go into listing them-to protect both the innocent and the guilty. Jason (Stinky), you always knew what to say to make me feel better when things were going bad. Your ability to always make me laugh helped get me through these past six years! I have been, and will always be, very grateful for your friendship. Chris (PhePhe), you are like the brother I never had (and never really wanted). What will I do without your daily teasing?! In all seriousness, I am really going to miss talking and laughing with you on a daily basis. You have been one of the best friends a person could ask for. I would also like to thank all my friends and family. Particularly my parents, who have always supported and believed in me. I especially want to thank my husband, Kyle. You have been so patient with me through this whole process. From dealing with my moods to fixing an endless number of dinners, your support has only made me love you more. I am looking forward to spending the rest of our lives together. Just remember, you have to call me “Doctor” now. viii CHAD RA? 31v. LIST OF TABLES ................................................................................. xii LIST OF FIGURES ............................................................................... xiii LIST OF SCHEMES ............................................................................................ xviii KEY TO SYMBOLS OR ABBREVIATIONS .................................................. xx CHAPTER 1 PROTEIN PHOSPHORYLATION: PROJECT BACKGROUND ............................. 1 1.1 Introduction to post-translational modifications ................................. 1 1.2 Introduction to the study of protein phosphorylation .......................... 3 1.3 Detection Of phosphorylated proteins ................................................ 9 a. 32F radiolabeling ..................................................................... 9 b. Gel electrophoresis/phosphoprotein staining ........................ 10 c. Phosphospecific antibodies .................................................. 12 1.4 Enrichment of phosphorylated proteins ........................................... 13 a. Immunoprecipitation ............................................................. 15. b. Affinity chromatography ........................................................ 16 i. Immobilized metal affinity chromatography ..................... 17 ii. Strong ion exchange chromatography ............................ 21 iii. Metal oxide affinity chromatography ................................ 22 c. B-elimination-phosphate modification ................................... 25 i. Chemical tagging ............................................................. 25 ii. Proteolytic phosphopeptide mapping .............................. 30 d. Covalent phosphate addition to a solid support .................... 32 1.5 Quantitative phosphoproteomic analysis ......................................... 35 a. Stable isotope labeling In cell culture (SILAC) ...................... 36 b. Isotope labeling/isotope affinity tags ..................................... 39 c. Phosphospecific staining ...................................................... 43 1.6 Global phosphoproteomic studies ................................................... 45 a. Using IMAC ........................................................................... 46 b. Using TIOz ............................................................................. 49 0. Additional technologies ......................................................... 51 1.7 Current work .................................................................................... 54 1.8 References ...................................................................................... 56 ix TABLE OF CONTENTS CHAPTERZ SYNTHESIS OF DIAZO FUNCTIONALIZED SOLID SUPPORTS AND QUANTIFICATION OF THE DEGREE OF FUNCTIONALIZATION ..................... 77 2.1 Reactivity of diazo materials with Bronsted acids ........................... 77 2.2 Concept of diazo-phosphate binding ............................................... 80 2.3 Original synthesis of diazo functionalized solid support .................. 83 2.4 Problems associated with original studies ....................................... 86 2.5 Improved synthesis of diazo acetate solid support .......................... 88 2.6 Application of improved synthesis to functionalize additional diazo supports ........................................................................................... 93 2.7 Synthesis of diazo ketone solid supports ........................................ 96 2.8 Synthesis of phenyl diazo methane solid supports .......................... 97 2.9 Quantification of solid support diazo functionalization ..................... 98 2.10 Experimental procedures ............................................................... 104 2.11 Appendix ........................................................................................ 110 2.12 References .................................................................................... 1 34 CHAPTER 3 APPLICATION OF DIAZO SOLID SUPPORTS IN THE ENRICHMENT OF PHOSPHOPEPTIDES ........................................................................................ 139 3.1 Introduction to diazo solid support phosphopeptide enrichment ..................................................................................... 139 3.2 Preliminary phosphopeptide enrichment from a peptide mixture and a protein digest .............................................................................. 141 3.3 Evaluation of synthetic phosphopeptide enrichment by diazo supports ......................................................................................... 143 a. radiolabeled quantification .................................................. 143 b. MS-based quantification ..................................................... 147 3.4 Scope of diazo ketone solid support ............................................. 152 3. proteins ............................................................................... 152 b. residues .............................................................................. 158 3.5 Experimental procedures ............................................................... 162 3.6 Appendix ........................................................................................ 1 69 3.7 References .................................................................................... 205 CHAPTER 4 APPLICATION OF PHOSPHOPEPTIDE ENRICHMENT ON A BIOLOGICAL SAMPLE CONTAINING NF-KB PROTEIN-PRELIMINARY WORK AND FUTURE PROJECT DIRECTIONS .................................................................................... 208 4.1 Impact Of application towards biological samples ......................... 208 4.2 Introduction to NF-KB ..................................................................... 211 4.3 General signaling pathway of NF-KB ............................................. 214 4.4 Phosphorylation events in NF-KB and their impact in cancer therapy ........................................................................................... 217 4.5 Determination of phosphorylation events in NF-KB pathway ......... 218 a. previous work ...................................................................... 218 x 4.6 4.7 4.8 4.9 4.10 4.11 4.12 b. proposed work using our approach .................................... 223 Activation and purification of NF-KB .............................................. 228 Enrichment attempts made using diazo ketone solid supports ..... 235 Future project directions ................................................................ 242 a. application on cellular proteins ........................................... 242 b. improvement on enrichment efficiency ............................... 246 i. solution-phase enrichment approach ............................ 246 ii. preparation of new diazo solid support ......................... 251 Conclusions ................................................................................... 254 Experimental procedures ............................................................... 256 Appendix ........................................................................................ 266 References ................................................................................... 278 xi hr. - .- an; n- a 5. am VU A I a 3i. LIST OF TABLES Table 1-1. Summary Of some select global phosphoproteome studies and the enrichment tools used ...................................................................................... 51-52 Table 2-1. Average percent diazo functionalization results ............................ 103 Table 3-1. Enrichment recovery results using a 32P labeled peptide .............. 146 Table 3-2. Average enrichment recovery for each diazo functionalized solid support ......................................................................................................... 149-150 Table 4-1. IEF result comparison between unmodified and phosphorylated peptides from a digested whole cell protein extract ............................................ 221 Table 4-2. IEF result comparison between unmodified and phosphorylated proteins from a whole cell protein extract ............................................................ 221 Table 4-3. Predicted peptide masses in a p65 tryptic digest containing known phosphorylation sites .......................................................................................... 245 xii Figure 1-1. Figure 1-2. Figure 1-3. Figure 1-4. Figure 1-5. Figure 1-6. Figure 1-7. Figure 2-1. Figure 2-2. Figure 2-3. Figure 2-4. Figure 2-5. Figure 2—6. Figure 2-7. Figure 2-8. Figure 2-9. Figure 2-10. Figure 2-11. Figure 2-12. Figure 2-13. Figure 2-14. LIST OF FIGURES Top five most common post-translational modifications ................. 2 Phosphoprotein enrichment and detection technologies ................ 8 Coordination Of metal cations and IMAC chelating reagents ........ 18 Carboxylic vs. Phosphoric acid Ti02 coordination comparison....23 General concept of SILAC approach for MS quantification .......... 36 General approach for MS quantification from EGF stimulation....38 Global phosphoproteomic analysis approach using IMAC ........... 46 Set-up for diazotization of amino-functionalized resin .................. 87 Prepared diazo-functionalized solid supports ............................... 95 IR of a-diazo acetate Wang resin using HONO gas 1 ................ 110 IR of a-diazo acetate Wang resin using DIC coupling ................ 111 IR of a-diazo acetate Wang resin using EDCI coupling .............. 112 IR of a-diazo acetate Wang resin 2-1 ......................................... 113 IR of a-diazo acetate Wang resin 2-2 ......................................... 114 IR of a-diazo amide tentagel 2-3 ................................................. 115 IR of a-diazo amide megabead 2-4 ............................................ 116 IR Of PEGA a-diazo amide 2-5 ................................................... 117 IR of PEGA HMPA a—diazo amide 2-6 ........................................ 118 IR of PEGA HMPA a-diazo amide dendn’mer 2-7 ....................... 119 IR of 4-butyl a—diazo acetate polystyrene 2-8 ............................. 120 IR of a-diazo acetate polystyrene 2-9 ......................................... 121 xiii Figure 2-15. IR Of NovaSyn a—diazo acetate polystyrene 2-10 ....................... 122 Figure 2-16. IR of NovaSyn TG HMP a-diazo acetate polystyrene 2-11 ........ 123 Figure 2-17. IR of NovaSyn a—diazo anhydn'de polystyrene 2-12 ................... 124 Figure 2-18. IR of vicinal a-diazo acetate silica 2-13 ...................................... 125 Figure 2-19. IR of NHNMPTS a-diazo amide silica 2-14 ................................. 126 Figure 2-20. IR of a-diazo ketone polystyrene 2-15 ........................................ 127 Figure 2-21. IR of a-diazo ketone polystyrene 2-16 ........................................ 128 Figure 2-22. IR of a-diazo ketone silica 2-17 .................................................. 129 Figure 2-23. IR of benzyloxy phenyl diazo methane polystyrene 2-18 ............ 130 Figure 2-24. IR of phenyl diazo methane polystyrene 2-19 ............................ 131 Figure 2-25. IR of benzyloxy phenyl diazo methane polystyrene 2-20 ............ 132 Figure 2-26. Fmoc—glycine standard curve ...................................................... 133 Figure 3-1. Enrichment recovery quantification procedure using 32P-labeled peptide ................................................................................................................ 145 Figure 3-2. Enrichment of ovalbumin phosphopeptides ................................ 154 Figure 3—3. Enrichment of B-casein phosphopeptides ................................... 157 Figure 3-4. Enrichment of phosphotyrosine peptide (angiotensin II) ............. 159 Figure 3-5. Enrichment of a synthetic phosphothreonine peptide ................. 161 Figure 3—6. Enrichment of 25 pmol angiotensin II, acidic cleavage ............... 169 Figure 3-7. Enrichment of 25 pmol angiotensin II, basic cleavage ................ 170 Figure 3-8. Enrichment of 100 pmol B-casein non-sequencing grade trypsin digest ................................................................................................................... 171 Figure 3-9. Enrichment of 100 pmol B-casein sequencing grade trypsin digest, acidic cleavage ........................................................................................ 172 xiv Figure 3-10. Enrichment Of 100 pmol B-casein sequencing grade trypsin digest, basic cleavage ......................................................................................... 173 Figure 3-11. MS of CHKtide standard ............................................................. 174 Figure 3-12. MS of CHKtide phosphorylation assay ....................................... 175 Figure 3-13. MS/MS of phosphorylated CHKtide ............................................ 176 Figure 3-14. H5/D5 calibration curve ............................................................... 177 Figure 3-15. MS of 05 enrichment using Resin 2-1 ....................................... 178 Figure 3-16. MS of D5 enrichment using Resin 2-2 ....................................... 179 Figure 3-17. MS of 05 enrichment using Resin 2-3 ....................................... 180 Figure 3-18. MS of 05 enrichment using Resin 2-4 ....................................... 181 Figure 3-19. MS of D5 enrichment using Resin 2-5 ....................................... 182 Figure 3-20. MS of 05 enrichment using Resin 2-6 ....................................... 183 Figure 3-21. MS of DS enrichment using Resin 2-7 ........................................ 184 Figure 3-22. MS Of D5 enrichment using Resin 2-8 ....................................... 185 Figure 3-23. MS Of 05 enrichment using Resin 2-9 ....................................... 186 Figure 3-24. MS of D5 enrichment using Resin 2-10 ..................................... 187 Figure 3-25. MS Of DS enrichment using Resin 2-11 ..................................... 188 Figure 3-26. MS of 05 enrichment using Resin 2-12 ..................................... 189 Figure 3-27. MS Of 05 enrichment using Resin 2-13 ..................................... 190 Figure 3—28. MS of D5 enrichment using Resin 2-14 ..................................... 191 Figure 3-29. MS of D5 enrichment using Resin 2-15 ..................................... 192 l=‘lgure 3-30. MS of DS enrichment using Resin 2-16 ..................................... 193 l=igure 3-31. MS of D5 enrichment using Resin 2-17 ..................................... 194 Figure 3-32. MS of D5 enrichment using Resin 2-18 ..................................... 195 xv Figure 3-33. MS of 05 enrichment using Resin 2-19 ..................................... 196 Figure 3-34. MS of D5 enrichment using Resin 2-20 ..................................... 197 Figure 3-35. MS of D5 enrichment using IMAC ZipTips .................................. 198 Figure 3-36. MS Of D5 enrichment using Glygen Ti02 NuTips ....................... 199 Figure 3-37. MS Of D5 enrichment using Glygen ZnOz NuTips ...................... 200 Figure 3-38. MSIMS of ion 2173 from ovalbumin enrichment ......................... 201 Figure 3-39. MSIMS of ion 2553 from ovalbumin enrichment ......................... 202 Figure 3-40. MSIMS of ion 2160 from B-casein enrichment ............................ 203 Figure 3-41. MSIMS Of ion 3234 from B-casein enrichment ............................ 204 Figure 4-1. The classical NF-KB signaling pathway ...................................... 213 Figure 4-2. Known NF-KB phosphorylation sites ........................................... 215 Figure 4-3. General approach for the purification of NF-KB .......................... 225 Figure 4-4. Western blot analysis of nuclear extracts from THP-1 cells probing for NF-kB p65 subunit + actin ............................................................... 229 Figure 4-5. Western blot analysis of p65 immunoprecipitated nuclear extracts from CEM and L363 cells .................................................................................... 232 Figure 4-6. Silver stain of p65 immunoprecipitated nuclear extracts from CEM and L363 cells .................................................................................................... 233 Figure 4-7. Western blot and silver stain comparison ................................... 234 Figure 4-8. MS of the p65 antibody tryptic digest .......................................... 266 Figure 4-9. MS of THP-1 untreated nuclear extract immunoprecipitate tryptic digest ................................................................................................................... 267 Figure 4-10. MS of THP-1 treated nuclear extract immunoprecipitate tryptic digest ................................................................................................................... 268 l=igure 4-11. MS of THP-1 treated immunoprecipitate digest enrichment ....... 269 p. .134 DA ‘Uv U mPuh‘ 4‘. in In... _‘ '7‘.an - b “I Ip’ :“v Figure 4-12. MSIMS Of ion 1738 from the THP—1 treated immunoprecipitate digest enrichment ................................................................................................ 270 Figure 4-13. MS3 of ion 1738 from the THP-1 treated immunoprecipitate digest enrichment ........................................................................................................... 271 Figure 4-14. MSIMS of ion 2302 from the THP-1 treated immunoprecipitate digest enrichment ................................................................................................ 272 Figure 4-15. MS3 of ion 2302 from the THP-1 treated immunoprecipitate digest enrichment .......................................................................................................... 273 Figure 4-16. MS of THP-1 treated immunoprecipitate digest hydrolyzed enrichment .......................................................................................................... 274 Figure 4-17. MSIMS Of ion 2231 from the THP-1 treated immunoprecipitate digest hydrolyzed enrichment ............................................................................. 275 Figure 4—18. MSIMS of ion 1653 from the THP-1 treated immunoprecipitate digest hydrolyzed enrichment ............................................................................. 276 Figure 4-19. MS of angiotensin II-diazo biotin coupling .................................. 277 xvii LIST OF SCHEMES Scheme 1-1. General process of reversible protein de/phosphorylation .............. 4 Scheme 1-2. General B-elimination-phosphate modification approach ............. 26 Scheme 1-3. Enrichment approach using modified phosphopeptide-biotin tag coupling ................................................................................................................. 28 Scheme 14. Improved modified phosphopeptide-solid support coupling .......... 29 Scheme 1-5. Methodology used in phosphospecific proteolysis ........................ 30 Scheme 1-6. Phosphoamidate modification enrichment scheme ...................... 33 Scheme 1-7. Triphenylphosphine mediated phosphoamidate formation on a solid support .......................................................................................................... 34 Scheme 1-8. Solid phase phosphopeptide enrichment using diazo- functionalized polystyrene ..................................................................................... 55 Scheme 2-1. Reactivity of diazo carbonyl moieties towards acids ..................... 79 Scheme 2—2. Theory Of diazo carbonyl-phosphate binding ................................ 81 Scheme 2-3. Scheme for the formation of the covalent phosphoester bond ..... 84 Scheme 2-4. HONO gas mediated diazotization of amino resins ...................... 85 Scheme 2-5. Diazotization reaction using glyoxylic acid p- toluenesulfonylhydrazone ..................................................................................... 88 Scheme 2-6. Synthesis of glyoxylic acid p-toluenesulfonylhydrazone ............... 89 Scheme 2-7. Synthesis Of a-diazo acetate Wang resin using DIC coupling ...... 90 Scheme 2-8. Optimized synthetic route for a-diazo acetate Wang .................... 92 Scheme 2-9. Application of Optimized diazo synthetic route for both hydroxyl and amino functionalized solid supports ............................................................... 93 Scheme 2-10. Synthesis of diazo ketone resins .................................................. 96 SCheme 2-11. Synthesis of phenyl diazo methane polystyrene ........................... 98 xviii Scheme 2-12. Quantification of diazo materials with Fmoc-glycine ................... 100 Scheme 3-1. General mechanism for covalent enrichment of phosphopeptides from diazo-functionalized solid support ............................................................... 140 Scheme 3-2. Formation Of the 32P labeled synthetic peptide CHKtide ............. 144 Scheme 4-1. Formation of diazo-functionalized biotin derivative ..................... 248 Scheme 4-2. Coupling of angiotensin II to diazo biotin .................................... 249 Scheme 4-3. Synthetic route to prepare diazo ketone biotin derivative ........... 251 Scheme 4—4. Mechanism for the enrichment of phosphopeptides ................... 252 Scheme 4-5. Comparison between diazo ketone and diazo dicarbonyl solid support ................................................................................................................ 253 xix II! '\ NUI' PTM pl IEF SCX SAX HPLC LC-MS 2DE CID MALDI-MS ESI-MS MSIMS ETD ECD IMAC IDA NTA ICAT SILAC PFZD HCC KEY TO SYMBOLS AND ABREVEATIONS Post-translational modification isoelectric point isoelectric focusing strong cation exchange strong anion exchange high-performance liquid chromatography liquid chromatography-liquid chromatography 2-dimensional gel electrophoresis collision induced dissociation matrix-assisted laser desorption ionization mass spectrometry electrospray ionization liquid chromatography tandem mass spectrometry electron transfer dissociation electron capture dissociation immobilized metal affinity chromatography iminodiacetic acid nitrilotriacetic acid isotopic-coded affinity tag stable isotOpe labeling by amino acids in cell culture 2 dimensional liquid phase fractionation hepatocellular carcinoma XX .‘l? w J UV ‘AAI ZJ‘e "II'J J'I'I EGF EGFR SIMAC NF-KB IKK TNF DCC EDCI DMF THF DBU DBF NMR TFA TMS FMOC epidermal growth factor epidermal growth factor receptor sequential immobilized metal affinity chromatography nuclear factor-k8 I-KB kinases tumor necrosis factor dicyclohexylcarbodiimide ethyldimethylaminopropyl carbodiimide N,N—dimethyl formamide tetrahydrofuran 1 ,8-Diazabicyclo[5.4.0]undec-7-ene dibenzofulvene nuclear magnetic resonance trifluoroacetic acid trimethylsilyl fluoren-9-ylmethoxycarbonyl CHAPTER 1 PROTEIN PHOSPHORYLATION: PROJECT BACKGROUND INFORMATION 1.1 Introduction to post-translational modifications The complexity of the human proteome (the proteins encoded by ~30,000 genes) is a staggeringly two to three orders of magnitude larger than its corresponding genome, resulting in more than 1,000,000 various forms of proteins.1 The substantial increase in complexity is the result of both RNA transcriptional regulation‘?’3 and post-translational modifications (PTMs).1 The ladder includes ~300 known covalent modifications that occur in proteins.4 These modifications are responsible for protein inlactivation, folding, signaling, stability, and localization.“5 The importance of these modifications can also be reflected by the 5% of the genome reserved for encoding the enzymes responsible for these PTMs.1 The various enzymes involved in the catalysis of PTMs can be divided into two sub-groups; the covalent addition of some chemical group (i.e. phosphate, acyl, alkyl groups) and the covalent cleavage of the peptide backbone. Additionally, many of these transformations require some sort of co-factor to aid in the covalent addition/cleavage; such as acetyl COA dependent acetylation, ATP dependent phosphorylation, S-adenosylmethionine (SAM)-dependent methylation, etc.1 Illustrated in Figure 1-1 are the top five most common types Of covalent addition; phosphorylation, acylation, alkylation, glycosylation and oxidation. Phosphorylation Alkylation OH NH2 HN/ OH ATP /A9P O=I'=—OH SAM SA“ I ; O ‘§N’£fl§’ H O ‘IN E’ ‘IN E’ ‘SN E’ H O H O H O Acetylation O Oxidation NH2 JL OH COAS\R jASH H” R é N 1* \ II/ E” \ N O 0 ii; é- \EN é’ . , O H 0 EN 2 H O Glycosylation OH UDP-GI UDP 0 OH \uco'f/ Hamm ‘EN I’ F OH 0 H O \ I ill/{,2 H 0 Figure 1-1. Top five most common PTMs. All of these modifications change the physical properties of the amino acid to which they covalently attach. Whether it be from the addition of a negatively charged phosphate or by changing the hydrogen bonding properties of the N- terrninus by acetylation of the amine to form the amide. These changes not only affect the modified amino acid, but the protein it belongs to as a whole.1 The structural changes that take place in the protein as a result of PTMs, help illustrate their involvement in signal transduction pathways and how the activity of the protein changes as a result of PTMs as well. However, the dynamic role that 2 PTMs play in cellular activity/response/grOMh is still largely in question. Although this area of research is Of great interest, it remains challenging due to the need to isolate the post-translationally modified protein of interest for analysis. Isolation and subsequent analysis of the modified protein from an exceptionally complex proteome is why characterization of these modifications and the amino acid residues in which they bind is a challenge that has only been met in a few selective modifications. 1.2 Introduction to the study of protein phosphorylation In the previous section, it was already addressed that protein phosphorylation was one of the most ubiquitous PTMs known. The dramatic change in the physical properties of the amino acid as the result of becoming phosphorylated is quite substantial. As one might expect, the significant physical change in both the amino acid as well as the protein itself, proves advantageous in cellular communication. Many cellular processes including internal cellular signaling, differentiation, cell survival, metabolism, regulation and proliferation are regulated by the reversible process of protein phosphorylation, which is governed by protein kinases and phosphatases.6‘8 This ATP dependent transformation occurs by a kinase prompting the addition of phosphate to a protein and a phosphatase enzyme catalyzing its removal (Scheme 1-1). Kinase Ox‘P/OH ’ \ Cf” (I) OH Protein Protein Phosphatase ATP ADP Scheme 1-1. General process of reversible protein de/phosphorylation. While acyl phosphorylation on glutamic/aspartic acid residues and N- phosphorylation on histidine and lysine residues can occur in eukaryotes, O- phosphorylation on serine, threonine and tyrosine residues are the predominant forms of this modification.9 The human genome encodes for about 500 protein kinases, about 100 protein phosphatases, and it is estimated that one third of all 10,11 human proteins contain at least one site of phosphorylation. Given the de/phosphorylation events’ involvement in signal transduction pathways and the large percentage of proteins that can be phosphorylated, it is understandable that disruption of this process has been implicated in diseases such as cancer?” ”'15 cystic fibrosis,16 and Alzheimer’s disease?17 Due to the type II diabetes, impact phosphorylation has on cell function, phosphoproteins and kinases have been explored for the last two decades as either possible drug targets or biomarkers to treat or diagnose a variety of diseases (reviewed elsewhere in 18' 21). girls": 933351 his er 235.2 ificspiia mien . lama. If? D“: .I,RP ‘4 ”‘93:": As a result of the importance in studying protein phosphorylation, conventional biological techniques, such as gel electrophoresis and 32F radiolabeling of phosphopeptides/proteins have been utilized as the traditional tools employed in studying/detecting the phosphoproteome. While these classical methods have well documented protocols available to observe phosphoproteins (reviewed elsewhere in 2233 ), they do require a large amount of protein starting material. The implementation of mass spectrometry (MS) technologies to probe the phosphoproteome has added new depth in elucidating the phosphoproteome. Not only has mass spectrometry been used in phosphorylation site identification,2“'25 but when coupled with additional phosphoprotein enrichment methods, MS has become the phosphoproteomic analytical tool of choice due to both its speed and sensitivity.26 The cooperative use of phosphoprotein enrichment and mass spectrometry analysis has been employed in discovering new phosphorylation sites, which could lead to the discovery Of cancer biomarkers and drug targets.20 Regardless of the development of classical biological assays or mass spectrometry, proteomic techniques relevant to the elucidation of signal transduction pathways have still lagged in development when compared to genomic technologies. The challenge in phosphoproteomic identification is in part due to the complexity of cellular samples and the low abundance typical of phosphoproteins relative to their unphosphorylated counterparts. This is why the development of efficient enrichment technologies is a vital step to probing the phosphoproteome on a cellular level. Enrichment of specific post-translational x ...I ”0‘1 =5 - .. u\4 r221?) c ISQIrW‘n Ur. b ‘ U Supt”? ' k:’:vLII EtEP V PL?! ungma’r u. modified proteins can reduce the complexity of a sample and allow one to more readily determine qualitative discrepancies between samples. To quantify the changes in the phosphoproteome between samples, additional methodologies have also been developed (recently reviewed in 27'28). Both qualitative and quantitative data of phosphoproteins is pertinent to identifying potential biomarkers in particular biological pathways or diseases. In an effort to understand how phosphorylation-mediated signaling pathways occur, many different phosphoprotein/peptide separation techniques have been developed in the last 50 years that utilize physical, structural, or chemical properties pertaining to the phosphate moiety. The physical effects resulting from the addition of a phosphate include the potential change in the isoelectric point (pl) Of the protein, which can be separated from other proteins by either isoelectric focusing (IEF)29 or strong anion/cation exchange chromatography (SAX and SCX).30 Additionally, the negatively charged phosphate has affinity for metal cations, which has been exploited in several types of chromatography techniques including iron31 or gallium32 and metal oxides such as titanium dioxide33 and zirconium dioxide.34 The 3-dimensional structural changes that arise as a result Of phosphorylation allow for the development of phospho—specific antibodies, which can be used to isolate a specific phosphorylated form of a protein.35 The specific reactivity of the phosphate group can also be manipulated to further modify the protein/peptide, which can subsequently undergo enrichment of the 23,36) modified group (techniques reviewed elsewhere in ‘I"PP“ ~ . eel F‘F F :A‘ ':)y.:t,|, .. I (I . .3 (:3 A basic overview illustrating all of the current phosphoprotein detection and enrichment techniques discussed within this chapter are shown in the diagram below (Figure 1-2). New tools and modifications on the listed techniques are constantly emerging in an effort to develop more sensitive, selective, and quantitative approaches. Currently, there is no one particular approach that appears to be the ‘holy grail’ in phosphoproteomic analysis. With respect to global phosphorylation analysis, the application of multiple enrichment techniques, either in a series or parallel fashion, is generally required to Obtain the most information possible (reviewed elsewhere in 37). Phosphoprotein] cell Iysate sample Phosphoprotein detection . 32.; or 33p labeling Phosphoprotein . 41 enrichment Gel electrophoreSIs l I Edman degradation 42-45 Immunoprecipitation pSer/pThr- antibodies Affinity ”’93 chromatography Strong ion exchange Phospho specific stain 48-50 Phosphate modification phospho—amidate 129,131 .132 pTyr-antibodies 61 ,85-87 phosphate elimination 1 16,1 19 \g§ Fe3+ 31.66.91 .92.96.97 phosphospecific proteolysis 126,127 nucleophilic linker affinity label 117,118,121 reactive solid support 1 20,1 22—124 TIOZ 105,105,109-111 Figure 1-2. Phosphoprotein enrichment and detection technologies flow chart. 8 1.3 Detection of phosphorylated proteins Prior to routine mass spectrometry analyses, biologists and chemists alike had to utilize other means of phosphoprotein detection or analysis. The typical tools employed were either 32F labeling, gel electrophoresis, Edman degradation, the use of phosphospecific antibodies, or any combination of the aforementioned 38'39). These methods are still valid tools techniques (reviewed elsewhere in today and are used routinely in studying phosphoproteomics, but have since been coupled to mass spectrometry (MS) analysis because of its sensitivity, versatility and speed (reviewed elsewhere in 28'38'40). While the limitations of the aforementioned biological tools have been addressed by the employment of MS, these biological tools are still important when dealing with complex mixtures, large datasets, and low abundances of protein; all typical scenarios when dealing with phosphoproteomic analysis. As such, it is important to briefly discuss the validity of these biological approaches to phosphoproteomic analysis. 1.3.a 32F Radiolabeling The incorporation of radiolabeled phosphate into a phosphoprotein in vitro via 32P or 33P—ATP or the in vivo addition of radiolabeled organic phosphate (32F) and subsequent identification of the radiolabeled protein is considered one of the classical methods in the detection of phosphorylated proteins. This assay was first used to confirm that both glucagon and epinephrine stimulate the phosphorylation of dog liver phosphorylase.41 05...: O .‘ 1‘ r Since then, this technique has been implemented for many different in vitro and in vivo phosphorylation studies. Typically, the site of phosphorylation has been characterized by first purifying and digesting the radiolabeled phosphoprotein, then isolating the radiolabeled peptides by high-performance liquid chromatography (HPLC) followed by sequence analysis using Edman c:legradation.“‘°"14 While this approach has successfully been performed on cell culture to identify phosphorylation sites,45 there are limitations to this approach in vivo as 32F labeling is inefficient due to high amounts of endogenous ATP present within cells and some phosphorylated proteins have low phosphate turnover rates, both of which will result in little incorporation of the radiolabel. Additionally, the i r'ItrOduction of radioactivity can cause a cellular stress response resulting in artificial phosphorylation events.38 Edman degradation can also become problematic by causing phosphate elimination on phosphoserine residues during analysis and is not capable of sequencing peptides with blocked N-termini. The employment of radiolabeling on its own is somewhat limited, which is even more true for complex samples and requires coupling of another technique besides Edman degradation, such as gel electrophoresis, for further phosphoproteomic a halysis.46'47 1 - 3.6 Gel electrophoresis/Phosphoprotein staining In order to separate and detect phosphoproteins within a complex mixture, SL1 ch as a cellular extract, 1 and 2 dimensional gel electrophoresis or HPLC is 10 A 3 55.8 arcs? JETU “Pa 3:9 “I“ 3. A... \. 'N ;i..3 em. .51 typically the method of choice. In 2-D gel electrophoresis, one dimension is usually a separation by charge state (ie. the pl), which is referred to as isoelectric focusing (IEF). The other dimension is separated by molecular weight of the protein using a polyacrylamide gel. In separating proteins by 1D or 20 gel electrophoresis, individual phosphoproteins can be selectively visualized using phosphoprotein specific stains.48 Early in the development of these stains some issues arose due to a lack of specificity of the stains, which limited the utility of this technique. However, in recent years a fluorescent-based dye called Pro-Q Diamond was found to selectively bind directly to the phosphate moiety of 3:)roteins.“9'50 Pro-Q Diamond stain shows great promise in phosphoproteomic analysis because of its specificity, selectivity and its compatibility with mass spectrometry analysis. It also is capable of staining either all types of O- phosphorylated residues, or specifically phosphotyrosine, depending on the staining protocol. This approach has been used for phosphoprotein analysis on a variety Of different biological systems/cellular samples.39 The utility of gel electrophoresis and its subsequent staining can also be fI-J rther characterized by MS analysis. The identification of phosphopeptides by application Of both techniques has been performed by first separating the protein Via gel electrophoresis followed by in-gel digestion and MS analysis. The digest was then subjected to phosphate removal via alkaline phosphatase and d i1=l"erential analysis is performed, monitoring the loss of 80 within a peptide thus (:0 hfirming the peptide was phosphorylated.51 Another example of the application 11 In) C) RAAR| | Eta: ‘n 5;»; UV'b-II I I‘Ai Ll‘» h it. n+, ‘ a “b.“ of these two technologies was illustrated by Patton and co-workers who selectively stained and characterized phosphoproteins from a Jurkat cell Iysate.49 1.3.c Phosphospecific antibodies In addition to phosphospecific staining on a polyacrylamide gel, another means to visualize femtomole amounts of a phosphoprotein of interest is western blotting.52 This technique, however, is limited by the availability and selectivity Of an antibody corresponding to the protein of interest.” Ideally, the antibody should recognize either the specific phosphorylation site or a type of phosphoamino acid (i.e. phosphotyrosine), in order to avoid selectivity issues associated with the detection of unphosphorylated forms Of the same protein. There are many phospho-serine, threonine, and tyrosine antibodies that have been developed and are available with limited cross-reactivity to the unphosphorylated forms of these residues.“"“‘56 The use of phosphotyrosine antibodies to study the phosphorylation state of various neural proteins in vitro and in vivo has proven the technique to be successful.57‘59 Additionally, this approach can be used to monitor the amounts Of phosphoprotein levels in a plate-based assay forrnat.60 Foley and co-workers grew 82 cells in a 96-well plate, stimulated the cells, subsequently fixed the cells onto the plate and probed the plate directly with an anti-phospho JNK antibody. The primary antibody was then probed with a fiucrophore linked secondary antibody for visualization. This technique allowed for the quantification of JNK activation in cells directly.60 12 Chin and co-workers took a different approach employing immunoprecipitation by first immobilizing 50-100 different antibodies on a PVDF membrane and then subsequently introduced the membrane to mammalian cell extracts. The cellular proteins were labeled with a fluorescent dye to visualize the presence of the protein of interest.61 The application of probing a ZD gel for the different types of phosphoamino acids using different monoclonal antibodies followed by subsequent protein mass fingerprinting has been utilized in phosphoprotein characterization.47 Instead of employing the antibody to visualize the protein of interest, an alternative application is the use of antibodies for selective immunoprecipitation of a desired phosphoprotein, which will be discussed further in the enrichment section. 1.4 Enrichment of phosphorylated proteins Despite recent advancements in mass spectrometry techniques, including tandem mass spectrometry (MS/MS), the characterization of phosphopeptides still remains challenging. Identification of phosphopeptides is Often hampered by their sub-stoichiometric abundance relative to their unphosphorylated counterparts, resulting in signal suppression.62 Additional ion suppression can also result from the formation of phosphopeptide-metal ion complexes.63 Enrichment of phosphorylated peptides/proteins prior to their introduction into a mass spectrometer can therefore help facilitate phosphoprotein analysis 23,27,28,36,64,65 (reviewed elsewhere in ). Additional problems relating to the lability 13 “an!!!" iv ' . ~‘~fl I h u Hal" 1 I prispr 363.91 1C‘IE‘II ET IV'I CeZE’T'ii WET; “II-”hf. an I V '|. «A. . a of the phosphate group when introduced into the mass spectrometer, includes dominant neutral losses of 98 and 80 Da in both MS and MSIMS analysis, creating difficulty in both sequencing the peptide and assigning the site of 7 phosphorylation.66'6 Consequently, an alternative approach uses MS3 to sequence the phosphopeptides when dominant H3PO4 loss in MS2 occurs.2'°"68'69 However, recent evidence supports the loss of H3PO4 results in a cyclic product ion whose subsequent MS3 product ions could provide additional insight to determine the site of phosphorylation within a peptide containing multiple potential phosphorylation sites.70 Additional difficulty in MS analysis is the potential for the phosphate to transfer to an unmodified hydroxyl containing amino acid when analyzed by collision induced dissociation (CID)-MS/MS, potentially resulting in mis-identification of the site of phosphorylation.71 The problems associated with CID-MSIMS analysis in both the facile H3PO4 loss and the gas-phase phosphate rearrangement of phosphoproteins has been addressed via the use of either ETD (electron transfer dissociation)72'73 or ECD (electron capture dissociation)74 to induce molecular ion fragmentation. The fragmentation occurs for both ETD and ECD primarily at the N-Ca bonds, creating c and 2 type ions.75'76 This type of fragmentation allows the phosphate to remain intact along the peptide backbone, enabling phosphorylation site identification.77""'2 Identification of the sequence of a peptide and its” respective protein, can be assisted by comparing the spectra with an MS database, however, the database only aids in identification if the protein in question is in the database. 14 Additional software utilized to process the excessive amounts Of phosphoproteomic data acquired from large sample sets, Gygi and Schwartz developed an algorithm to aid in the identification and classification Of sequence motifs of phosphorylation sites.83 They applied this algorithm to characterize phosphorylation sites found from HeLa nuclear protein analysis,84 and found 77% Of phosphopeptides that were characterized had a proline residue 1+ position (C- terrninal) and/or a glutamate at the 3+ position. This bioinforrnatics tool can both be used to analyze large phosphoprotein datasets and as a comparative tool in the sequence homology between different types of phosphoproteins collected from global phosphorylation studies described below. The sub-stoichiometric problem has also been addressed by many groups using a variety of different techniques, which will be discussed in more detail herein. The most widely employed enrichment method has utilized the phosphate moiety’s affinity for metal cations and metal oxides. Alternative approaches include elimination of the phosphate followed by subsequent introduction of a more stable modification. Thus, identification assignment of the phosphorylation site is based on identifying the modified residue (enrichment procedures reviewed elsewhere in 27""8"’"3'38'40'64). 1.4.a Immunoprecipitation As illustrated in the previous section, antibodies have been used as a means of detecting phosphorylated proteins, however, they also can serve as a phosphoprotein/peptide enrichment tool. Instead of employing the antibody to 15 visualize the protein of interest, an alternative application is the use of antibodies for selective immunoprecipitation Of a desired phosphoprotein. The antibody of choice is bound by either affinity (i.e. biotin) or covalently (i.e. agarose) to a solid support and introduced to a cellular fraction containing the protein Of interest. The protein is subsequently purified from the rest of the cellular proteins by collecting the solid support-antibody—protein pellet. The protein is then eluted from the pellet and enzymatically digested and subsequent MS analysis of the immunoprecipitated sample can lead to identification of the phosphorylation sitesaf”86 The application of probing a 2D gel for the different types of phosphoamino acids using different monoclonal antibodies followed by subsequent protein mass fingerprinting has been utilized in phosphoprotein characterization.47 Alternatively, the cellular extract may first be digested, then introduced to an antiphosphotyrosine antibody, allowing for the global MS identification of phosphotyrosine peptides.87 1.4.b Affinity chromatography As mentioned earlier, the addition of the negatively charged phosphate on the protein can be manipulated to be purified by chromatography. Specifically, affinity chromatography has been employed utilizing the affinity of the negatively charged phosphate towards metal cations. This purification approach has been exploited in several types of chromatography techniques including iron31 or gallium32 and metal oxides such as titanium dioxide33 and zirconium dioxide.34 16 Additionally, the physical effects resulting from the addition of a phosphate include the potential change in the isoelectric point (pl) of the protein, enabling purification by either strong anion/cation exchange chromatography (SAX and sexy” 1.4.b.i. Immobilized metal ion affinity chromatography (IMAC) Identifying phosphorylation sites on proteins by isolating phosphopeptides through exploitation of their affinity for metal cations has been explored for many years. Originally coined “metal chelating affinity chromatography’, this technique was introduced in 1975 by Porath and co-workers and used to purify histidine- tagged proteins.88 When subsequent studies revealed that phosphorylated amino acids also bind to iron(III)-charged species coordinated to agarose-bound iminodiacetic acid, immobilized metal ion affinity chromatography (IMAC) was bom.31 IMAC has been one of the most widely employed and well-known techniques used in phosphoproteomic studies. This method utilizes metal ions, such as Ga3+ or Fe‘”, which have been found to have the highest percent recovery of the negatively charged phosphate.32 The metal ions are first introduced to chelating agents, such as iminodiacetic acid (IDA) or nitrilotriacetic acid (NTA), which are immobilized on a resin, bead, glass chip or membrane. The IDA-metal complex and the NTA complex has three sites and two sites of available coordination, respectively (Figure 1-3). The coordination sites are only available to the phosphate at a pH range between 2.0-3.5.32 In an effort to 17 Figure develop an IMAC material capable of enrichment at physiological pH, an alkoxide-bridged di-nuclear Zn (II) was developed, referred to as phos—tag (Fig. 1-3).89 0 O > O NH 9/15 \ / \ O \ 1’ I \ /N\ ’N / ‘ 2+ ’2+ I \/ . / N\/K/N \ (a) 0 (C) (3"gk 0) + E ,0 O -n" (D ‘\ N2 C) Figure 1-3. Coordination between metal cations and IMAC chelating reagents. The phosphopeptide enrichment can be carried out by introducing a complex biological sample, such as a protein digest or cellular extract, to the column or surface containing the immobilized metal cations. The phosphate moiety coordinates to the metal cations allowing for the removal of unbound peptides. Liberation of the bound phosphopeptides from the affinity material is accomplished by either washing the column with a basic pH solution, or by introducing excess free ligand, such as a phosphate buffer. The liberated peptides are typically analyzed directly by either matrix assisted laser desorption/ionization mass spectrometry (MALDI-MS) or electrospray ionization mass spectrometry (ESI-MS) to sequence the peptide and determine the site of phosphorylation. There are some problems associated with using IMAC to determine phosphopeptides/proteins. These challenges include the recovery of multiply phosphorylated peptides which have a higher binding affinity to the metal cations 18 3'13 'ES'JI ibe'ate finna Hi e'I’eI H 'J\ v :cior res, :5 SSLE EST-3'3 us? 55265531. Jfl‘wflp‘n q Gi .6. “motor. and result in poor elution from the support, requiring harsher conditions to liberate the peptides.66'90 The most problematic issue using IMAC is non-specific binding due to acidic residues (i.e. aspartic acid and glutamic acid) and electron donor residues (i.e histidine), which also have affinity to the metal ions. In part, this issue has been addressed by esterifying the carboxylic acids to methyl esters using in situ formed HCI in dry methanol.66 It is important to follow the esterification conditions explicitly to avoid either incomplete conversion or unwanted side reactions, both of which increase the complexity Of the sample. Phosphorylation site identification may also become more challenging as a result of other amino acids such as tyrosine, histidine, and tryptophan binding to the IMAC material. There have been many advancements to improve current IMAC technologies, including fabricating higher loading solid supports such as mesoporous nanoparticles,91 glass microchips,92 magnetic particles,93'94 metal- chelating plastic MALDI chip95 and functionalized gold MALDI plates.96'97 Higher loading has increased the sensitivity of the supports and decreased the amount of IMAC material required to carry out the enrichment. In an effort to develop a high-throughput and automated approach to IMAC, HPLC columns have also been fabricated with metal chelating silica to allow for large-scale analysis or front-end LC-MS analysis."""98'99 The scope of IMAC technology has been explored by a variety of research groups, measuring the capacity, selectivity, and enrichment ability of different materials. The application of IMAC has also been used to probe the 19 . AAHF‘A x2. ‘ .rv ? n. r ' Is III'TI‘ :. gr II,“ .i, .H phosphorylation sites of a variety of other commercially available I66 as well as phosphoproteins and characterizing the phosphoproteome of yeas various cancer cell lines (discussed later within this review), indicating the capability Of this technique to handle large datasets. In addition to the aforementioned limitations of IMAC, an additional drawback is the inability to fractionate IMAC enriched samples as an additional purification measure, as IMAC peptides are typically eluted in only one fraction. This limitation has been addressed by the development of Sequential elution from IMAC (SIMAC).1°° In this technique, mesenchymal stem cells were harvested, the proteins digested and the peptide mixture was introduced to iron-coated PHOS-select metal chelate beads. The beads were washed with 1% TFA in 20% acetonitrile to elude the singly phosphorylated peptides and contaminating unphosphorylated peptides, then multiply phosphorylated peptides were eluted from the IMAC beads upon introduction of ammonium hydroxide (pH 11.3). All of the IMAC enriched fractions including the flow-through and washing solutions were then further enriched using Ti02 chromatography and then analyzed by either LC-MS or MALDI-MS,100 resulting in the number of monophosphorylated peptides identified as 179, 232 and 80, respectively. Taking into consideration phosphopeptide overlaps, a total of 306 unique monophosphorylated peptides were detected, 55 Of which were identified from the IMAC flow-through, indicating the importance of a secondary enrichment technique to capture phosphopeptides that do not bind well to the IMAC column. The resulting multiply phosphorylated 20 peptces EigIeOI' VII of scar-g “inf-0' “V'V . . be has Th 2% pics were ml: peptides that were detected in the flow-through, 1% TFA elution and NH4OH elution were 8, 11, and 179 respectively.100 The results illustrate the importance of adding a basic solution to the IMAC column in order to obtain the multiply phosphorylated peptides, as more than 96% of the those peptides were identified in the basic elution wash and not in the 1% TFA elution. Further application of IMAC for global phosphoproteome analysis will be discussed later within the text. The SIMAC approach was compared to using just Ti02 enrichment and 286 phosphopeptides were identified using TIO2, where 54 Of those peptides were multiply phosphorylated. The use of the SIMAC technique resulted in the identification of 492 phosphopeptides where 186 of those were multiply phosphorylated, illustrating the improvement of enrichment for multiply phosphorylated peptides when using SIMAC.100 1.4.b.ii Strong ion exchange chromatography (SCX, SAX) Another technology that employs the negative character of the phosphate as means of separation is ion exchange chromatography. Enrichment of phosphopeptides by the use of strong cation exchange (SCX) is achieved by the negatively charged phosphate’s inability to bind well to the SCX material, whereas unphosphorylated peptides are more readily retained on the support. Successful application of this approach has been illustrated on cellular protein extracts that had been separated by gel electrophoresis and then in-gel digested.“101 Cooperatively, SCX can also be used as a front-end purification 21 technique prior to TIOz mediated phosphopeptide enrichment as shown by Mann and co-workers.69 Additionally, strong anion exchange (SAX) can also be used to enrich phosphorylated peptides.102 As opposed to SCX, in SAX the negatively charged nature of the phosphate moiety allows it to bind SAX material while their unphosphorylated counterparts are not retained. Comparatively, both SAX and SCX have been used to enrich phosphopeptides and it was found that each ion- 3 Dai et al. exchange method enriched different types of phosphopeptides.10 reported some loss of phosphopeptides from the use of SCX, which was confirmed by the detection Of phosphopeptides collected from the SCX flow through fractions.104 Gu and co-workers probed the liver phosphoproteome with SAX and compared the results obtained with SAX versus using the IMAC approach. The technique was compared qualitatively using standard protein digests to IMAC and found to have identified >95% of the singly phosphorylated peptides, where IMAC identified 66.7%.102 The benefit of SAX over IMAC is the ability for SAX to fractionate samples, whereas IMAC typically produces only one fraction, unless SIMAC is performed. 1.4.b.iii Metal oxide affinity chromatography (MOAC) In an effort to find a material with a higher degree of specificity for the phosphate group, metal oxides can be explored. In this approach the phosphate is proposed to bind to the titanium dioxide through a bidentate interaction (Figure 1-4).33 Heck and co-workers described a procedure to trap phosphopeptides in 22 a pre-column packed with Ti02 (titansphere) under acidic conditions. Subsequently, the phosphopeptides can be desorbed from the column under basic conditions.105 These fractions are trapped on a reversed-phase column for further separation and subsequently analyzed by LC-ESl-MS/MS. a) O b) HO \\ ,OR(H) /P\ O O O l l O. /0 T102 TI02 T102 Figure 1-4. Hypothesized coordination on Ti02 support comparison between carboxylic acids and phosphoric acid by Larsen et al.106 While this technique is capable of enriching low femtomole quantities of phosphopeptides, some acidic, non-phosphorylated peptides also show affinity to the titanium dioxide column. This selectivity issue was addressed by Larsen et al., who prepared microcolumns packed with Ti02 and observed a higher degree of phosphopeptide specificity when 2,5-dihydroxybenzoic acid (DHB) was present in the loading solution.106 The increase in selectivity of Ti02 for phosphopeptides under these conditions was thought to result from competition between DHB and non-phosphorylated peptides for the remaining Ti02 binding sites. This hypothesis was confirmed with spectroscopic studies indicating that substituted aromatic carboxylic acids, such as DHB, coordinate strongly to the surface of TiOz, while monofunctional carboxylic acids (i.e. benzoic acid or glutamic acid) coordinate much more weakly with titanium dioxide 107. Larsen 23 also suggested that while there is competition between DHB and other non- phosphorylated peptides, it is not competing with phosphorylated peptides. This is due to the difference between DHB and phosphate in their coordination geometry with TiOz. DHB is a bidentate chelator,107 whereas the phosphate group is a bridging bidentate,108 which results in both species differing in Optimal binding sites (Figure 1-4). Improvement in phosphopeptide selectivity with Ti02 versus IMAC enrichment was confirmed by Krause and co-workers, who evaluated several phosphopeptides and their unphosphorylated counterparts for Ti02 enrichment.109 Recently, reproducible protocols have been published using Ti02 packed pipette tips in phosphopeptide enrichment and analysis.“"'111 Additionally, zirconium dioxide packed pipette tips have also been implemented in the enrichment of phosphopeptides. Comparatively, the two types of commercially available metal oxide tips were found to be equivalent in phosphopeptide enrichment, however, the ZI'Oz tips were found to be more selective for singly phosphorylated peptides while the Ti02 tips were superior for the enrichment of multiply phosphorylated peptides.34 Both of these materials have been useful in global phosphoproteome characterization, which will be explored later on within the text. Ti02 is the most widely employed metal oxide in phosphoprotein enrichment; however, its development brought about the exploration Of additional metal oxides for their enrichment capabilities. Zhang and co-workers have coated Fe304 magnetic microspheres with either Al203112 or Ga203113 and both 24 materials were capable of enriching phosphopeptides from commercially available phosphoproteins and rat liver protein digests. Additionally, Sn02114 and Nb205115 have also recently been identified as phosphopeptide enrichment materials. 1.4.c B-Eliminatlon-phosphate modification Another phosphopeptide enrichment technique that became established around the same time as phosphO-affinity enrichment, was phosphopeptide derivatization. Manipulation of the reactivity of the phosphate moiety can be implemented to purify phosphorylated peptides/proteins from their unphosphorylated counterparts through the addition of base. Under basic conditions the phosphate moiety can be eliminated upon the deprotonation of a carbonyl and the resulting eliminated product can be further modified and analyzed. The addition of the nucleophile can then be corresponded back to the original site of phosphorylation. 1.4.c.i Chemical tagging In the chemical tagging approach, the phosphopeptide is first modified by elimination of the phosphate group on either phosphoserine or phosphothreonine, followed by nucleophilic attack of the newly formed Michael acceptor (Scheme 1 -2). 25 O=Fl’-OH l h'l Nuc O 'OH ~ \ . nuc eop re; T g ”H; V \ExNjWE’ ' §' O H phosphoserine dehydroalanine Scheme 1-2. General scheme for B-elimination-phosphate modification approach for phosphoserine. The formation of the unphosphorylated derivative eliminates the problem of difficult sequence analysis from dominant -H3PO4 loss in the MSIMS spectrum. Prior to routine mass spectrometric analysis, the phosphorylation site on the peptide derivatives were characterized by Edman degradation.116 Meyer and coworkers found the nucleophile of choice to be ethanethiol, due to its addition efficiency and its significant change in elution time allowing for definitive identification.116 More recently, this route has been used to modify a variety of phosphopeptides and subsequently analyze the modified product by ESI or MALDI MS."7'119 The modified proteins undergo proteolytic digestion and their phosphate site is characterized by the +44 mass unit shift of the modified peptide observed in MSIMS analysis.119 Validation of the approach was confirmed by the commercially available protein and additionally, 28 phosphorylation sites were characterized on the purified human protein.119 While this technique does prove advantageous over the affinity approaches as it removes the challenges of phosphopeptide sequence analysis by modifying the phosphate first, there are limitations. The most notable drawback for the B-elimination approach is its incompatibility to detect 26 phosphotyrosine residues, as they do not undergo B-elimination. Another limitation is the elimination of unmodified serinelthreonine residues and O- glycosylated peptides which can result in a “false positive’ modification and create ambiguous phosphorylation site determination. Aside from the direct MS analysis of the modified peptide without additional purification, there are other applications for the phosphopeptide derivatives that involve phosphopeptide enrichment. The B-elimination product can be introduced to a nucleophile that is also attached to either an affinity tag or immobilizing reagent, allowing for selective enrichment of the modified phosphopeptides. Application of this concept has been employed to enrich phosphopeptides using the nucleophile ethanedithiol to react with the B- 1" Prior to the introduction of the elimination species (dehydroalanine). ethanedithiol, cysteine residues are oxidized to prevent any unwanted nucleophilic attack by adjacent residues. The terminal sulfur on the modified (previously phosphorylated) peptide can undergo subsequent nucleophilic attack to a biotin linked maleimide, forming an affinity labeled (formerly phosphorylated) peptide. The electrophilic maleimide-biotin incorporated within the modified peptide can then be purified by avidin beads, which retain the biotinylated species on the column (Scheme 1-3). The flow-through and washing steps effectively remove any unmodified (i.e. unphosphorylated) peptides.117 27 O=FI’-OH /\/SH OH HS 0 ifl H3PO4 3‘Njfi"? H O Biotin ii 017” B' . O IOtInw-NH 002” H BiotinW-N KI H20 0 H <—-— S 3 ml“? it lift H N O H 0 Scheme 1-3. Enrichment approach using modified phosphopeptide coupled to a biotin affinity tag. While biotin affinity chromatography is an attractive approach because of its availability and well documented protocols, there are a few limitations using biotin as an affinity tag. The biotin-avidin interaction is quite strong, making Blution of the biotin-peptide species sometimes difficult. As a result, other researchers have utilized this established enrichment strategy but developed a Solution to the current issues associated with biotin affinity tags. The incorporation of both the ethanedithiol and the maleimide was performed, however, the maleimide was bound to an acid cleavable solid support instead of biotin, thereby eliminating the problems associated with biotin.120 Additional work maintained the use of biotin as an affinity label, but was first coupled to an acid 28 g cleavable linker prior to its introduction to the modified peptide and then subsequently liberated the modified peptide from the affinity column under acidic conditions.121 The implementation of either the ethanedithiol phosphopeptide derivative122 or a solid-support bound sulfur123 as the nucleophile has also been thoroughly explored towards Optimization of this technique. Further problems arise in the sequence analysis of the biotin-containing peptides which may be challenging due to fragmentation patterns that are hard to interpret. This limitation has been addressed in a follow-up publication by Chait and co—workers, who again incorporated ethanedithiol, but then performed the enrichment using an activated thiol affinity resin followed by liberation Of the bound peptide by reduction of the disulfide bond (Scheme 1-4).124 The absence 0f the biotin tag increased the sensitivity of the enrichment, however, the authors acknowledged still having difficultly addressing the unwanted side reaction, B- elrrn i nation of serine. H S S N e, ”O H O Scheme 1-4. Improved modified phosphopeptide coupled directly to a solid $342734)? Q’Sslgr H3P04 support. 29 Cramer and co-workers explored the complimentary use of both IMAC as an initial enrichment step followed by a subsequent B—elimination of the phosphate on the IMAC solid support, which eluted to modified phosphopeptides 125. The benefit of coupling the two techniques simultaneously avoids aforementioned complications inherent of each method, such as improving the elution Of the phosphorylated peptides by eliminating the phosphate on the IMAC support. Additionally, the use of IMAC prior to the B-elimination allows glycosylated and phosphorylated peptides to be distinguished. 1.4-c.ii Proteolytic phosphopeptide mapping To date there is currently no known protease to selectively hydrolyze residues at their phosphorylation site, however there has been a way to accomplish this through phosphopeptide modification. This approach still implements the documented B-elimination-derivatization sequence, however, the peptide is modified to form a lysine residue mimic, which upon introduction to trIv’Psin, will selectively cleave at the formerly phosphorylated amino acids?”127 (Scheme 1-5) OH H2N NH2 NH2 NH2 e a. a. %. -OH SH + \: = $5,311 TwJKgI-Ficujwy a.” = . — snlfl O O O O H3PO4 Scheme 1-5. Methodology used in phosphospecific proteolysis. 30 Independently, Shokat and co-workers and Hathaway and co-workers each developed similar chemistry by derivatizing phosphoserine to form aminoethyl cysteinem"127 Structurally, aminoethyl cysteine behaves similar to the natural amino acid lysine, and by introducing the modified peptide to a protease that cleaves at lysine residues, such as trypsin, it will also selectively cleave at the modified phosphorylated residue (i.e. aminoethyl cysteine). Subsequent MSIMS analysis of the digested modified protein can identify the presence of pseudo-lysine residues, therefore identifying the phosphorylation sites present within the protein. The same limitations apply for this method as stated earlier, including the exclusion of phosphotyrosine identification and unwanted side reactions. Additionally, characterizing phosphothreonine residues is challenging, as they do not undergo phospho-ester cleavage readily.126 In a follow-up publication, Hathaway and co-workers developed a modification of the original procedure to reduce the problem with unwanted side reactions by discriminating between Phosphorylated and glycosylated peptides.128 Hathaway observed complete conversion to the B-elimination product for both phosphorylated and glycosylated Peptides in the presence of barium hydroxide. However, when ammonium hydroxide was substituted for the barium hydroxide, 60% of the glycopeptides remained unconverted. While this reduces the amount of false characterization Of a glycopeptide as a phosphopeptide, caution still must be issued when making PhOSphorylation site assignments based solely on this methodology. as the PhOSphate is no longer intact to confirm its identity. Later studies determined the 31 Optimized conditions to enable the selective elimination of phosphopeptides over glycopeptides by reducing the amount of barium hydroxide used and decreasing the amount of reaction time under these conditions.122 1.4.d Covalent phosphate addition to a solid support Other than techniques relying on B-elimination to derivatize the phosphate moiety, the phosphate moiety is not inert from other types of nucleophilic addition. The phosphate itself has access to an arsenal of additional reactivity utilizing both its nucleophilic and its electrophilic properties. For example, adaptation of a known condensation between carboxylic acids and carbodiimides can be applied to the phosphate moiety for phosphopeptide enrichment.129 Prior to the condensation, this method requires protection Of all the carboxylic acid residues and C-terminus with ethanolamine to eliminate any unwanted side reactions, as their reactivity is similar to that of phosphate. Aebersold and co- workers performed the condensation reaction catalyzed by the carbodiimide between cysteamine and the phosphate, and upon nucleophilic addition of ditI'IiOIthreitOI, the disulfide bond was reduced, generating a free thiol. The thiol 0" the modified phosphopeptide was then covalently bound to a solid support by reacting it with iodoacyl functionalized glass beads followed by cleavage Of the PhOSphate from the solid support was achieved in the presence of acid (Scheme 1-6), 32 I I 0—P-OH H N/VOH 0=P-N/\/OH TFA 0=P"OH : ' H ——-> O EDC ° t-Boc OH t-Boc N t-Boc H \N \N \/\O \ \/\OH H O H O NH2 s—s’ EDC, DTT H N o 2 if ?H S 0 9H SH 0 < o t-Boc III t-Boc N H O H O lTFA OH O=P-OH O O + S H2N V‘OH 0 Scheme 1-6. Phosphoamidate modification-enrichment scheme. This technique was measured for its enrichment efficiency with a radiolabeled peptide formed from 32P-ATP, a tyrosine kinase and myelin basic PTOtein. The labeled protein was digested and its subsequent labeled PhOSphOpeptide underwent the enrichment protocol and its radioactive counts were measured after each step, determining an enrichment recovery of ~ 20%. Furthermore, this approach was introduced to a yeast Iysate, where multiple Phosphopeptides were enriched and their corresponding phosphoproteins were identified.130 In a more recent publication, Aebersold and co-workers performed 33 the enrichment by implementing the same EDC-mediated formation of the phosphoamidate between an amino functionalized dendrimer and the phosphopeptides.131 This approach first protected the carboxylic acids by converting them to their corresponding methyl esters as previously described by Hunt and co-workers.66 Pflum and co-workers developed an oxidation-reduction method for enrichment utilizing the electrophilic character of the phosphate group, which can be accomplished by activating the phosphate with phosphine and disulfide, then coupling the phosphate to a solid-support bound nucIeophiIe.132 The solid suF>|:Jort-bound phosphoamidate enabled the removal of unphosphorylated Peptides and subsequent hydrolysis of the phosphate liberated the peptide from the support (Scheme 1-7).132 NH2 9H 9H <9“ ,VO 0=I';"'OH MeOH 0=I'3-OH 0:5,-” O HCI O O ACLRRA AcLRRA Pph3 PYSSPY; A LRRA Nln/OH OMe DIPEA DMF C \N OMe H 0 Scheme 1-7. Triphenylphosphine mediated phosphoamidate formation on a SOIId support Additionally, the current work described within this dissertation illustrates a method for the enrichment of phosphorylated serinelthreonine and tyrosine containing substrates using a unique solid phase enrichment technique. The method employs the use of a functionalized solid support, which reacts 34 specifically with the phosphate moiety allowing for non-phosphorylated peptides to be efficiently removed by filtration. This technique is an alternative approach to the solid phase enrichment approaches discussed above. Further elaboration of our approach will be addressed later on within the current work section of this chapter. 1.5 Quantitative phosphoproteomic analysis Upon the development Of phosphoproteomic technologies, additional effort to quantify the phosphopeptides/proteins is important for two different reasons. First, the importance of the development of quantitative phosphoproteomic technologies should not be trivialized. A critical facet of phosphoproteomics is determination of the relative change in abundance of phosphoproteins. The Change in abundance of phosphoproteins is of particular interest when biological systems are exposed to stimuli, under duress, or as a result of pathogenic tissue Signaling. There are a variety of different technologies that have been (and are currently being) developed. In-gel based quantitative protein staining/labeling f0I|C>wed by subsequent MS analysis is a common techniques used in protein quantificationfawfi but it suffers limitations with reproducibility and problems with efficiently separating very large or very small, >200 kDa and <10 kDa proteins.136 Additionally, phosphospecific stains can be utilized in a microarray format instead 0f the traditional gel electrophoresis.137 The most widely employed quantitative method, however, involves the implementation of various isotopic labels followed by sUbsequent MS analysis.69'13°'13& ‘42 35 Secondly, the quantification of the recovered phosphopeptides can be used to determine the efficiency of these enrichment techniques. This information can then be used to identify optimal enrichment conditions and materials. This also can identify biases of different approaches with respect to the type (and extent) of the phosphopeptides enriched. 1.5-a SILAC route to quantification of cellular proteins Stable isotope labeling by amino acids in cell culture (SILAC) involves the metabolic labeling of cellular proteins using stable isotope-coded amino acicls.13‘"1‘"'142 The isotopically heavy and light labeled cell culture samples can be treated under different conditions (i.e. exposed to certain drugs or activators in a particular pathway), then pooled together and the changes in phosphoprotein levels can be determined quantitatively by MS (Figure 1-5). Cell Sample #1: ‘ Cell Sample #2: control-light isotope stressed-heavy isotope "heavy" label IIII "light" label Pooled samples illIIIs FIN-Ire 1-5. SILAC approach using mass spectrometry for the quantification of Protein expression levels between normal cells and cells under stress. 36 Work by Neubert and coworkers monitored the cellular response to stimulation by brain derived neurotrophic factor (BDNF), utilizing SILAC to quantitatively measure changes in the phosphotyrosine proteome in primary neurons.143 Upon stimulation, the cells were lysed, immunoprecipitated with phosphotyrosine antibodies, separated by gel electrophoresis and in-gel digested. The digested samples were analyzed by LC-MS/MS and Neubert found that after stimulation 18 phosphoproteins had ratios greater than 1.25 or less than 0.80 and of those phosphoproteins, 13 had ratios greater than 1.5 or less than 0.67.143 Mann and coworkers illustrated the capability of the SILAC to determine global phosphorylation changes by employing it to identify and quantify phosphorylated proteins expression levels in HeLa cells either unstimulated or stimulated with epidermal growth factor (EGF).69 Different plates of cells were supplemented with one of the following; unlabeled lysine and arginine, or labeled lysine “4” and arginine “6”, or lysine “8” and arginine “10”. The cells were collected after exposure to EGF after 0, 1, 5, 10, and 20 minutes. The collected cellular proteins were further separated by strong-cation exchange chromatography and titanium dioxide chromatography for enrichment of phosphopeptides and then subsequently analyzed by MSIMS (Figure 1-6). 37 Lys “0" Lys "8" Arg ROI Arg I10" 0 min EGF 10 min EGF 1 min EGF 5 min EGF 20 min EGF Pool cell samples /\ Cytoplasm proteins Nuclear proteins 1 digest l digest SCX fractionation SCX fractionation l l Ti02 enrichment Ti02 enrichment Figure 1-6. Scheme for SILAC incorporation and enrichment strategy adapted from 69. The SILAC global quantification and phosphopeptide enrichment by Mann and co-workers led to the detection of more than 10,000 phosphopeptides and the characterization of 6,600 phosphorylation sites (4901 phosphoserine, 670 phosphothreonine, and 103 phosphotyrosine) on 2,244 proteins. In comparison to the SwissProt database, more than 90% of the identified phosphorylation sites within the experiment were found to be novel. As a result of the employed SILAC technique, 1,046 phosphopeptides were found to be more than a 2-fold increase 38 as a result of EGF stimulation, which includes 724 phosphoserine, 106 phosphothreonine, and 53 phosphotyrosine, which all are in some way regulated byEGF. Further analysis on identified phosphopeptides containing more than one phosphorylation site had their corresponding phosphopeptides independently quantified to determine if different phosphorylation sites on the same protein are regulated differently. It was found that 77% of those proteins were regulated differently on different phosphorylation sites. This Observation indicates the utility of targeting post-translational modifications as biomarkers by identifying specific forms of a protein as a result of activation. This could also aid in the identification of drugs targeting inhibition of a specific phosphorylated form of the protein without inhibiting all of the additional functions the protein is responsible for. Ultimately, this study determined there are at least 26 transcription factors, 20 transcriptional co-regulators, 21 proteins involved with regulating cytoskeleton function, 27 phosphopeptides belonging to actin-binding proteins, 12 ubiquitin Iigases, among others that are involved with EGF signal activation. Additional quantitative phosphoproteome studies using SILAC has been 145 performed on mouse liver cells,“4 lung cancer cells, and cervical cancer ceIIs.146 1.5.b The use of stable isotopic labeling or isotope affinity tags to quantify Many LC-MS/MS approaches have been utilized to determine relative protein changes?“149 but the method of choice in accurately comparing protein 39 levels within a sample set is stable isotopic labeling of proteins, either metabolically in cell culture (SILAC) or employing labeling reagents on already extracted proteins. One type of labeling reagent is incorporated within the peptide via nucleophilic attack of the labeling reagent by cysteine residues within the peptide, which is referred to as “isotOpe-coded affinity tag” (ICAT).138'14° The ICAT reagent consists of three sections; the biotin affinity tag, the linker which contains either the light or the heavy label and the reactive portion of the molecule. A newer version of the ICAT reagent is labeled with carbon-13 instead of deuterium and contains an acid-cleavable linker attached the biotin tag.150 The introduction of the ICAT reagent can label two separate protein mixtures (light and heavy), the two mixtures combined, and their proteins are subsequently digested using trypsin. The pooled, labeled digests are first purified using strong ion exchange chromatography (SXC) to remove excess ICAT reagent, separated further by HPLC, and subsequently analyzed by MS to Observe the mass difference in the same protein between the light and heavy chain labels and determine relative abundance differences between the two sample sets.150 ICAT has been successfully used in quantifying protein changes in chromatin,151 mitochondria,152 and microsomes,153 even though there is an issue regarding incomplete coverage with ICAT technology, since the labeling reagent only reacts with cysteines, which are present in only 26.6% of the total human tryptic peptides.154 This labeling approach has also been implemented in the 138,155 analysis of whole cell protein changes, including analysis of prostate cancer 40 cells 156 and pancreatic cancer juice.157 However, this technique is limited with regards to phosphoprotein quantification as this reagent requires the presence of both a cysteine and phosphorylated residue within the same peptide in order to quantify the phosphoprotein. Another approach similar to ICAT but with fewer limitations with respect to phosphoproteome quantification is called iTRAQ, and is comprised of isobaric amine-reactive tagging reagents capable of differentially labeling up to eight samples.158 Pappin and co-workers utilized these reagents to monitor global protein expression of both wild-type and mutant yeast strains.158 In regards to quantification of phosphorylated proteins specifically, the iTRAQ approach has been employed to perform a quantitative comparison between some enrichment materials such as IMAC and Ti02 surfaces.159 Burlingame and coworkers utilized iTRAQ technology to quantitatively monitor differences in the phosphorylation sites from proteins extracted from various brain tissues such as the murine cortex, midbrain, cerebellum, and hippocampus.160 The various brain tissues were digested, isotopically labeled, pooled together, subsequently fractionated by SCX and the phosphopeptides were enriched by TiOz and then analyzed by LC-MS/MS. The results provided the quantification of 1564 phosphorylation sites on 831 unique proteins. Additionally, isotopic labels can be incorporated while simultaneously performing the enrichment. Goshe and co-workers, prepared two yeast protein samples which underwent cysteine oxidation, B-elimination, and Michael addition by either isotopically labeled ethanedithiol-D4 or ethanedithiol-H4 and the two 41 separate samples were then pooled together followed by biotin functionalized affinity tag purification.118 The enriched peptides were subsequently analyzed to compare differences in abundances of the enriched phosphopeptides. The isotopic labeling and enrichment developed by Goshe allowed for not only identification of new phosphorylated proteins, but also changes in abundance of the same phosphopeptides within two different sample sets. In another previously discussed enrichment strategy, Aebersold and co- workers used EDC to condense a phosphopeptide on a dendrimer and quantified the enrichment efficiency to be >35% by labeling the peptides via methyl 131 In esterification using methanol (CH30H) or deuterated methanol (CD30D). the same publication, Aebersold also performed this enrichment-labeling procedure to determine tyrosine phosphorylation sites in human T cells (Jurkat cells). The cells were treated with pervanadate for 2 or 10 minutes, then lysed and immunO-purified using phosphotyrosine antibodies. The immunoprecipitated samples were digested and the carboxylic acids were converted to their methyl- do or methyl-da esters. The labeled peptides underwent the dendrimer enrichment and the products were subsequently analyzed by microcapillary reverse-phase LC-MS/MS. The analysis led to the identification and quantification of 97 phosphotyrosine-containing proteins.131 Additional labels, such as 18O, can be incorporated into peptides by 18O -Iabeled water during 161-163 proteolytic digestion and used to further quantify phosphoprotein differences between two sample sets. 42 1.5.c Quantification of protein phosphorylation using a phosphospecific stain Protein microarrays have been valid tools in the discovery and analysis of protein phosphorylation as well as protein quantification. The concept of Pro-Q Diamond dye, which as previously discussed, selectively binds to the phosphate moiety Of the peptide and can enable the extent of phosphorylation in a sample to be visualized in a microarray without the use Of fluorescently labeled antibodies.137 Initial studies by Patton and co-workers employed this technique using serial dilutions of four phosphorylated proteins and three unphosphorylated proteins. The microarray slides were incubated with Pro-Q Diamond stain, subsequently visualized, and quantified on a microarray analysis scanning system. The stained phosphoprotein standards were quantified to determine if Pro-Q Diamond provided linear dynamic range and the technique was found suitable for the quantification Of proteins at unknown concentrations. The limit Of detection was also determined for each of the phosphoproteins and found to vary between 300-600 femtograms, depending on the protein.137 This approach was employed to detect kinase reactions on microarrays by stamping three specific kinase substrates onto a polyacrylamide slide. The kinase reactions were completed by incubating the array with the appropriate kinase and ATP. The success of the phosphorylation, as a result of the kinase assay, was determined by staining the array with the phospho—specific Pro-Q Diamond dye. It was found that 156 picograms of phosphorylated peptide were labeled with the Pro-Q dye, proving the validity of this approach to quantify phosphoproteins and the changes that take place within the phosphoproteome. 43 This technique was also tested for specificity by arraying a thiophosphorylated peptide against an in vitro phosphorylated tyrosine kinase peptide. The sensitivity of detection by Pro-Q was found to be 10.4 picograms for the phosphotyrosine and 325 femtograms for thiophosphopeptide, which confirmed the stain’s specificity for the phosphate.137 An additional purification tool is a multidimensional separation system, PFZD which can be used for separation and quantification of biological samples‘“‘168 and specifically phosphopeptides when coupled with the Pro-Q Diamond stain. The PFZD technique involves a peptide-(or protein) based 2-D liquid phase fractionation, which has shown to be capable of high-throughput analysis and can be fully automated, enabling excellent reproducibility and great resolution. Liquid-based fraction collection can be utilized in large-scale proteomic separation and profiling.169'170 The use of 2-D fractionation has also been employed to specifically perform differential phosphoprotein mapping. The 2-D separation was carried out by first separating the proteins by their pl via chromatofocusing (CF) and then further separating the cell lysate by RP-HPLC.171 Two breast cancer cell samples were cultured and either treated with a kinase inhibitor (P0173074) or a DMSO control then harvested and 2-D separated. The separated fractions were deposited on a microarray surface and stained with Pro-Q Diamond stain to visualize the presence and relative changes between Iysate samples. Only the fractions corresponding to the phosphorylated proteins detected in the array data were subjected to digestion and subsequent MS analysis. Upon analysis it was 44 found the +25 phosphoproteins were identified and their difference in expression levels between the two sets of lysates were characterized.171 1.6 Application of phosphoproteomic enrichment technologies in global phosphorylation analysis Aside from the initial application of the developed enrichment technologies on commercially available proteins, is the importance of their subsequent use on practical cellular samples. The significance of global phosphoproteomic analysis is imperative in qualitatively studying the impact of a phosphoprotein and the extent that its phosphorylation site has on the function Of healthy cells versus pathogenic cells (i.e. tumorgenic). Global phosphoproteomic analysis allows for the ‘whole picture’ view of the phosphoproteome, providing a more synergistic and cooperative system identification. Instead of looking at one particular protein of interest, global analysis provides information on the complete ramifications of cellular changes occurring as a result of activation or inhibition of one particular protein or pathway. This also provides insight on proteins relationship with one another, either directly or through cross talk. A variety of phosphoproteomic enrichment technologies have been employed either singularly or cooperatively to tackle the sample complexity inherent in global analysis. The ultimate goal of this phosphoproteome enrichment project Is its application towards elucidating phosphorylation sites within a cellular protein of interest. Further elaboration of this aspect of the project will be discussed in more depth in chapter four of this dissertation. 45 1.6.a. The use of IMAC to characterize the phosphoproteome Since IMAC has been the technique that has been the most widely used, it is the most common approach employed to identify the global phosphoprotein in a wide variety Of cell lines and tissue samples. In one of the preliminary global phosphoproteome IMAC enrichment experiments, White and co-workers looked at a colon adenocarcinoma cell line (HT-29) in an effort to profile the phosphorylation sites. Characterization of the phosphopeptides within these cells was accomplished by subjecting the lysates to IMAC capillary chromatography and subsequently analyzing the fractions by LC-MS.172 The general scheme for this enrichment is shown in Figure 1-7. o f”; (96) Cell lysis \ Protein digestion ‘5} © © 0 l >|§ ”W \" © © Trizol . - Trypsin I‘Ew’o't HT—29 cells proteins peptides IMAC Enrichment MASCOT LC/MS/MS a“. \ Identified < I Separated _ 9’- phosphopeptides phosphopeptides attern phosphopeptides explorer Figure 1-7. Global phosphoproteomic analysis via IMAC enrichment (Scheme adapted from figure in 172). As a result of this method, 238 phosphorylation sites were found from HT- 29 cells. The experiment was run in duplicate to determine reproducibility, which 46 was found that about 60% of phosphorylated peptides from one analysis were also identified in a duplicate analysis. The subsequent MASCOT automated sequence explorer was found to give a ‘false-positive’ identification rate of 11.5 %, as a result of large mass errors. Most of the characterized phosphorylation sites originated from phosphoproteins involved in basic structural support or cellular function, such as transcription, translation and mRNA processing. There were some characterized sites, however, that corresponded to proteins involved in signaling pathways including Protein kinase C, AMP-activated protein kinase 0, p53, and MAPK 14."?- In addition to the global study discussed above, quantitative phosphoproteomic analysis was performed in cells observing the tumor necrosis factor (TNF) pathway.146 While the impact of this study directly relates to the pathway we are interested in and will be discussed in more depth in chapter four, its importance in IMAC-based global phosphoproteome enrichment requires its mentioning here. Briefly, whole cell extracts of stimulated and unstimulated cells labeled with either 14N or 15N, digested and separated on a gel-free isoelectric focusing instrument, which separated the peptides by their pl. IEF is a tool of choice based on the general idea that upon phosphorylation the isoelectric point (pl) of a protein or peptide will shift from a higher to lower value based on the additional negative charge from the phosphate moiety. Further enrichment of the IEF fractions collected at pH 3 and 4.6 were subjected to IMAC purification followed by LC-MS. The enriched samples were subsequently pooled together and analyzed by LC-MS/MS/MS. The results identified a total of 701 47 phosphopeptides between the triplicate analyses, characterizing phosphoserine, phosphothreonine and phosphotyrosine form both the 14N and 15N isotopically labeled fractions. From the MSIMS analysis, the comparison between the two labeled fractions led to the quantification of 223 Of the phosphopeptides. This study utilized quantification via isotopic labeling of followed by sequential phosphopeptide purification by both isoelectric focusing and IMAC technologies. Additional global phosphoproteome analysis using IMAC enrichment has been explored by other researchers in a variety of cell lysates.68'100'1”"7‘5 The global phosphoproteomic identification in prostate cancer cells, as reported by Beranova-Giorgianni and co-workers, was performed by tryptic digestion Of the whole cell extract and subsequent IMAC purification.175 The IMAC was performed using gallium spin columns and both the flow-through and elution fraction were collected and analyzed by LC-MS/MS. Upon analysis of the fractions collected, 137 phosphorylation sites were identified from 81 phosphoproteins and out of the 137 phosphorylation sites, 124 sites have previously been identified in various protein databases. For all of the identified phosphopeptides, their corresponding proteins categorized by cellular function and 35% of the proteins were involved with RNA processing and transcriptional regulation, 17% for signal transduction and 14% for translation function. With respect to biomarker identification, one of the phosphoproteins identified in the enrichment study was the kinase BRAF, which is known to be mutated in about 7% of all cancers.177 Another identified phosphorylation site within the study corresponded to heat shock 27-kDa protein (HSP27), which is a typically over- 48 expressed protein in prostate cancer and has been used as biomarker used to determine poor prognosis in prostate cancer patients.178 1.6.b. The use of Ti02 in probing the phosphoproteome Veenstra and co-workers employed Ti02 enrichment technique to probe 179 The cells were harvested and the phosphoproteome of HeLa cells. subsequently labeled with either 18O-water or 16O-water in the presence of trypsin, to quantitatively measure phosphoproteome differences and the digests were enriched for phosphopeptides using a Ti02 column followed by LC-MS analysis.179 When no enrichment was performed prior to analysis, only 2.1% Of the unique peptides identified were phosphorylated. This improved to 17.1% with an enrichment step using 0.1% TFA as the washing solution. Improvement of the enrichment protocol to 38.2% was achieved upon introduction of ammonium glutamate as the washing solution. The subsequent MSIMS analysis identified and characterized a total of 1034 phosphorylation sites from 858 unique phosphopeptides representing 607 proteins. The characterized phosphorylated proteins were found to have a variety of different cellular functions including cytoskeleton maintenance, tumorigenesis, transcription and translational control, cell proliferation and signal transduction. The identified phosphopeptides were cross-validated by comparing the MS2 and MS3 data Obtained to if both sets of data would result in identification of the same phosphopeptide and phosphorylation site. The data was also analyzed manually to determine the rate at which ‘false-positives’ are identified by the 49 automated system. From both sets Of analysis, 24% of the phosphopeptides were identified by the MS2 and MS3 data, while 76% were identified by either MS2 or MS3 data alone. 74 of the 204 identified phosphorylation sites within the study had not been previously reported.179 There were many phosphorylation sites found in this study that were also identified using the same cell line in global phosphorylation study,69 confirming the validity of this approach in cellular phosphoproteome analysis. Recent global phosphoproteomic analysis has been performed by other research groups, also employing TiOz enrichment, albeit preparing the material in a different form in an effort to Obtain higher sensitivity and selective results."‘°'182 Ishirama and co-workers developed an on-Iine ZD-LC-MS system where they manufactured a titania/C18 biphasic column to perform the enrichment of collected HeLa Iysate trypsinized proteins. The subsequent MS analysis yielded 696 non-redundant phosphopeptides from 512 phosphoproteins.180 In a follow-up publication, Mann and co-workers applied a similar global phosphoproteome purification study as previously discussed within this dissertation,69 using SCX and Ti02 enrichment.183 Solid melanoma tumors were homogenized, the Iysate extracted, digested and subsequently purified by either SCX fractionation and Ti02 enrichment or multiple incubations of TiOz enrichment and then analyzed by LC-MS/MS. The SCX-TiOz enrichment sequence led to the identification of 3999 phosphorylation sites, corresponding to 3621 phosphoserine, 355 phosphothreonine and 23 phosphotyrosine. The multiple TiOz incubations enrichment identified 2666 phosphorylation sites 50 consisting of 2385 phosphoserine, 266 phosphothreonine and 15 phosphotyrosine residues. Collectively, 2250 phosphoproteins were identified containing a total of 5250 different phosphorylation sites. 1.6.c. Additional technologies used to determine global phosphorylation While the affinity approaches listed above have proven to be the method of choice in phosphoproteome analysis, there have been additional methods employed to enrich cell Iysates that deserve to be recognized as well. In a previously discussed publication, Aebersold performed the aforementioned phosphoamidate-dendrimer enrichment and methyl esterification labeling procedure to determine tyrosine phosphorylation sites in human T cells (Jurkat cells). The cells were treated with pervanadate for 2 or 10 minutes, then lysed and immunO-purified using phosphotyrosine antibodies. The immunoprecipitated samples were digested and the carboxylic acids were converted to their methyl- do or methyl-d3 esters. The labeled peptides underwent the dendrimer enrichment and the products were subsequently analyzed by microcapillary reverse-phase LC-MS/MS. The analysis led to the identification and quantification of 97 phosphotyrosine-containing proteins.131 Another approach couples the separation ability of 20 gel electrophoresis and an additional enrichment procedure, such as either phospho-specific staining,"""185 IMAC,136 or immunoaffinity,"’7'188 followed by MS analysis to identify phosphoproteins present in cellular extracts. The immunoaffinity approach towards enrichment is an attractive approach for the identification of 51 phosphotyrosine residues as they are less abundant than phosphoserine and phosphothreonine residues. This point is illustrated in the brief summary of some global phosphoproteome studies on different human cell lines, using various enrichment techniques all analyzed by MS, shown in Table 1-1. The use of immunoaffinity for global phosphotyrosine analysis was shown by Rush et al., who harvested Jurkat cells (leukemia cell line), digested the proteins and enriched the phosphopeptides with immobilized anti- phosphotyrosine antibodies.87 The enriched peptides were liberated from the antibody and then separated and analyzed by LC-MS/MS. This strategy identified 194 phosphotyrosine sites from 185 phosphopeptides. Table 1-1. . Unique . Cellular Enrichment . Ref. Cell lIne . . phos.peptides fraction technique (pS e rszhrszyr) pTyr Ab.- 9., . ._ pTyr proteins 131 Jurkat T cells Anti pTyr IP covalent 75 pTyr. sites binding Human HeLa SCX, then , , 69 cells Whole cell “02 (4,901.670.103) Human HeLa SDS-PAGE, phos.peptides 84 cells Nuc'ea' scx 1631219820 Human colon . 172 ad en 0 carcin cm a Whole cell IMAC 238 phos. srtes 174 Humeli(n5?2yeloid Whole cell IMAC 689 phos.peptides Human prostate 137 phos. sites 175 cancer cells Whole cell IMAC 115 phos.peptides Human 10° mesenchymal Whole cell SIMAC 306 monophos, 186 muItIply phos. stem cells Human HeLa . . 179 cells Whole cell TI02 858 phos.peptides Table 1-1 (cont’d). C18-titania 18° Human HeLa Whole cell biphasic 2D— 696 phos.peptides cells LC Human embryonic TiOz chip . 192 kidney 292T Whole cell based HPLC >100 phos.peptides cells Human liver Tissue . 102 tissue samples Iysate SAX 305 phos.peptrdes Human Chang pTyr AD] 2- 16 phos.proteins 184 liver cells Whole cell DE 15 pTyr proteins Human myeloid Multiple Anti- . 187 K562 Whole cell pTyr Ab. 67 pTyr peptides Human A431 . 189 cells, xenograft Whole cell IMAC phgg-gpggllg es tumors ' ' Human 19° mesenchymal Memtbrane TiOz 703 phosphopeptides stem cells pro eIns Jurkat T-cells/ . Combined: 209 phos. 19‘ A549 and HCT- Egg; SDmZPéGE' Sites, 185 phos. 116 peptides P19 embryonal 472 phos. sites on ‘92 carcinoma cells Whole cell MAC 151 phos.proteins Human HeLa 79 phos.peptides (97 145 cells Whole cell IEF” IMAC sites); TNF-regulated 268 phos.peptides 193 Jurkat T-cells Whole cell IMAC tot al.138 pT y r. 190 phos.peptides 19.. ”#3203232 Whole cell IMAC from 152 p phos.proteins 195 Human heart Tissue IM AC 75 phos. sites tissue samples Iysate 47 phos.peptides Table 1-1. Brief summary of some global phosphoproteome studies and the enrichment tool employed 53 1.7 Current work Clearly the phosphate moiety can be manipulated in a plethora of different ways, as illustrated within this chapter. Many research groups employ the 8- elimination-subsequent modification/enrichment material couplings and a few additional groups have designed enrichment approaches targeting the electrophilic character of the phosphate moiety. We have taken the opposite approach and utilized both the nucleophilicity and acidity of the phosphate group to selectively bind to a diazo functionalized solid support.196 The carboxylic acids and C-terrninus are first protected via conversion into methyl esters, leaving the most acidic proton within the peptide to be on the phosphate. Upon a proton transfer by the phosphate to the diazo resin, the solid support becomes activated as a result of the positively charged N2 leaving group. This activation allows for the negatively charged deprotonated oxygen on the phosphate to attack the solid support, simultaneously liberating N2 gas in an irreversible reaction. The phosphopeptide can then be released from the solid support under acidic conditions by hydrolysis of the phosphoester bond (Scheme 1-8). This approach has successfully enriched a phosphorylated peptide from a complex protein digest. O R O HzN—ggerj/ankéi/u‘ome 0 CH2 H O. ,OH 9P 0 \OH O "NM—O O R O H O R o HZN-Ei NW/IL J\ A H N- H \le u EE OMe TFA 2 fix“, #Lngé/ILOMe 0 CH2 _. 0 CH2 H °:p’°H o O. ,OH 0’ \ 9P 0 O \OH Scheme 1-8. Solid phase phosphopeptide enrichment by a diazo-functionalized polystyrene In the upcoming chapters, I will address this approach to phosphopeptide enrichment. The development of the synthesis of the various diazo- functionalized materials and determination of their extent of phosphorylation will be discussed. Additionally, all of the diazo-functionalized materials were all evaluated for their phosphopeptide enrichment capabilities. The Optimal diazo materials were then evaluated for the abilities to enrich phosphorylated peptides from proteolytic digests of commercially available phosphoproteins. The effort towards the application of this technology on a cellular system and future directions will also be addressed. 55 10. REFERENCES Walsh, C. T.; Gameau-Tsodikova, S.; Gatto, G. J. Protein posttranslational modifications: The chemistry of proteome diversifications. Angewandte Chemie-Intemational Edition 2005, 44, (45), 7342-7372. Hanger, D. P.; Betts, J. C.; Loviny, T. L. F.; Blackstock, W. P.; Anderton, B. H. New phosphorylation sites identified in hyperphosphorylated tau (paired helical filament-tau) from Alzheimer's disease brain using nanoelectrospray mass spectrometry. Journal of Neurochemistry 1998, 71, (6), 2465-2476. Maniatis, T.; Tasic, B. Alternative pre-mRNA splicing and proteome expansion in metazoans. Nature 2002, 418, (6894), 236-243. Witze, E. S.; Old, W. M.; Resing, K. A.; Ahn, N. G. Mapping protein post- translational modifications with mass spectrometry. Nature Methods 2007, 4, (10), 798-806. Wold, F. In vivo chemical modification of proteins (post-translational modification). Annual Review of Biochemistry 1981 , 50, 783-814. Graves, J. D.; Krebs, E. G. Protein phosphorylation and signal transduction. Pharmacology & Therapeutics 1999, 82, (2-3), 111-121. Manning, G.; Whyte, D. B.; Martinez, R.; Hunter, T.; Sudarsanam, S. The protein kinase complement of the human genome. Science 2002, 298, (5600), 1912-1916. Miklos, G. L. G.; Maleszka, R. Integrating molecular medicine with functional proteomics: Realities and expectations. Proteomics 2001, 1, (1), 30-41. Hunter, T.; Sefton, B. M. Transforming gene-product of rous-sarcoma virus phosphorylates tyrosine. Proceedings of the National Academy of Sciences of the United States of America-Biological Sciences 1980, 77, (3), 1311-1315. Hubbard, M. J.; Cohen, P. On target with a new mechanism for the regulation of protein-phosphorylation. Trends in Biochemical Sciences 1993, 18, (5), 172-177. 56 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. Venter, J. 0.; Adams, M. D.; Myers, E. W.; Li, P. W.; Mural, R. J.; Sutton, G. G., et al. The sequence of the human genome. Science 2001, 291, (5507), 1304-1351. Sherr, C. J. Cancer cell cycles. Science 1996, 274, (5293), 1672-1677. Klumpp. 3.; Krieglstein, J. Serine/threonine protein phosphatases in apoptosis. Current Opinion in Pharmacology 2002, 2, (4), 458-462. DesboisMouthon, C.; Danan, C.; Amselem, S.; Eggelpoel, M. J. B. V.; SertLangeron, C.; Goossens, M.; Besmond, C.; Capeau, J.; Caron, M. Severe resistance to insulin and insulin-like growth factor-i in cells from a patient with leprechaunism as a result of two mutations in the tyrosine kinase domain of the insulin receptor. Metabolism-Clinical and Experimental 1996, 45, (12), 1493-1500. Saltiel, A. R. Diverse signaling pathways in the cellular actions of insulin. American Journal of Physiology-Endocrinology and Metabolism 1996, 33, (3), E375-E385. Gadsby, D. C.; Naim, A. C. Regulation of CFT R CI- ion channels by phosphorylation and dephosphorylation. Ion Channel Regulation 1999, 33, 79-106. Goedert, M.; Jakes, R.; Spillantini, M. G.; Crowther, R. A.; Cohen, P.; Vanmechelen, E.; Probst, A.; Gotz, J.; Burki, K. Tau-protein in Alzheimers- disease. Biochemical Society Transactions 1995, 23, (1), 80-85. Cohen, P. Protein kinases - the major drug targets of the twenty-first century? Nature Reviews Drug Discovery 2002, 1, (4), 309-315. SopkO, R.; Andrews, B. J. Linking the kinome and phosphorylome - a comprehensive review of approaches to find kinase targets. Molecular Biosystems 2008, 4, (9), 920-933. Moran, M. F.; Tong, J. F.; Taylor, P.; Ewing, R. M. Emerging applications for phospho-proteomics in cancer molecular therapeutics. Biochimica Et Biophysica Acta-Reviews on Cancer 2006, 1766, (2), 230-241. Lim, Y. P. Mining the tumor phosphoproteome for cancer markers. Clinical Cancer Research 2005, 11, (9), 3163-3169. 57 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. Yan, J. X.; Packer, N. H.; GOOley, A. A.; Williams, K. L. Protein phosphorylation: Technologies for the identification Of phosphoamino acids. Journal of Chromatography A 1998, 808, (1 -2), 23-41. D'Ambrosio, C.; Salzano, A. M.; Arena, 8.; Renzone, G.; Scaloni, A. Analytical methodologies for the detection and structural characterization of phosphorylated proteins. Journal of Chromatography B-Analytical Technologies in the Biomedical and Life Sciences 2007, 849, (1-2), 163- 180. Bruce, C.; Shifman, M. A.; Miller, P.; Gulcicek, E. E. Probabilistic enrichment Of phosphopeptides by their mass defect. Analytical Chemistry 2006, 78, (13), 4374-4382. Schroeder, M. J.; Shabanowitz, J.; Schwartz, J. C.; Hunt, D. F.; Coon, J. J. A neutral loss activation method for improved phosphopeptide sequence analysis by quadrupole ion trap mass spectrometry. Analytical Chemistry 2004, 76, (13), 3590-3598. Loyet, K. M.; Stults, J. T.; Arnott, D. Mass spectrometric contributions to the practice of phosphorylation site mapping through 2003 - a literature review. Molecular & Cellular Proteomics 2005, 4, (3), 235-245. Smith, J. C.; Figeys, D. Recent developments in mass spectrometry- based quantitative phosphoproteomics. Biochemistry and Cell Biology- Biochimie Et Biologie Cellulaire 2008, 86, (2), 137-148. Paradela, A.; Albar, J. P. Advances in the analysis of protein phosphorylation. Journal of Proteome Research 2008, 7, (5), 1809-1818. Zhu, K.; Zhao, J.; Lubman, D. M.; Miller, F. R.; Barder, T. J. Protein pl shifts due to posttranslational modifications in the separation and characterization of proteins. Analytical Chemistry 2005, 77, (9), 2745- 2755. Ramasubbu, N.; Reddy, M. S.; Bergey, E. J.; Haraszthy, G. G.; Soni, S. D.; Levine, M. J. Large-scale purification and characterization of the major phosphoproteins and mucins of human submandibular-sublingual saliva. Biochemical Journal 1991, 280, 341 -352. Andersson, L.; Porath, J. Isolation of phosphoproteins by immobilized metal (Fe-3*) affinity-chromatography. Analytical Biochemistry 1986, 154, (1), 250-254. 58 32. 33. 35. 36. 37. 38. 39. 40. 41. 42. Posewitz, M. C.; Tempst, P. Immobilized gallium (Ill) affinity chromatography of phosphopeptides. Analytical Chemistry 1999, 71, (14), 2883-2892. Matsuda, H.; Nakamura, H.; Nakajima, T. New ceramic titania - selective adsorbent for organic-phosphates. Analytical Sciences 1990, 6, (6), 911- 912. Kweon, H. K.; Hakansson, K. Selective zirconium dioxide-based enrichment of phosphorylated peptides for mass spectrometric analysis. Analytical Chemistry 2006, 78, (6), 1743-1749. Ross, A. H.; Baltimore, 0.; Eisen, H. N. Phosphotyrosine-containing proteins isolated by affinity-chromatography with antibodies to a synthetic hapten. Nature 1981, 294, (5842), 654-656. Collins, M. 0.; Yu, L.; Choudhary, J. S. Analysis of protein phosphorylation on a proteome-scale. Proteomics 2007, 7, (16), 2751-2768. Ptacek, J.; Snyder, M. Charging it up: Global analysis of protein phosphorylation. Trends in Genetics 2006, 22, (10), 545-554. Reinders, J.; Sickmann, A. State-of-the-art in phosphoproteomics. Proteomics 2005, 5, (16), 4052-4061. Stasyk, T.; Morandell, S.; Bakry, R.; Feuerstein, I.; Huck, C. W.; Stecher, G.; Bonn, G. K.; Huber, L. A. Quantitative detection of phosphoproteins by combination of two-dimensional difference gel electrophoresis and phosphospecific fluorescent staining. Electrophoresis 2005, 26, (14), 2850-2854. Salih, E. Phosphoproteomics by mass spectrometry and classical protein chemistry approaches. Mass Spectrometry reviews 2005, 24, 828-846. Rall, T. W.; Sutherland, E. W.; Wosilait, W. D. Relationship of epinephrine and glucagon to liver phosphorylase 3. Reactivation of liver phosphorylase in slices and in extracts. Joumal of Biological Chemistry 1956, 218, (1), 483-495. Hunter, T. Protein-kinase classification. Methods in Enzymology 1991, 200, 3-37. 59 43. 45. 46. 47. 48. 49. 50. 51. 52. Sefton, B. M. Measurement of stoichiometry of protein-phosphorylation by biosynthetic labeling. Methods in Enzymology 1991, 201, 245-251. Salih, E.; Zhou, H. Y.; Glimcher, M. J. Phosphorylation of purified bovine bone sialoprotein and osteopontin by protein kinases. Journal of Biological Chemistry 1996, 271, (28), 16897-16905. Salih, E.; Ashkar, S.; Gerstenfeld, L. C.; Glimcher, M. J. Identification of the phosphorylated sites of metabolically p-32-labeled osteopontin from cultured chicken osteoblasts. Journal of Biological Chemistry 1997, 272, (21), 13966-13973. Hathaway, D. R.; Haeberle, J. R. A radioimmunoblotting method for measuring myosin light chain phosphorylation levels in smooth-muscle. American Joumal of Physiology 1985, 249, (3), C345-C351. Bendt, A. K.; Burkovski, A.; Schaffer, S.; Bott, M.; Farwick, M.; Hermann, T. Towards a phosphoproteome map of corynebacterium glutamicum. Proteomics 2003, 3, (8), 1637-1646. Cutting, J. A.; Roth, T. F. Staining of phosphO-proteins on acrylamide-gel electropherograms. Analytical Biochemistry 1973, 54, (2), 386-394. Steinberg, T. H.; Agnew, B. J.; Gee, K. R.; Leung, W. Y.; Goodman, T.; Schulenberg, B., et al. Global quantitative phosphoprotein analysis using multiplexed proteomics technology. Proteomics 2003, 3, (7), 1128-1144. Schulenberg, B.; Aggeler, R.; Beechem, J. M.; Capaldi, R. A.; Patton, W. F. Analysis of steady-state protein phosphorylation in mitochondria using a novel fluorescent phosphosensor dye. Journal of Biological Chemistry 2003, 278, (29), 27251-27255. Larsen, M. R.; Sorensen, G. L.; Fey, S. J.; Larsen, P. M.; Roepstorff, P. Phospho-proteomics: Evaluation of the use of enzymatic de- phosphorylation and differential mass spectrometric peptide mass mapping for site specific phosphorylation assignment in proteins separated by gel electrophoresis. Proteomics 2001, 1, (2), 223-238. Kaufrnann, H.; Bailey, J. E.; Fussenegger, M. Use of antibodies for detection of phosphorylated proteins separated by two-dimensional gel electrophoresis. Proteomics 2001, 1, (2), 194-199. 60 53. 55. 56. 57. 58. 59. 60. 61. 62. Berwick, D. C.; Tavare, J. M. Identifying protein kinase substrates: Hunting for the organ-grinder's monkeys. Trends in Biochemical Sciences 2004, 29, (5), 227-232. Otvos Jr., J. F., E.; Lang, E.; Szendrei, G. l.; Lee, V. M.-Y. Monoclonal antibody PHF-1 recognizes tau protein phosphorylated at serine residues 396 and 404. Journal of Neuroscience Research 1994, 39, 669-673. Zhao, J. Y.; Kuang, J.; Adlakha, R. C.; Rao, P. N. Threonine phosphorylation is associated with mitosis in hela-cells. FEBS Letters 1989, 249, (2), 389-395. Flick, M. B.; Sapi, E.; Perrotta, P. L.; Maher, M. G.; Halaban, R.; Carter, 0.; Kacinski, B. M. Recognition of activated CSF-1 receptor in breast carcinomas by a tyrosine 723 phosphospecific antibody. Oncogene 1997, 14, (21), 2553-2561. Naim, A. C.; Detre, J. A.; Casnellie, J. E.; Greengard, P. Serum antibodies that distinguish between the phospho-forrns and dephosphO-forms of a phosphoprotein. Nature 1982, 299, (5885), 734-736. Czemik, A. J.; Girault, J. A.; Naim, A. C.; Chen, J.; Snyder, G.; Kebabian, J.; Greengard, P. Production of phosphorylation state-specific antibodies. Methods in Enzymology1991, 201, 264-283. Snyder, G. L.; Girault, J. A.; Chen, J. Y. C.; Czemik, A. J.; Kebabian, J. W.; Nathanson, J. A.; Greengard, P. Phosphorylation of DARPP-32 and protein phosphatase inhibitor-1 in rat choroid-plexus - regulation by factors other than dopamine. Journal of Neuroscience 1992, 12, (8), 3071-3083. Bond, 0.; Primrose, D. A.; Foley, E. Quantitative evaluation of signaling events in drosophila s2 cells. Biological Procedures Online 2008, 10, (1), 20-28. lvanov, S. S.; Chung, A. S.; Yuan, 2.; Guan, Y.; Scachs, K. V.; Reichner, J. 8.; Chin, E. Antibodies immobilized as arrays to profile protein post- translational modification in mammalian cells. Molecular & Cellular Proteomics 2004, 3, 788-795. Steen, H.; Jebanathirajah, J. A.; Rush, J.; Morrice, N.; Kirschner, M. W. Phosphorylation analysis by mass spectrometry - myths, facts, and the consequences for qualitative and quantitative measurements. Molecular & Cellular Proteomics 2006, 5, (1 ), 172-181. 61 63. 65. 66. 67. 68. 69. 70. 71. 72. Liu, S.; Zhang, C. J.; Campbell, J. L.; Zhang, H. X.; Yeung, K. K. 0.; Han, V. K. M.; Lajoie, G. A. Formation Of phosphopeptide-metal ion complexes In liquid chromatography/electrospray mass spectrometry and their influence on phosphopeptide detection. Rapid Communications in Mass Spectrometry 2005, 19, (19), 2747-2756. Schmezle, K.; White, F. M. Phosphoproteomic approaches to elucidate cellular signaling networks. Current Opinion in Biotechnology 2006, 17, (4), 406-414. Garcia, B. A.; Shabanowitz, J.; Hunt, D. F. Analysis of protein phosphorylation by mass spectrometry. Methods 2005, 35, 256-264. Ficarro, S. B.; McCleland, M. L.; Stukenberg, P. T.; Burke, D. J.; Ross, M. M.; Shabanowitz, J.; Hunt, D. F.; White, F. M. Phosphoproteome analysis by mass spectrometry and its application to saccharomyces cerevisiae. Nature Biotechnology 2002, 20, (3), 301 -305. Tholey, A.; Reed, J.; Lehmann, W. D. Electrospray tandem mass spectrometric studies of phosphopeptides and phosphopeptide analogues. Journal of Mass Spectrometry 1999, 34, (2), 117—123. Lee, J.; Xu, Y. D.; Chen, Y.; Sprung, R.; Kim, S. C.; Xie, S. H.; Zhao, Y. M. Mitochondrial phosphoproteome revealed by an improved IMAC method and MSIMS/MS. Molecular & Cellular Proteomics 2007, 6, (4), 669-676. Olsen, J. V.; Blagoev, B.; Gnad, F.; Macek, B.; Kumar, 0.; Mortensen, P.; Mann, M., Global, in vivo, and site-specific phosphorylation dynamics in signaling networks. Cell 2006, 127, (3), 635-648. Palumbo, A. M.; Tepe, J. J.; Reid, G. E. Mechanistic insights into the multistage gas-phase fragmentation behavior of phosphoserine- and phosphothreonine-containing peptides. Journal of Proteome Research 2008, 7, (2), 771-779. Palumbo, A. M.; Reid, G. E. Evaluation of gas-phase rearrangement and competing fragmentation reactions on protein phosphorylation site assignment using collision induced dissociation-MSIMS and M83. Analytical Chemistry 2008, 80, (24), 9735-9747. Coon, J. J.; Syka, J. E. P.; Schwartz, J. C.; Shabanowitz, J.; Hunt, D. F. Anion dependence in the partitioning between proton and electron transfer in ion/Ion reactions. International Joumal of Mass Spectrometry 2004, 236, (1-3), 33-42. 62 73. 74. 75. 76. 77. 78. 79. 80. 81. Syka, J. E. P.; Coon, J. J.; Schroeder, M. J.; Shabanowitz, J.; Hunt, D. F. Peptide and protein sequence analysis by electron transfer dissociation mass spectrometry. Proceedings of the National Academy of Sciences of the United States of America 2004, 101, (26), 9528-9533. Zubarev, R. A.; Kelleher, N. L.; McLafferty, F. W. Electron capture dissociation Of multiply charged protein cations. A nonergodic process. Journal of the American Chemical Society 1998, 120, (13), 3265-3266. Savitski, M. M.; Kjeldsen, F.; Nielsen, M. L.; Zubarev, R. A. Hydrogen rearrangement to and from radical 2 fragments in electron capture dissociation of peptides. Joumal of the American Society for Mass Spectrometry 2007, 18, (1 ), 113-120. Zubarev, R. A.; Haselmann, K. F.; Budnik, B.; Kjeldsen, P.; Jensen, F. Towards an understanding of the mechanism of electron-capture dissociation: A historical perspective and modern ideas. European Journal of Mass Spectrometry 2002, 8, (5), 337-349. Shi, S. D. H.; Hemling, M. E.; Carr, S. A.; Horn, D. M.; Lindh, l.; McLafferty, F. W. Phosphopeptide/phosphoprotein mapping by electron capture dissociation mass spectrometry. Analytical Chemistry 2001, 73, (1), 19-22. Stensballe, A.; Jensen, 0. N.; Olsen, J. V.; Haselmann, K. F.; Zubarev, R. A. Electron capture dissociation of singly and multiply phosphorylated peptides. Rapid Communications in Mass Spectrometry 2000, 14, (19), 1793-1800. Creese, A. J.; Cooper, H. F. The effect of phosphorylation on the electron capture dissociation of peptide ions. Journal of the American Society for Mass Spectrometry 2008, 19, (9), 1263-1274. Kjeldsen, F.; Savitski, M. M.; Nielsen, M. L.; Shi, L.; Zubarev, R. A. On studying protein phosphorylation patterns using bottom-up LC-MS/MS: The case of human a-casein. Analyst 2007, 132, (8), 768-776. Sweet, S. M. M.; Mardakheh, F. K.; Ryan, K. J. P.; Langton, A. J.; Heath, J. K.; Cooper, H. J. Targeted online liquid chromatography electron capture dissociation mass spectrometry for the localization of sites of in vivo phosphorylation in human sprouty2. Analytical Chemistry 2008, 80, (17), 6650-6657. 63 82. 83. 85. 86. 87. 88. 89. 90. 91. Zabrouskov, V.; Ge, Y.; Schwartz, J.; Walker, J. W. Unraveling molecular complexity of phosphorylated human cardiac troponin I by top down electron capture dissociation/electron transfer dissociation mass spectrometry. Molecular & Cellular Proteomics 2008, 7, (10), 1838-1849. Schwartz, D.; Gygi, S. P. An iterative statistical approach to the identification of protein phosphorylation motifs from large-scale data sets. Nature Biotechnology 2005, 23, (11), 1391-1398. Beausoleil, S. A.; Jedrychowski, M.; Schwartz, 0.; Elias, J. E.; Villen, J.; Li, J. X.; Cohn, M. A.; Cantley, L. C.; Gygi, S. P. Large-scale characterization of Hela cell nuclear phosphoproteins. Proceedings of the National Academy of Sciences of the United States of America 2004, 101, (33), 12130-12135. Lehr, S.; Kotzka, J.; Herkner, A.; Sikmann, A.; Meyer, H. E.; Krone, W.; Muller-Wieland, D. Identification of major tyrosine phosphorylation sites in the human insulin receptor substrate gab-1 by insulin receptor kinase in vitro. Biochemistry 2000, 39, (35), 10898-10907. Lehr, S.; Kotzka, J.; Herkner, P.; Klein, E.; Siethoff, C.; Knebel, B., et al. Identification of tyrosine phosphorylation sites in human Gab-1 protein by EGF receptor kinase in vitro. Biochemistry 1999, 38, (1), 151-159. Rush, J.; Moritz, A.; Lee, K. A.; Guo, A.; Goss, V. L.; Spek, E. J., et al. lmmunoaffinity profiling of tyrosine phosphorylation in cancer cells. Nature Biotechnology 2005, 23, (1 ), 94-101 . Porath, J.; Carlsson, J.; Olsson, I.; Belfrage, G. Metal chelate affinity chromatography, a new approach to protein fractionation. Nature 1975, 258, (5536), 598-599. Kincshita, E.; Yamada, A.; Takeda, H.; KInoshita-Kikuta, E.; Koike, T. Novel immobilized zinc (ll) affinity chromatography for phosphopeptides and phosphorylated proteins. Journal of Separation Science 2005, 28, (2), 155-162. Jensen, S. S.; Larsen, M. R. Evaluation of the impact of some experimental procedures on different phosphopeptide enrichment techniques. Rapid Communications in Mass Spectrometry 2007, 21, (22), 3635-3645. Pan, C. S.; Ye, M. L.; Liu, Y. G.; Feng, S.; Jiang, X. G.; Han, G. H.; Zhu, J. J.; Zou, H. F. Enrichment of phosphopeptides by Fey-immobilized 64 92. 93. 97. 99. 92. 93. 95. 96. 97. 98. 99. mesoporous nanoparticles of mcm-41 for MALDI and nano-LC-MS/MS analysis. Joumal of Proteome Research 2006, 5, (11), 3114-3124. Yue, G. E.; Roper, M. G.; Balchunas, C.; Pulsipher, A.; Coon, J. J.; Shabanowitz, J.; Hunt, D. F.; Landers, J. P.; Ferrance, J. P. Protein digestion and phosphopeptide enrichment on a glass microchip. Analytica Chimica Acta 2006, 564, (1 ), 116-122. Li, Y. C.; Lin, Y. S.; Tsai, P. J.; Chen, C. T.; Chen, W. Y.; Chen, Y. C. Nitrilotriacetic acid-coated magnetic nanoparticles as affinity probes for enrichment of histidine-tagged proteins and phosphorylated peptides. Analytical Chemistry 2007, 79, (19), 7519-7525. Chen, C. T.; Chen, W. Y.; Tsai, P. J.; Chien, K. Y.; Yu, J. S.; Chen, Y. C. Rapid enrichment Of phosphopeptides and phosphoproteins from complex samples using magnetic particles coated with alumina as the concentrating probes for maldi ms analysis. Journal of Proteome Research 2007, 6, (1 ), 316-325. Ibanez, A. J.; Muck, A.; Svatos, A. Metal-chelating plastic MALDI (p MALDI) chips for the enhancement of phosphorylated-peptide/protein signals. Journal of Proteome Research 2007, 6, (9), 3842-3848. Dunn, J. D.; Igrisan, E. A.; Palumbo, A. M.; Reid, G. E.; Bruening, M. L. Phosphopeptide enrichment using MALDI plates modified with high- capacity polymer brushes. Analytical Chemistry 2008, 80, (15), 5727- 5735. Dunn, J. 0.; Watson, J. T.; Bruening, M. L. Detection of phosphopeptides using Fe(l|I)-nitrilotriacetate complexes immobilized on a MALDI plate. Analytical Chemistry 2006, 78, (5), 1574-1580. Ficarro, S.; Chertihin, 0.; Westbrook, V. A.; White, F.; Jayes, F.; Kalab, P., et al. Phosphoproteome analysis of capacitated human sperm - evidence of tyrosine phosphorylation of a kinase-anchoring protein 3 and valosin- containing protein/p97 during capacitation. Journal of Biological Chemistry 2003, 278, (13), 11579-11589. Salomon, A. R.; Ficarro, S. B.; Brill, L. M.; Brinker, A.; Phung, Q. T.; Ericson, C., et al. Profiling of tyrosine phosphorylation pathways in human cells using mass spectrometry. Proceedings of the National Academy of Sciences of the United States of America 2003, 100, (2), 443-448. 65 til]. II. In It; 100. 101. 102. 103. 104. 105. 106. 107. 108. Thingholm, T. E.; Jensen, 0. N.; Robinson, P. J.; Larsen, M. R. SIMAC (sequential elution from IMAC), a phosphoproteomics strategy for the rapid separation of monophosphorylated from multiply phosphorylated peptides. Molecular & Cellular Proteomics 2008, 7, (4), 661-671. Ballif, B. A.; Villen, J.; Beausoleil, S. A.; Schwartz, D.; Gygi, S. P. Phosphoproteomic analysis Of the developing mouse brain. Molecular & Cellular Proteomics 2004, 3, (11), 1093-1101. Han, G. H.; Ye, M. L.; Zhou, H. J.; Jiang, X. N.; Feng, S.; Jiang, X. G., et al. Large-scale phosphoproteome analysis Of human liver tissue by enrichment and fractionation of phosphopeptides with strong anion exchange chromatography. Proteomics 2008, 8, (7), 1346-1361. Nuhse, T. S.; Bottrill, A. R.; Jones, A. M. E.; Peck, S. C. Quantitative phosphoproteomic analysis of plasma membrane proteins reveals regulatory mechanisms Of plant innate immune responses. Plant Journal 2007, 51, (5), 931-940. Dai, J.; Jin, W. H.; Sheng, Q. H.; Shieh, C. H.; Wu, J. R.; Zeng, R. Protein phosphorylation and expression profiling by yin-yang multidimensional liquid chromatography (yin-yang MDLC) mass spectrometry. Journal of Proteome Research 2007, 6, (1 ), 250-262. Pinkse, M. W. H.; Uitto, P. M.; Hilhorst, M. J.; Ooms, B.; Heck, A. J. R. Selective isolation at the femtomole level of phosphopeptides from proteolytic digests using 2D-nano LC-ESI-MS/MS and titanium oxide precolumns. Analytical Chemistry 2004, 76, (14), 3935-3943. Larsen, M. R.; Thingholm, T. E.; Jensen, 0. N.; Roepstorff, P.; Jorgensen, T. J. D. Highly selective enrichment of phosphorylated peptides from peptide mixtures using titanium dioxide microcolumns. Molecular & Cellular Proteomics 2005, 4, (7), 873—886. Dobson, K. D.; McQuilIan, A. J. In situ infrared spectroscopic analysis of the adsorption of aromatic carboxylic acids to TiOz, ZrOz, Al203, and Ta205 from aqueous solutions. Spectrochimica Acta Part a-Molecular and Biomolecular Spectroscopy 2000, 56, (3), 557-565. Connor, P. A.; McQuilIan, A. J. Phosphate adsorption onto Ti02 from aqueous solutions: An in situ internal reflection infrared spectroscopic study. Langmuir 1999, 15, (8), 2916-2921. 66 109. 110. 111. 112. 113. 114. 115. 116. 117. 118. Klemm, C.; Otto, 8.; Wolf, 0.; Haseloff, R. F.; Beyerrnann, M.; Krause, E. Evaluation of the titanium dioxide approach for ms analysis of phosphopeptides. Journal of Mass Spectrometry 2006, 41, (12), 1623- 1632. Thingholm, T. E.; Jorgensen, T. J. D.; Jensen, 0. N.; Larsen, M. R. Highly selective enrichment of phosphorylated peptides using titanium dioxide. Nature Protocols 2006, 1, (4), 1929-1935. Mazanek, M.; Mituloviae, G.; Herzog, F.; Stingl, C.; Hutchins, J. R. A.; Peters, J. M.; Mechtler, K. Titanium dioxide as a chemo-affinity solid phase in Offline phosphopeptide chromatography prior to HPLC-MSIMS analysis. Nature Protocols 2007, 2, (5), 1059-1069. Li, Y.; Liu, Y.; Tang, J.; Lin, H.; Yao, N.; Shen, X.; Deng, C.; Yang, P.; Zhang, X. Fe304@AI203 magnetic core-shell microspheres for rapid and highly specific capture of phosphopeptides with mass spectrometry analysis. Journal of ChromatographyA 2007, 1172, (1 ), 57-71. Li, Y.; Lin, H. O; Deng, C. H.; Yang, P. Y.; Zhang, X. M. Highly selective and rapid enrichment of phosphorylated peptides using gallium oxide- coated magnetic microspheres for MALDI-TOF-MS and nano—LC-ESI- MSIMS/MS analysis. Proteomics 2008, 8, (2), 238-249. Sturm, M.; Leitner, A.; Smatt, J. H.; Linden, M.; Lindner, W. Tin dioxide microspheres as a promising material for phosphopeptide enrichment prior to liquid chromatography-(tandem) mass spectrometry analysis. Advanced Functional Materials 2008, 18, (16), 2381 -2389. Ficarro, S. B.; Parikh, J. R.; Blank, N. C.; Marto, J. A. Niobium (V) oxide (Nb205): Application to phosphoproteomics. Analytical Chemistry 2008, 80, (12), 4606-4613. Meyer, H. E.; Hoffmannposorske, E.; Korte, H.; Heilmeyer, L. M. G. Sequence-analysis of phosphoserine-containing peptides - modification for picomolar sensitivity. FEBS Letters 1986, 204, (1 ), 61 -66. Oda, Y.; Nagasu, T.; Chait, B. T. Enrichment analysis of phosphorylated proteins as a tool for probing the phosphoproteome. Nature Biotechnology 2001, 19, (4), 379-382. Goshe, M. B.; Conrads, T. P.; Panisko, E. A.; Angeli, N. H.; Veenstra, T. D.; Smith, R. D. Phosphoprotein isotOpe-coded affinity tag approach for 67 119. 120. 121. 122. 123. 124. 125. 126. isolating and quantitating phosphopeptides in proteome-wide analyses. Analytical Chemistry 2001, 73, (11), 2578-2586. Jaffe, H.; Veeranna; Pant, H. C. Characterization of serine and threonine phosphorylation sites in beta-elimination ethanethiol addition-modified proteins by electrospray tandem mass spectrometry and database searching. Biochemistry 1998, 37, (46), 16211-16224. Chowdhury, S. M.; Munske, G. R.; Siems, W. F.; Bruce, J. E. A new maleimide-bound acid-cleavable solid-support reagent for profiling phosphorylation. Rapid Communications in Mass Spectrometry 2005, 19, (7), 899-909. van der Veken, P.; Dirksen, E. H. C.; Ruijter, E.; Elgersma, R. C.; Heck, A. J. R.; Rijkers, D. T. S.; Slijper, M.; Liskamp, R. M. J. Development of a novel chemical probe for the selective enrichment of phosphorylated serine- and threonine-containing peptides. Chembiochem 2005, 6, (12), 2271-2280. Poot, A. J.; Ruijter, E.; Nuijens, T.; Dirksen, E. H. C.; Heck, A. J. R.; Slijper, M.; Rijkers, D. T. S.; Liskamp, R. M. J. Selective enrichment of ser- lthr-phosphorylated peptides in the presence of ser-lthr-glycosylated peptides. Proteomics 2006, 6, (24), 6394—6399. Tseng, H. C.; Ovaa, H.; Wei, N. J. C.; Ploegh, H.; Tsai, L. H. Phosphoproteomic analysis with a solid-phase capture-release—tag approach. Chemistry & Biology 2005, 12, (7), 769-777. McLachlin, D. T.; Chait, B. T. Improved beta-elimination-based affinity purification strategy for enrichment of phosphopeptides. Analytical Chemistry 2003, 75, (24), 6826-6836. Thompson, A. J.; Hart, S. R.; Franz, C.; Bamouin, K.; Ridley, A.; Cramer, R. Characterization of protein phosphorylation by mass spectrometry using immobilized metal ion affinity chromatography with on-resin beta- elimination and michael addition. Analytical Chemistry 2003, 75, (13), 3232-3243. Knight, 2. A.; Schilling, B.; Row, R. H.; Kenski, D. M.; Gibson, B. W.; Shokat, K. M. Phosphospecific proteolysis for mapping sites of protein phosphorylation. Nature Biotechnology 2003, 21, (9), 1047-1054. 68 Ill. Rusr and Jour #28. Rus glit ten 6 w (J7 127. 128. 129. 130. 131. 132. 133. 134. 135. 136. Rusnak, F.; Zhou, J.; Hathaway, G. M. Identification of phosphorylated and glycosylated sites in peptides by chemically targeted proteolysis. Journal of Biomolecular Techniques 2002, 13, (4), 228-237. Rusnak, F.; Zhou, J.; Hathaway, G. M. Reaction Of phosphorylated and o- glycosylated peptides by chemically targeted identification at ambient temperature. Journal of Biomolecular Techniques 2004, 15, (4), 296-304. Zhou, H. L.; Watts, J. D.; Aebersold, R. A systematic approach to the analysis of protein phosphorylation. Nature Biotechnology 2001, 19, (4), 375-378. Chen, X.; Smith, L. M.; Bradbury, E. M. Site-specific mass tagging with stable isotopes in proteins for accurate and efficient protein identification. Analytical Chemistry 2000, 72, (6), 1134-1143. Tao, W. A.; Wollscheid, B.; O'Brien, R.; Eng, J. K.; Li, X. J.; Bodenmiller, 3.; Watts, J. D.; Hood, L.; Aebersold, R. Quantitative phosphoproteome analysis using a dendrimer conjugation chemistry and tandem mass spectrometry. Nature Methods 2005, 2, (8), 591-598. Warthaka, M.; Kanrvowska-Desaulniers, P.; Pflum, M. K. H. Phosphopeptide modification and enrichment by oxidation-reduction condensation. ACS Chemical Biology 2006, 1, (11), 697-701. Orenes-Pinero, E.; Corton, M.; Gonzalez-Peramato, P.; Algaba, F.; Casal, l.; Serrano, A.; Sanchez-Carbayo, M. Searching urinary tumor markers for bladder cancer using a two-dimensional differential gel electrophoresis (2d-dige) approach. Joumal of Proteome Research 2007, 6, (11), 4440- 4448. Tannu, N. S.; Hemby, S. E. Two-dimensional fluorescence difference gel electrophoresis for comparative proteomics profiling. Nature Protocols 2006, 1, (4), 1732-1742. Gronborg, M.; Bunkenborg, J.; Kristiansen, T. Z.; Jensen, 0. N.; YeO, C. J.; Hruban, R. H.; Maitra, A.; Goggins, M. G.; Pandey, A. Comprehensive proteomic analysis of human pancreatic juice. Journal of Pmteome Research 2004, 3, (5), 1042-1055. Lee, H. J.; Kang, M. J.; Lee, E. Y.; Cho, S. Y.; Kim, H.; Paik, Y. K. Application of a peptide-based PF2D platform for quantitative proteomics in disease biomarker discovery. Proteomics 2008, 8, (16), 3371-3381. 69 lII. ‘38. ‘39. ill. I42. 1143. hfiTlf‘ 137. 138. 139. 140. 141. 142. 143. 144. 145. Martin, K. S., T. H.; Cooley, L. A.; Gee, K. R.; Beechem, J. M.; Patton, W. F. Quantitative analysis of protein phosphorylation status and protein kinase activity on microarrays using a novel fluorescent phosphorylation sensor dye. Proteomics 2003, 3, 1244-1255. Gygi, S. P.; Rist, B.; Gerber, S. A.; Turecek, F.; Gelb, M. H.; Aebersold, R. Quantitative analysis of complex protein mixtures using isotope-coded affinity tags. Nature Biotechnology 1999, 17, (10), 994-999. Zhang, Y.; Wolf-Yadlin, A.; Ross, P. L.; Pappin, D. J.; Rush, J.; Lauffenburger, D. A.; White, F. M. Time-resolved mass spectrometry of tyrosine phosphorylation sites in the epidermal growth factor receptor signaling network reveals dynamic modules. Molecular & Cellular Proteomics 2005, 4, (9), 1240-1250. Zhou, H. L.; Ranish, J. A.; Watts, J. D.; Aebersold, R. Quantitative proteome analysis by solid-phase isotope tagging and mass spectrometry. Nature Biotechnology 2002, 20, (5), 512-515. Ong, S. E.; Blagoev, B.; Kratchmarova, |.; Kristensen, D. B.; Steen, H.; Pandey, A.; Mann, M. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Molecular & Cellular Proteomics 2002, 1, (5), 376-386. Zhu, H. N.; Pan, S. Q.; Gu, S.; Bradbury, E. M.; Chen, X. Amino acid residue specific stable isotope labeling for quantitative proteomics. Rapid Communications in Mass Spectrometry 2002, 16, (22), 2115-2123. Spellman, D. S.; Deinhardt, K.; Darie, C. C.; Chao, M. V.; Neubert, T. A. Stable isotopic labeling by amino acids in cultured primary neurons. Molecular & Cellular Proteomics 2008, 7, (6), 1067-1076. Pan, C. G., F.; Olsen, J.; Mann, M. Quantitative phosphoproteome analysis of a mouse liver cell line reveals specificity of phosphatase inhibitors. Proteomics 2008, 8, 4534-4546. Guha, U.; Chaerkady, R.; Marimuthu, A.; Patterson, A. S.; Kashyap, M. K.; Harsha, H. C., et al. Comparisons Of tyrosine phosphorylated proteins in cells expressing lung cancer-specific alleles of EGFR and KRAS. Proceedings of the National Academy of Sciences of the United States of America 2008, 105, (37), 14112-14117. 70 146. 147. 148. 149. 150. 151. 152. 153. 154. Cantin, G. T.; Venable, J. D.; Cociorva, D.; Yates, J. R. Quantitative phosphoproteomic analysis of the tumor necrosis factor pathway. Journal of Proteome Research 2006, 5, (1 ), 127-134. Old, W. M.; Meyer-Arendt, K.; Aveline-Wolf, L.; Pierce, K. G.; Mendoza, A.; Sevinsky, J. R.; Resing, K. A.; Ahn, N. G. Comparison of label-free methods for quantifying human proteins by shotgun proteomics. Molecular & Cellular Proteomics 2005, 4, (10), 1487-1502. Qian, W. J.; Jacobs, J. M.; Camp, D. G.; Monroe, M. E.; Moore, R. J.; Gritsenko, M. A., et al. Comparative proteome analyses of human plasma following in vivo lipopolysaccharide administration using multidimensional separations coupled with tandem mass spectrometry. Proteomics 2005, 5, (2), 572-584. Wiener, M. C.; Sachs, J. R.; Deyanova, E. G.; Yates, N. A. Differential mass spectrometry: A label-free LC-MS method for finding significant differences in complex peptide and protein mixtures. Analytical Chemistry 2004, 76, (20),)6085-6096. Shiio, Y.; Aebersold, R. Quantitative proteome analysis using isotope- coded affinity tags and mass spectrometry. Nature Protocols 2006, 1, (1), 139-145. Shiio, Y.; Donohoe, S.; Yi, E. C.; Goodlett, D. R.; Aebersold, R.; Eisenman, R. N. Quantitative proteomic analysis of myc oncoprotein function. Embo Journal 2002, 21, (19), 5088-5096. Jin, J. H.; Meredith, G. E.; Chen, L.; Zhou, Y.; Xu, J.; Shie, F. S.; Lockhart, P.; Zhang, J. Quantitative proteomic analysis of mitochondrial proteins: Relevance to lewy body formation and parkinson's disease. Molecular Brain Research 2005, 134, (1), 119-138. Han, D. K.; Eng, J.; Zhou, H. L.; Aebersold, R. Quantitative profiling Of differentiation-induced microsomal proteins using isotOpe-coded affinity tags and mass spectrometry. Nature Biotechnology 2001, 19, (10), 946- 951. Zhang, H.; Yan, W.; Aebersold, R. Chemical probes and tandem mass spectrometry: A strategy for the quantitative analysis of proteomes and subproteomes. Cun'ent Opinion in Chemical Biology 2004, 8, 66-75. 71 155. 156. 157. 158. 159. 160. 161. 162. 163. Ideker, T.; Thorsson, V.; Ranish, J. A.; Christmas, R.; Buhler, J.; Eng, J. K., et al. Integrated genomic and proteomic analyses of a systematically perturbed metabolic network. Science 2001, 292, (5518), 929-934. Martin, D. B.; Gifford, D. R.; Wright, M. E.; Keller, A.; Yi, E.; Goodlett, D. R.; Aebersold, R.; Nelson, P. S. Quantitative proteomic analysis of proteins released by neoplastic prostate epithelium. Cancer Research 2004, 64, (1 ), 347-355. Chen, R.; Sheng, P.; Yi, E. C.; Donohoe, S.; Bronner, M. P.; Potter, J. D.; Goodlett, D. R.; Aebersold, R.; Brentnall, T. A. Quantitative proteomic profiling of pancreatic cancer juice. Proteomics 2006, 6, (13), 3871 -3879. Ross, P. L.; Huang, Y. L. N.; Marchese, J. N.; Williamson, 8.; Parker, K.; Hattan, S., et al. Multiplexed protein quantitation in saccharomyces cerevislae using amine-reactive isobaric tagging reagents. Molecular & Cellular Proteomics 2004, 3, (12), 1 154-1 169. Liang, X. Q.; Fonnum, G.; Hajivandi, M.; Stene, T.; Kjus, N. H.; Ragnhildstveit, E.; Amshey, J. W.; Predki, P.; Pope, R. M. Quantitative comparison of IMAC and TiOz surfaces used in the study of regulated, dynamic protein phosphorylation. Journal of the American Society for Mass Spectrometry 2007, 18, (11), 1932-1944. Trinidad, J. C.; Thalhammer, A.; Specht, C. G.; Lynn, A. J.; Baker, P. R.; Schoepfer, R.; Burlingame, A. L. Quantitative analysis of synaptic phosphorylation and protein expression. Molecular & Cellular Proteomics 2008, 7, (4), 684-696. Shevchenko, A.; Chemushevich, l.; Ens, W.; Standing, K. G.; Thomson, 8.; “film, M.; Mann, M. Rapid 'de novo' peptide sequencing by a combination Of nanoelectrospray, isotopic labeling and a quadrupole/time- Of-flight mass spectrometer. Rapid Communications in Mass Spectrometry 1997, 11, (9), 1015-1024. Shevchenko, A.; Shevchenko, A. Evaluation of the efficiency of in-gel digestion of proteins by peptide isotopic labeling and maldi mass spectrometry. Analytical Biochemistry 2001, 296, (2), 279-283. Kosaka, T.; Takazawa, T.; Nakamura, T. Identification and C-terrninal characterization Of proteins from two-dimensional polyacrylamide gels by a combination of isotopic labeling and nanoelectrospray fourier transform ion cyclotron resonance mass spectrometry. Analytical Chemistry 2000, 72, (6), 1179-1185. 72 164. 165. 166. 167. 168. 169. 170. 171. 172. Lee, H. J.; Lee, E. Y.; Kwon, M. S.; Paik, Y. K. Biomarker discovery from the plasma proteome using multidimensional fractionation proteomics. Cun'ent Opinion in Chemical Biology 2006, 10, (1 ), 42-49. Billecke, C.; Malik, l.; Movsisyan, A.; Sulghani, S.; Sharif, A.; Mikkelsenn, T.; Farrell, N. P.; Bogler, 0. Analysis of glioma cell platinum response by metacomparison Of two-dimensional chromatographic proteome profiles. Molecular & Cellular Proteomics 2006, 5, (1 ), 35-42. Falisse—Poirrier, N.; Ruelle, V.; EIMoualij, B.; Zorzi, D.; Pierard, O.; Heinen, E.; De Pauw, E.; Zorzi, W. Advances in immunoproteomics for serological characterization of microbial antigens. Journal of Microbiological Methods 2006, 67, (3), 593-596. Ruelle, V.; Falisse—Poirrier, N.; EIMoualij, B.; Zorzi, D.; Pierard, O.; Heinen, E.; De Pauw, E.; Zorzi, W. An immunO-PFZD-MS/MS proteomic approach for bacterial antigenic characterization: To bacillus and beyond. Journal of Proteome Research 2007, 6, (6), 2168-2175. Shin, Y. K.; Lee, H. J.; Lee, J. S.; Paik, Y. K. Proteomic analysis Of mammalian basic proteins by liquid-based two-dimensional column chromatography. Proteomics 2006, 6, (4), 1143-1150. Xie, H. W.; Bandhakavi, S.; Griffin, T. J. Evaluating preparative isoelectric focusing of complex peptide mixtures for tandem mass spectrometry- based proteomics: A case study in profiling chromatin-enriched subcellular fractions in saccharomyces cerevisiae. Analytical Chemistry 2005, 77, (10), 3198-3207. Heller, M.; Michel, P. E.; Morier, P.; Crettaz, D.; Wenz, C.; Tissot, J. D.; Reymond, F.; Rossier, J. S. Two-stage off-gel (tm) isoelectric focusing: Protein followed by peptide fractionation and application to proteome analysis of human plasma. Electrophoresis 2005, 26, (6), 1174-1188. Pal, M.; Moffa, A.; Sreekumar, A.; Ethier, S. P.; Barder, T. J.; Chinnaiyan, A.; Lubman, D. M. Differential phosphoprotein mapping in cancer cells using protein microarrays produced from 2-D liquid fractionation. Analytical Chemistry 2006, 78, (3), 702-710. Kim, J. E.; Tannenbaum, S. R.; White, F. M. Global phosphoproteome of ht-29 human colon adenocarcinoma cells. Journal of Proteome Research 2005, 4, (4), 1339-1346. 73 173. 174. 175. 176. 177. 178. 179. 180. 181. Schroeder, M. J.; Webb, D. J.; Shabanowitz, J.; Horwitz, A. F.; Hunt, D. F. Methods for the detection of paxillin post-translational modifications and interacting proteins by mass spectrometry. Journal of Proteome Research 2005, 4, (5), 1832-1841. Ndassa, Y. M.; Orsi, C.; Marto, J. A.; Chen, 8.; Ross, M. M. Improved immobilized metal affinity chromatography for large-scale phosphoproteomics applications. Journal of Proteome Research 2006, 5, (10), 2789-2799. Giorgianni, F.; Zhao, Y. X.; Desiderio, D. M.; Beranova-Giorgianni, S. Toward a global characterization of the phosphoproteome in prostate cancer cells: Identification of phosphoproteins in the lncap cell line. Electrophoresis 2007, 28, (12), 2027-2034. Zhang, X.; Ye, J. Y.; Jensen, 0. N.; Roepstorff, P. Highly efficient phosphopeptide enrichment by calcium phosphate precipitation combined with subsequent IMAC enrichment. Molecular & Cellular Proteomics 2007, 6, (11), 2032-2042. Davies, H.; Bignell, G. R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg. S., et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, (6892), 949-954. Comford, P. A.; Dodson, A. R.; Parsons, K. F.; Desmond, A. D.; Woolfenden, A.; Fordham, M.; Neoptolemos, J. P.; Ke, Y. Q.; Foster, C. S. Heat shock protein expression independently predicts clinical outcome in prostate cancer. Cancer Research 2000, 60, (24), 7099-7105. Yu, L. R.; Zhu, Z. Y.; Chen, K. C.; Issaq, H. J.; Dimitrov, D. S.; Veenstra, T. D. Improved titanium dioxide enrichment of phosphopeptides from Hela cells and high confident phosphopeptide identification by cross-validation of MSIMS and MSIMS/MS spectra. Journal of Proteome Research 2007, 6, (11), 4150-4162. Imami, K.; Sugiyama, N.; Kyono, Y.; Tomita, M.; Ishirama, Y. Automated phosphoproteome analysis for cultured cancer cells by two-dimensional nanOLC-MS using a calcined titania/C18 biphasic column. Analytical Sciences 2008, 24, (1), 161-166. Li, Y.; Xu, X. Q.; Qi, D. W.; Deng, C. H.; Yang, P. Y.; Zhang, X. M. Novel Fe304@Ti02 core-shell microspheres for selective enrichment of phosphopeptides in phosphoproteome analysis. Joumal of Proteome Research 2008, 7, (6), 2526-2538. 74 -.-—-.—._,. 182. 183. 184. 185. 186. 187. 188. 189. 190. Mohammed, S.; Kraiczek, K.; Pinkse, M. W. H.; Lemeer, S.; Benschop, J. J.; Heck, A. J. R. Chip-based enrichment and nanoLC-MS/MS analysis of phosphopeptides from whole Iysates. Journal of Proteome Research 2008, 7, (4), 1565-1571. Zanivan, S.; Gnad, F.; Wickstrom, S.; Geiger, T.; Macek, 8.; Cox, J.; Fassler, R.; Mann, M. Solid tumor proteome and phosphoproteome analysis by high resolution mass spectrometry. Journal of Proteome Research 2008, 7, 5314-5326. Kim, J. H.; In, Y. J.; Kim, W. K.; Bae, K. H.; Kang, 8.; Lee, S. C. Differential signatures of protein glycosylation and phosphorylation in human chang liver cells induced by TCDD treatment. Toxicology Letters 2008, 178, (1), 20-28. Kristjansdottir, K.; Wolfgeher, D.; Lucius, N.; Angulo, D. S.; Kron, S. J. Phosphoprotein profiling by page-LC-MS/MS. Journal of Proteome Research 2008, 7, (7), 2812-2824. Dubrovska, A.; Souchelnytskyi, S. Efficient enrichment of intact phosphorylated proteins by modified immobilizes metal-affinity chromatography. Proteomics 2005, 5, (18), 4678-4683. Lind, S. B.; Molin, M.; Savitski, M. M.; Emilsson, L.; Astrom, J.; Hedberg, L., et al. lmmunoaffinity enrichments followed by mass spectrometric detection for studying global protein tyrosine phosphorylation. Journal of Proteome Research 2008, 7, (7), 2897-2910. Zheng, H. Y.; Hu, P.; Quinn, D. F.; Wang, Y. K. Phosphotyrosine proteomic study of interferon alpha signaling pathway using a combination of immunoprecipitation and immobilized metal affinity chromatography. Molecular & Cellular Proteomics 2005, 4, (6), 721 -730. Stover, D. R.; Caldwell, J.; Marto, J. A.; Root, K.; Mestan, J.; Stumm, M., et al. Differential phosphoprofiles of EGF and EGFR kinase inhibitor- treated human tumor cells and mouse xenografts. Clinical Proteomics 2004, 1, 69-80. Thingholm, T. E.; Larsen, M. R.; lngrell, C. R.; Kassem, M.; Jensen, 0. N. TiOz-based phosphoproteomic analysis of the plasma membrane and the effects of phosphatase inhibitor treatment. Journal of Proteome Research 2008, 7, (8), 3304-3313. 75 191. 192. 193. 194. 195. 196. Wissing, J.; Jansch, L.; Nimtz, M.; Dieterich, G.; Homberger, R.; Keri, G.; Wehland, J.; Daub, H. Proteomics analysis of protein kinases by target class-selective prefractionation and tandem mass spectrometry. Molecular & Cellular Proteomics 2007, 6, (3), 537-547. Smith, J. C.; Duchesne, M. A.; Tozzi, P.; Ethier, M.; Figeys, D. A differential phosphoproteomic analysis of retinoic acid-treated p19 cells. Journal of Proteome Research 2007, 6, (8), 3174-3186. Brill, L. M.; Salomon, A. R.; Ficarro, S. B.; Mukherji, M.; Stettler-Gill, M.; Peters, E. C. Robust phosphoproteomic profiling of tyrosine phosphorylation sites from human T cells using immobilized metal affinity chromatography and tandem mass spectrometry. Analytical Chemistry 2004, 76, (10), 2763-2772. Gevaert, K.; Staes, A.; Van Damme, J.; De Groot, S.; Hugelier, K.; Demol, H.; Martens, L.; Goethals, M.; Vandekerckhove, J. Global phosphoproteome analysis on human heng hepatocytes using reversed- phase diagonal LC. Proteomics 2005, 5, (14), 3589-3599. Ruse, C. I.; Tan, F. L.; Kinter, M.; Bond, M. Intregrated analysis of the human cardiac transcriptome, proteome and phosphoproteome. Proteomics 2004, 4, (5), 1505-1516. Lansdell, T. A.; Tepe, J. J. Isolation of phosphopeptides using solid phase enrichment. Tetrahedron Letters 2004, 45, 91-93. 76 CHAPTER2 SYNTHESIS OF DIAZO FINCTIONALIZED SOLID SUPPORTS AND QUANTIFICATION OF THE DEGREE OF FUNCTIONALIZATION 2.1 Reactivity Of diazo materials with Bronsted acids As discussed briefly in the previous chapter, the goal of this project is to develop materials capable of selectively and covalently binding to the phosphate moiety of phosphorylated peptides. There are many moieties that can, and have, been used to react with phosphates and more specifically, phosphopeptides by manipulating the electrophilic character of the phosphate. In particular, there are additional approaches used to obtain covalent binding between a solid support and a phosphate. Specifically, a phosphate can become succeptable to nucleophilic attack upon the employment of a carbodiimide, in a similar fashion as a carboxylic acid. Additionally, nucleophilic attack may be obtained upon the implementation of triphenylphosphine to the phosphate. These approaches of employing a carbodiimide“2 or phosphine3 to activate the phosphate moiety have been applied to mediate the covalent coupling between the phosphate and a nucleophile-bound solid support, which then was applied towards phosphopeptide enrichment. As an alternative to manipulating the electrophilic character of the phosphate, deprotonation of the phosphoric acid can result in a nucleophilic oxygen, leading to further reactivity. A moiety that can both act as a proton acceptor, while also harboring a site succesptable towards nucleophilic attack 77 led to the exploration of diazo-functionalized materials and their ability to covalently bind to phosphates. Specifically, diazo carbonyl groups were explored as the adjacent carbonyl adds additional stabilization of the diazo moiety. While this stability of diazo carbonyl compounds enables the ease of preparation of these molecules, they are still reactive towards a myriad of different moieties.“'5 The synthetic utility and versatility of diazo carbonyl compounds has been illustrated in countless publications and elegantly reviewed by Ye and McKervey.4 Although solution chemistry exploiting the diazo carbonyl moiety has been widely explored, limited work has been published pursuing the synthetic 6'10 The main diazo solid support application of this moiety on solid supports. used for synthetic applications have been a-diazo-B-ketoesters. Upon the synthesis of these supports, their subsequent application was aimed towards the preparation of a variety of heterocycles including oxazolones,8 oxazoles,11 2 imidazoles,13 pyrazinones and pyrazines.14 The indoles,7 imidazolones,1 preparation of these molecules enabled by the reactivity of the diazo carbonyl resulted in a metal-catalyzed N-H insertion, while liberating N2 from the support. Additional diazo-functionalized solid supports have been implimented to prepare a variety of compounds by nucleophilic addition of an alcohol to the a carbon of the diazo support, again releasing N2 gas.6 The reactivity of the diazo carbonyl moiety towards nucleophilic addition with Brcnsted acids has also been documented. The addition of hydrogen 78 halides to a diazo carbonyl results in the preparation of a-halogenated ketones.15 The addition of a Bronsted acid towards an a-diazo carbonyl in Scheme 2-1 illustrates the potential of these materials to react with phosphoric acids (i.e. phosphorylated peptides) for enrichment applications, which will be discussed at more length later on. Reactive form W Riki H H H—rA‘ 0 Scheme 2-1. Reactivity of diazo carbonyl moieties towards acids The protonation of the negatively charged carbon in the above resonance form of the diazo carbonyl leads the material to susceptibility towards nucleophilic attack at the a-carbon. Upon the addition of the conjugate base nucleophile, N2 gas is simultaneously liberated. This results in the a-substituted conjugate base carbonyl product, such as the a-halogenated ketone discussed previously. It can then be hypothesized that additional acidic reagents can be used in a similar fashion. 79 2.2 Concept of diazo-phosphate binding The idea behind the covalent binding between the diazo moiety and the phosphate is identical to the use of the Brensted acid in Scheme 2-1; the acidic proton on the phosphate (pH 2.8) activates the diazo carbonyl species followed by the subsequent nucleophilic attack by the corresponding negatively charged oxygen (i.e. the conjugate base). Theoretically, the covalent binding of the a carbon by a phosphoric acid or a phosphorylated peptide should react in the same manner as any other Brcnsted acid, shown in Scheme 2-2. It is hypothesized that upon introduction of an immobilized diazo carbonyl material to a phosphoric acid (or phosphorylated peptide), the subsequent covalent binding (and immobilization) of the phosphate will then allow for its purification from any unbound materials by filtration. This hypothesis led to the both concept and development Of the synthesis of diazo- functionalized materials immobilized on solid supports. The use of these materials in covalent phosphate binding would provide a means of purifying phosphopeptides from a complex mixture, such as a proteolytic digest. The importance and utility of developing a selective phosphopeptide enrichment approach was illustrated in Chapter 1. 80 Reactive form R O O O H (+3.,N ®2N HZN-gg NW/IL J\ )L RJKfN R/U\(?,N + \H’ u §§ OMe H H RJK/Ox /O / HO, \9 H CH2 0 MGOTHYNqu’lkEE-th + ~21 O R 0 Scheme 2-2. Theory behind diazo carbonyl-phosphate (phosphopeptide) binding. chemistry that were addressed in Chapter 1.16 It can also be reasoned that this covalent enrichment approach to purify phosphopeptides can reduce the potential specificity problems associated with affinity chromatography and the limitations inherent to phosphate elimination 81 The current covalent phosphopeptide enrichment strategies typically include the elimination of the phosphate moiety from serine or threonine, under alkaline conditions, creating a Michael acceptor.2'17’19 This allows the peptide to be further modified at the site of the eliminated phosphate moiety. Disadvantages to this approach include undesired reactions at the site of glycosylated amino acids within the peptides, dehydration of unmodified serine or threonine residues, and the inability to functionalize phosphotyrosine sites within peptides. As mentioned earlier in this chapter, other covalent enrichment strategies have been reported that utilize the electrophilic character of the phosphate moiety by employing a carbodiimide“2 or phosphine3 to mediate the coupling between the phosphate and a nucleophilic solid support. It can be envisioned that a diazo-functionalized material can covalent bind to the phosphate moiety of a phosphopeptides and be applied towards the enrichment of phosphorylated serine/threonine and tyrosine containing substrates. The use of a diazo-functionalized solid support to react specifically with the phosphate moiety would allow for non-phosphorylated peptides to be efficiently removed by filtration. Liberation of the phosphorylated substrates from the resin can be accomplished using aqueous acid to hydrolyze the phosphoester bond, while still keeping the phosphate intact. The enriched phosphopeptides can be readily analyzed and characterized by tandem mass spectrometry. 82 2.3 Original synthesis of diazo functionalized solid support The implementation of solid phase enrichment and solid phase purification techniques have become increasingly common in the chemical and pharmaceutical industries.’"°'23 One of the advantages of solid support- mediated purification approaches is the ability to easily isolate the product by filtration with subsequent removal of any unbound materials by washing. Further purification can be Obtained by selective/specific cleavage of the product Off the solid support. Numerous solid phase purification/enrichment techniques for chemical reactions have been reported in the literature to illustrate the utility of this approach in synthetic organic chemistry?“30 Applications of this technology in the form of immobilized reagents have also been applied in total syntheses.“32 The biological equivalent of solid phase enrichment in the field of proteomics has been illustrated by various covalent and non-covalent affinity chromatography techniques. One of the most common solid-phase purification strategies of biological samples is immunoaffinity chromatography?”4 This approach is used to purify proteins by binding to its immobilized antibody.35 Additional solid-phase purification on biological samples has been performed by exploiting the strong biotin-avidin interaction in which a protein or antibody Of interest is covalently bound to biotin and subsequently purified on immobilized avidin.36 Additional examples of purification strategies for biological molecules include synthetically or metabolically driven modifications of proteins followed by subsequent covalent 83 addition by a reactive affinity probe/solid support?”0 This has proven to be an effective tool in protein labeling and protein identification. The use of solid phase enrichment to selectively purify/enrich specific post-translational protein modifications, however, remains less developed. The application of a functionalized solid support to specifically purify a post- translational modified protein from its unmodified counterpart could provide unprecedented insight into protein mediated cell signaling. As illustrated above, our approach is based on the reaction between the diazo functionalized material and the acidic character of the phosphate moiety, allowing for covalent capture of the phosphate functional group. In this approach, the covalent enrichment is performed on a solid support. Protonation of the a carbon of the diazo moiety by the phosphate moiety is followed by nucleophilic attack by the deprotonated oxygen of the phosphate on the a carbon resulting in the liberation of N2 (Scheme 2-3). Scheme 2-3. Scheme for the formation of the covalent phosphoester bond. Utilizing the benefits of solid-phase organic reactions, this enrichment approach employs the functionalization of the reactive diazo carbonyl moiety 84 bound to a solid support, allowing for the subsequent purification/removal of the the peptides that do not contain a phosphate moiety. The initial synthetic strategy to form a diazo-functionalized solid support employed the procedure previously used within the Tepe group to prepare a diazo acetate Wang resin (Scheme 2-4).41 A fluoren-9-ylmethoxycarbonyl (Fmoc)-glycine protected Wang resin first underwent liberation of the Fmoc group in the presence Of piperdine to form the resulting free amino- functionalized Wang resin. The free amine was then oxidized to the corresponding diazo-functionalized Wang resin upon exposure to the HONO gas generated from sulfuric acid and sodium nitrite. “Che . 04L NHFmoc 1.) 20% piperdine, DMF 2.) NaNOz, H20, H2804, CH2CI2 Woes . O £N2 Scheme 24. HONO gas mediated diazotization of an amino-functionalized resin. Upon the formation of the diazo-functionalized Wang resin, the material was filtered to remove any reagents or solvent. Confirmation of the formation of 85 the diazo moiety was confirmed by IR, indicating a corresponding diazo stretch at 2110 cm". While this method was capable of preparing the desired diazo- functionalized material, the diazo IR stretch was never one of the most dominant stretches even though it is supposed to be a very strong stretch. It is hypothesized that the reason for the small peak illustrates a poor conversion of the free amine support to the diazo material. 2.4 Problems associated with original studies A potential reason for the poor diazotization could be a result of the nature Of the reaction; incorporation of a gas across a solid support. As described in the previous section, the preparation of the diazo-functionalized solid support was Obtained by the addition of H2SO4 to a solution of sodium nitrite. Upon the addition of acid, HONO gas was generated and bubbled into a vented flask containing dichloromethane and the amino-functionalized resin (Figure 2-1). The reaction vessel/s in Figure 2-1 illustrates why set-up was time consuming and the nature of the reaction required the absence of water (or any other proton source) in the secondary flask containing the resin, which was accomplished upon purging the system with nitrogen gas. Although this procedure did afford the corresponding diazo resin, the conversion was variable at best and the diazo stretch present in the IR was modest (see Appendix, Figure 2-2). Additionally, the reproducibility of this approach was challenging as one day the reaction would afford a modest diazo stretch, while another day the reaction would yield a significantly smaller diazo 86 stretch. The greatest limitation of this approach was the procedure could only be applied to primary amino functionalized materials, which limited the scope of the materials that could be employed and their subsequent further application towards phosphopeptide enrichment. Amino resin H2804, HNO3 in DCM Figure 2-1 Set-up for diazotization of the amino functionalized resin. As a result of the aforementioned limitations Of this approach, this prompted the effort to develop an improved route to obtaining diazo- functionalized solid supports. While developing a new synthetic strategy, emphasis was placed on developing a route to be applied to a multitude of materials. Additionally, a more “user-friendly" set-up that could be performed in a one-pot manner was also desired. 87 2.5 Improved synthesis Of diazo acetate solid support In the initial effort to move towards a synthetic strategy that could be applied to a multitude of materials, work by Ouihia and co-workers began the concept of a new approach. In their work, they performed a coupling of N- hydroxylsuccimamide to glyoxylic acid p-toluenesulfonyl hydrazone in the presence of dicyclocarbodiimide.42 Upon the coupling of these two materials, the hydrazone was subsequently deprotonated, resulting in the liberation of the tosyl moiety and the formation of the diazo acetate functionalized succinimide (Scheme 2-5). While they used the prepared diazo materials for other synthetic applications, this approach however, provided a potential alternative to diazo moiety preparation. " DCC QN-OH + HOJVNNRQ ———> N-O H O O 0 Scheme 2-5.Diazotization reaction using glyoxylic acid p—toluenesulfonyl hydrazone. This reaction led to pursuing glyoxylic acid p-toluenesulfonyl hydrazone as a potential source of forming the desired diazo acetate moiety. Upon further exploration of the use of this material to prepare diazo-functionalized materials, additional methods were also found. Modifications of those approaches towards the solid support synthesis of the diazo moiety were also investigated. To explore this avenue, however, first the glyoxylic acid p-toluenesulfonyl hydrazone 88 must be synthesized. The material was prepared as previously described by Blankley and co-workers by coupling glyoxylic acid and p- toluenesulfonylhydrazide in the presence of hydrochloric acid to obtain the hydrazone product (Scheme 2-6). Compound 2-1 was purified by recrystallization in 1:2 ethyl acetatezcarbon tetrachloride. This material was prepared on a large, 20 9 scale, with no problems, in a 70% yield.43 0 O O H 26MHO 9 H + H2N\ ’8 ; N O Scheme 245. Synthesis of glyoxylic acid p-toluenesulfonyl hydrazone. Upon the preparation of compound 2-1, its carboxylic acid moiety was activated in the presence of diisopropylcarbodiimide and then subsequently coupled to the Wang resin to form the diazo Wang support in a similar manner to the reaction used by Oihua and co-workers. 89 N=C=N ll WOQTOJQ Resin 2-1 Scheme 2-7. Synthesis of diazo acetate Wang resin using glyoxylic acid p- toluenesulfonyl hydrazone and and carbodiimide. The reason for the use of diisopropylcarbodiimide instead of dicyclohexylcarbodiimide (000) was that the dicyclohexylurea by—product formed as a result of the carboxylic acid-DCC coupling, crashes out in the reaction. Typically, this is advantageous in solution-phase reactions. However, as the product is a resin and purification after the reaction is obtained by filtration, a different carbodiimide, whose resultant urea by-product is soluble in the solvent, was desirable. The initial reaction was run overnight in dioxane, using 6 eq. of the glyoxylic acid p—toluenesulfonyl hydrazone, which did not afford any of the desired diazo-functionalized product. Different time points (1, 2, 3 and 4 hr) and solvents (THF and DMF) were also tried. The conditions that appeared to indicate the largest diazo stretch were at 2 and 3 hr in DMF (see Appendix, 90 Figure 24). Additionally, a different coupling reagent, EDCI, was used instead of DIC for different time points (45 min and 4 hrs.) at 0 °C and room temperature, both with and without lithium hydride. The optimal conditions for the diazotization were found to be using EDCI, 1.1 eq LiH for 45 minutes at room temperature in DCM (see Appendix, Figure 2-5). It is believed that the UH was required to deprotonate the hydrazone to allow for its decomposition in forming the diazo product. The IR data still indicated a prominent broad OH stretch at ~3400 cm", which could be a result of un-reacted starting material. This led to the exploration of converting the carboxylic acid to a moiety with more reactivity than with the use of a carbodiimide. The use of 1-chloro-N,N-2-trimethyl-1- propenylamine has been employed as a chlorinating agent to convert the carboxylic acid into an acid chloride.”45 As illustrated by Corey and co-workers, the preparation of the glyoxylic acid chloride p-toluenesulfonyl hydrazone led to subsequent coupling to an alcohol in the presence of base to obtain the diazo acetate moiety (Scheme 2-8). Corey then used the prepared diazo acetate to perform further chemistry. This approach though, to our knowledge, had not been previously attempted on a solid support. We believed that it could be applied towards the synthesis of the desired diazo acetate Wang resin. If this approach was successful, it could also be applied to additional materials containing either a nucleophilic oxygen or nitrogen. 91 Resins 2-1, 2-2 Scheme 2-8. Optimized route for the formation of diazo Wang resin. The desired diazo acetate Wang resin was prepared using the glyoxylic acid chloride p-tosylhydrazone approach outlined above, instead of the aforementioned previously reported diazotization using HONO gas.41 The free hydroxyl moiety on a Wang resin (0.89 and 2.2 mmol/g loading) was converted into the diazo-functionalized solid support in the presence of glyoxylic acid p-tosylhydrazone,43 1-chloro-N,N—2-trimethyl-1-propenylamine, N,N-dimethylaniline and triethylamine to afford resins 2-1 and 2-2 (Scheme 2- 8), respectively.“*44 Confirmation in the formation of the diazo-functionality was readily obtained using IR, in which the diazo stretch has a distinct absorption at 2107 cm'1 (see Appendix, Figures 2-6, 2-7). The yields of the diazo- functionalization on the solid supports were determined in a modified 92 approach to a previously reported procedure.46 The quantification procedure and its results will be discussed in a later section. 2.6 Application of improved synthesis to functionalize additional diazo supports Once the success of the diazotization using glyoxylic acid chloride p- tosylhydrazone had been confirmed on the diazo acetate Wang resin by IR, implementation of this synthetic approach was applied to additional resins containing hydroxyl moieties. Additionally, it was believed that this route could also be employed to nucleophilic amino functionalized materials as well, as illustrated in Scheme 2-9. 9 >::Nf O RBSiI‘I 2-13 \E \ N2 Resin 244 N2 Figure 2-2. Diazo-functionalized solid supports prepared using Scheme 2-9. 95 2.7 Synthesis of diazo ketone solid supports The reactivity of the diazo-functionality in resins 2-1 thru 2-14 is in large mediated by the resonance stabilization with the neighboring carbonyl moiety.5 As a result, diazo ketone supports were also explored for their potential use in phosphopeptide enrichment. Subsequently, diazo ketone derivatives 2-15 thru 17 were prepared by the addition of oxalyl chloride to a carboxylic acid functionalized solid support. Upon formation of the acid chloride, trimethylsilyl diazo methane was added to achieve the diazo ketone.6 The subsequent work-up for the reaction resulted in liberation of the labile TMS group to afford resins 2-15 thru 2-17. (Scheme 2-10). Confirmation of the diazo moiety was confirmed by the IR stretch at ~2100 cm'1 (see Appendix, Figures 2-20 thru 2-22 resin/silica IR data). Quantification of the extent of diazotization was performed in the same manner used for the previously made diazo acetate and amide solid supports. Quantification and its corresponding results will be discussed in a later section. 0 a C : OH benzene ——Ci>a C :: Cl 1:1 THF/ACN a 3 :NZ reflux, reflux, 50 hr 16 hr Resins 2-17 — 2-20 Scheme 2-10. The synthesis of diazo ketone resins. 96 2.8 Synthesis of phenyl diazo methane solid supports Upon the successful formation of the diazo ketone functionalized solid support, we decided to further explore the range of reactivity of the diazo moiety, as we replaced the neighboring carbonyl moieties with a phenyl group. Clearly this substitution would reduce the stability of the diazo moiety, however, it could yield more reactive towards the phosphate. The phenyl diazomethane resins were prepared using a modified approach as previously reported.47 Using three different polymer-bound aldehyde starting materials (mesh size 50-100, 100-200, and 200-400), each polymer was introduced to 2,4,6-triisopropylbenzenesulfonyl hydrazide to form the corresponding hydrazone. The resulting hydrazone was heated in anhydrous methanol and potassium hydroxide, then within minutes the diazo functionalized material was prepared (Scheme 2-11). It should be noted that the triisopropyl groups were required on the benzene ring to afford the corresponding product. The use of benzenesulfonyl hydrazide in the coupling to the aldehyde and subsequent degradation to the desired diazo product was not obtained. This coincides with previous work done by Reese and co-worker, who performed this reaction in solution phase and noticing a drop in yields when a less alkyl substituted aryl ring was employed.48 97 O I N ,2 \ O=CD=O .4 I 'n 23: O + N _.. , \ll H H 16hr NI fi O—cn/Hi 0 KOH in MeOH 90°C N2 Resins 2-18 - 2-20 Scheme 2-11. Synthesis of phenyl diazo methane polystyrene. Confirmation of the diazo moiety was confirmed by the IR stretch at ~2050 cm'1 (see Appendix, Figures 2-23 thru 2-25 resin/silica IR data). Quantification was performed in the same manner as the aforementioned materials and its corresponding results will be discussed in the next section. 2.9 Quantification of solid support diazo functionalization Upon the successful preparation of the twenty different diazo- functionalized solid supports 2-1 thru 2-20, an effort went in to develop an approach to determine the degree of functionalization for each material. This became less straight-forward than initially anticipated, as quantification of solid supports can be potentially more challenging than for solution materials. As such, unique techniques or methods can be employed to obtain reproducible results.”50 While quantification by IR was attempted, it proved to be inconsistent, which required the development of an alternative method to 98 obtain accurate representative yields of conversions.46 The implementation of the incorporation of fluoren-9- ylmethoxycarbonyl (Fmoc) as a means of quantification has been shown in “5'51 Its use has also been cited to characterize the previous publications. degree of functionalization in commercially available polymers. An established protocol that quantifies Fmoc-derivatized solid supports by measuring the amount of liberated dibenzofulvene (DBF) was modified to quantify the amount of diazo functionalization for each solid support. To determine the overall conversion of the starting alcohol, amine or carboxylic acid resin to its corresponding diazo functionalized solid support, an excess of Fmoc-glycine in DMF was added to obtain the Fmoc coupled resin product in a similar covalent binding manner to that of phosphopeptides. Initially, the Fmoc-glycine solution coupling was extended to two days, however, the resin was analyzed by IR and found a substantial amount of diazo still present (IR stretch at ~2000-2130 cm"). It is believed that this could be due to the difference in acidity of the carboxylic acid (pKa ~5) versus a phosphoric acid (pKa at 2.8). As a result of the hypothesis, the coupling was attempted again in the presence of an equivalent of p-toluene sulfonic acid. Upon reaction monitoring by IR, there was no diazo stretch present after 4 hours. These conditions were then introduced into two separately synthesized batches of each support. The reaction afforded the coupling of the Fmoc-group to the diazo material. The Fmoc-glycine addition to the diazo-functionalized 99 materials was confirmed using IR, resulting in the disappearance of the diazo stretch at 2100 cm'1 and the addition of a new ester stretch at 1720 cm”. The coupled product subsequently underwent liberation of the DBF group in the presence of DBU, which typically proceeds in a quantitative conversion (Scheme 2-12). The use of an Fmoc-glycine standard curve was employed for both batches of each solid support to determine the amount of released DBF (see Appendix, Figure 2-27). 0 O Hoi/HYO .0 O O—DJVNZ 0 s O—CI/‘H TsOH (cat) 5% DBU in DMF DBF C02 + O—D/EL’OVNHZ + 000 l I Measure concentration at 304 nm Scheme 2-12. Quantification of diazo functionalized solid support conversion using Fmoc-glycine coupling. 100 The amount of released DBF was converted back to the total Fmoc- glycine bound to the solid support, which corresponded to the amount of Fmoc-glycine that reacted with the diazo moiety. The amount of liberated DBF was then compared to the manufacturers loading value for the starting material to determine the overall percent diazo conversion. Quantification of each support was performed in two separate trials and then reported as an average (Table 2-1). While these values did indicate high reproducibility, it should be noted that these % diazo conversion values are a “minimum level of conversion” rather than % yield of diazo-formation, as they are a result of liberated DBF, not diazo formation. Out of all the resins (2-1 thru 2-12) prepared as shown in Scheme 2-9, the material found to have the largest percent diazo conversion was the dendrimer (2-7). It is hypothesized the reason for this is due to the length of the linker thereby increasing the accessibility of reagents in both the diazo functionalization and the Fmoc derivatization. In general, it appears that the mesh size is involved in the variability of functionalization for resins 2:1-12 as most of the materials around 100-200 mesh size appear to be the most functionalized (resins 1, 7, 9, 11). It is surprising that resins 1 and 2, however, are not closer in % conversion, as the only difference is in their level of substitution. This could be a result of a loss in accessibility of the hydroxyl moiety in the more functionalized support. Resins 4 and 8 have mesh sizes outside of the 100-200 range, which could explain their relatively low conversion as a result of poor interaction with the reagents as a result of their 101 mesh size. The poor conversion of resin 12 is likely due to the relative instability of the diazo anhydride product formed. The extent of functionalization of the diazo ketone supports 15-16 appear to correlate directly with their degree of substitution, which illustrates the likelihood that reagent accessibility is relative to each materials’ level of substitution. The diazo ketone silica 17 appears to be more accessible as its’ percent diazo conversion is the highest out of the three diazo ketones. The mesh size hypothesis also holds up when comparing the degree of functionalization for the diazomethylbenzene resins 18-20 as well, as resin 19 was found to have the highest degree of functionalization. 102 Table 2-1. Resin Solid support % diazo conversion 1 Diazo wanflesin (0.89 mmoI/g) 72 1 8 2 Diazo wang resin (2.2 mmoI/g) 46 13 3 Diazo amide tentagel 52 1 2 4 Diazo amide megabead 27.8 1 0.1 5 Diazo amide PEGA resin 76 1 2 6 Diazo amide HMPA PEGA resin 53 1 6 7 Diazo amide dendrimer 80 1 3 8 4-diazo acetate polystyrene 25.9 1 0.5 9 Diazo acetate polystyrene 68 1 2 10 Nova Syn TG diazo acetate 40 1 3 11 Nova Syn TG HMP diazo acetate 72 1 8 12 Nova Syn diazo anhydride 23 1 10 13 Diazo acetate silica N/A* 14 Diazo amide silica N/A* 15 Diazo ketone resin (1.0 mmomq) 22.3 1 0.1 16 Diazo ketone resin (2.0 mmol/gL 36 1 7 17 Diazo ketone silica 41 1 3 18 Diazomethylbenzene (50-100 mesh) 39 1 3 19 Diazometfylbenzene goo-200 mesh) 81 1 12 20 Diazomethylbenzene (200-400 mesh) 55 1 2 Table 2-1. Average percent diazo functionalization results from two separate batches of each diazo resin using Fmoc-coupling quantification. *The following materials were generously donated and starting material loading values were not available. 103 2.10 Experimental procedures Materials. All organic chemical reagents were obtained from Sigma Aldrich Chemical Co. All polystyrene resins were purchased from either Novabiochem, Sigma Chemical Co., or Advanced Chemtech. Carboxylic acid silica was purchased from Silicycle. The hydroxyl and amino functionalized silica was generously provided by the Pinniavaia lab. Initial diazo Wang resin synthesis. . O OQJOJLNZ Commercially available Fmoc-Glycine Wang resin 100-200 mesh (0.5 g) was added to 20% diperidine in N,N-dimethylformamide (10 mL). The resulting mixture was stirred at 25° C for 2 hr. The product was filtered off with N,N- dimethylformamide (3x 30 mL) and dichloromethane (3x 30 mL), collected, and placed in dichloromethane (10 mL). An adjacent flask containing NaNOz (4.6 g) in H20 (10 mL) was stirred at 25° C when 10% HZSO4/H20 (5 mL) was added. The resulting HONO gas produced, was bubbled into the dichloromethane/resin mixture. After 17 minutes at 25° C, the resin was filtered off, washed with dichloromethane (4x 30 mL), collected, and stored at 0° C. The resin was characterized by IR. IR: 2105 cm" (diazo stretch) KBr pellet 104 Diazo Wang resin synthesis using diisopropylcarbodiimide. w O OGJOJL—“Z Commercially available Wang resin (0.1 9, ~02 mmol) was added to a solution of glyoxylic acid tosylhydrazone (0.24 g, 10 equiv.) in N,N-dimethylformamide (10 mL) and the solution was stirred at 0 °C for 10 minutes. To the reaction flask a solution of diisopropylcarbodiimide (0.93 mmol) in N,N-dimethylformamide (1.5 mL) was added dropwise and allowed to stir at room temperature for 2 hours. The resin was filtered off, washed with dichloromethane (4x 30 mL), collected, and stored at 0° C. The resin was characterized by IR. IR: 2111 cm'1 (diazo stretch) KBr pellet Diazo Wang resin synthesis using EDCI. H_/ O O‘Q4OflN2 Commercially available Wang resin (0.1 g, ~0.1-0.2 mmol depending on substitution) was added to a solution of glyoxylic acid tosylhydrazone (10 equiv.) in dichloromethane (10 mL). To the reaction flask, EDCI (10 equiv.) and an excess of lithium hydride (1.1 eq) was added and the reaction was allowed to stir at room temperature for 45 minutes. The resin was filtered off, washed with 105 dichloromethane (4x 30 mL), collected, and stored at 0° C. The resin was characterized by IR. IR: 2111 cm'1 (diazo stretch) KBr pellet Diazo acetate/amide resin synthesis. o—ofJR Resins 2-1 - 2-14 Commercially available hydroxyl or amino resin (0.1 g, ~0.03 to 0.3 mmol depending on substitution) was added to a solution of glyoxylic acid tosylhydrazone (10 equiv.) and 1-chloro-N,N-2-trimethyl-1-propenylamine (10 equiv.) in dichloromethane (10 mL). N,N-dimethylaniline (10 equiv.) was added drop-wise to the resulting mixture, then stirred at room temperature for 5 min. Triethylamine (10 equiv.) was added to the mixture and stirred at 0°C for five minutes and allowed to warm to room temperature and stirred for 10 minutes. The resulting resins were washed thoroughly with dichloromethane (3x30 mL) and collected by filtration. IR: ~211o cm'1 (diazo stretch) KBr pellet. 106 Diazo ketone resin synthesis. 5.1, O Resins 2-15 - 2-17 In a 25 mL round bottom flask, commercially available carboxypolystyrene resin (0.5 g) was added to a solution of oxalyl chloride (10 equiv.) in benzene (15 mL) and refluxed at 85° C for 16 hr. The acid chloride product was filtered and washed three times in tetrahydrofuran (30 mL) and three times in dichloromethane (30 mL). The acid chloride (0.25 g) was added to a solution of 1:1 acetonitrile and tetrahydrofuran (15 mL) in a 25 mL round bottom flask. To the flask a solution of trimethylsilyl diazo methane (10 equiv.) was added and stirred at room temperature for 50 hr. The resulting resins were washed thoroughly with tetrahydrofuran (3x30 mL) and then dichloromethane (3x30 mL) and collected by filtration. IR: ~21oo cm“ (diazo stretch) KBr pellet. Phenyl diazo methane resin synthesis. /NZ W Resins 2-18 - 2-20 In a 25 mL round bottom flask, commercially available aldehyde functionalized polystyrene resin (0.5 g) was added to a solution of 2,4,6- triisopropylbenzenesuIfonyl hydrazide (2 equiv.) in tetrahydrofuran (10 mL) and stirred under nitrogen at room temperature for 16 hr. The hydrazone product 107 was filtered and washed three times in tetrahydrofuran (30 mL) and three times in dichloromethane (30 mL). The hydrazone (0.1 g) was added to tetrahydrofuran (10 mL) in a 25 mL round bottom flask. The flask was heated to 90 °C in an oil bath and a solution of potassium hydroxide (0.1 g) in anhydrous methanol (1 mL) was added dropwise. The reaction stirred until the resin turned red (about two minutes). The resin was washed three times in tetrahydrofuran (30 mL) and collected by filtration. l R: ~2050 cm‘1 (diazo stretch) KBr pellet. Determination of diazo functionalization. In a 10 mL vial, the diazo functionalized resin (0.050 g) was added to a solution of Fmoc-glycine (10 equiv.) in 1:2 DMF and tetrahydrofuran (1.5 mL), dry p- toluenesulfonic acid (1 equiv.) was added and stirred overnight. The resin was collected, washed three times in tetrahydrofuran (30 mL), and transferred to a 25 mL round bottom flask containing a solution of DBU (0.04 mL) in N,N- dimethylforrnamide (1.96 mL) and stirred at room temperature for 60 minutes. The reaction was diluted in acetonitrile (8 mL) and an aliquot of solution Containing the liberated Fmoc (2 mL) was removed. The 2 mL aliquot was diluted further in acetonitrile (23 mL) and the absorbance of the solution was measured at 304 nm. The amount of Fmoc present in each sample was then quantified using the prepared F moc standard curve. 108 Preparation of glyoxylic acid tosylhydrazone To a 100 mL water in a 250 mL round bottom flask, add glyoxylic acid (9.2 g, 0.1 mole) and warm in a hot water bath to 60 °C. Meanwhile, in a separate 250 mL round bottom flask add p-toluenesuIfonylhydrazide (18.6 g, 0.1 mole) to a 50 mL aqueous solution of hydrochloric acid (2.5 M) and heat to 60 °C in a hot water bath. Add the p-toluenesulfonylhydrazide solution to the glyoxylic acid flask, while maintaining the 60 °C temperature and then the reaction continued to stir at 60 °C for 10 minutes. The flask is allowed to cool to room temperature and then it was placed at 4 °C overnight to aid in crystalization. The crude product was collected by suction filtration, washed with cold water (4 x 200 mL), filtered off and dried in vacuo. The crude product is dissolved in boiling ethyl acetate (100 mL), filtered to remove any insoluable material and then diluted with carbon tetrachloride (200 mL), cooled to room temperature and placed 4 °C overnight to aid in recrystalization. The recrystallized product was washed with cold 1:2 ethyl acetatezcarbon tentrachloride (300 mL), collected by suction filtration and dried in vacuo. IR (NaCl plate): 3205, 1710, 1350, 1172 cm". 1HNMR (500 MHz, DMSO-d5): 6 = 2.37 (s, 3H), 3.41 (br s, 1H), 7.18 (d, J = 2.5 Hz, 1H), 7.42 (d, J = 6.5 Hz, 2H), 7.70 (d, J = 6.0 Hz, 2H), 12.27 (s, 1H). 13cullllR (500 MHz, DMSO- ds): 6 = 21.02, 127.11, 129.91, 135.74, 137.47, 144.06, 163.61. MS (calc. for M“ = 242.2); found 169.8 (M-CzHgNOZ), 155.0 (M-C2H3N202). 109 Appendix Figure2-3. o 0 (30013449 Resin 2-1 100.0r‘77 -111 s~r— 44* 2111--.,111fl «MI (I I II 99.5 MW I II IIIII I 1‘ MI II 99.0 ' I [I 'I I. I {'1 1 I 11,21,611 IIIIIIIIII III I I I J ' II'I'I ' II I I, l : I ,1, » 11) I I I ‘ I I I I I III I), . II . , 1744-41: I I» I I I I I I 2104.39 I I I I 97.5I I I I II I 1597.67 I 97.0I I] II I I 3440.36 III. : 96.5I I III I 3022-97IL291498 4000 3500 3000 2500 2000 1500 1000 500 Wavenumbers 2-3. IR spectrum of a-diazo ester wang resin (0.89 mmol/g loading) made from HONO gas. 110 Figure 2-4. 0 O o Q .0139 £0 Resin 2-1 1000 ”II II . , . , II ”/ I I’WWIIIII III III III I III II -. I ' I II I ,I;I II II I III I III I ' III I I III I , 1 9: I) II 3404.20 I I 1502.12 92 III I--____--- - - I'M-2020.40 - -__ - -_____ - Wavenumbers 2-4. IR spectrum of o-diazo ester wang resin (0.89 mmol/g loading) made from diisopropylcarbodiimide coupling. 111 Figure 2-5. 0 Q HQJM 1:4IIIIII 99 [VI/J I I III I I I III I III I I I I I 9 \ I \ 92 I 21 1 1 .44 340324 /I 1082.12 90- _ 1451-44 697.72 ‘ I 88* 3025.15I~--—2926.48 / 1598.81 V1168.68 4000 3500 3000 2500 2000 1500 1000 500 Wavenumbers 2-5. IR spectrum of a-diazo ester wang resin (0.89 mmol/g loading) made from EDCI with excess LiH, 45 minutes. 112 Figurl 95 85 8O 75' 70 2+3. 0 Q W34“ Resin 2-1 80‘ I 1511.64 I II I I I 1600.30 I II 2107.74 7°IL.__--_.._.__- -__-. -- .1 - - , , _ 4000 3500 3000 2500 2000 1500 1000 500 Wavenumbers I I I / \ 1451.27 1681.8 /192,oo 2-6. IR spectrum of a-diazo ester wang resin (0.89 mmol/g loading) made from route shown in Scheme 2-9. 113 Fig III 9! 90 85 80 75 2-7_ Figure 2-7. 0 Q W48 Resin 2-2 I I I I v‘ I I I I . I. I . I .I I III I II I 90I I II I I II I II I I I I I - _ I I I I I I III II I III III I III I I I I ' I ~ II /I/ I II I I I 1689.06 I w I 1451.54 1610.73 I II 1512.01 I I- _8°-8-_8 - _ _+ _ ___ I 4000— 3500 3000 2500 2000 1500 Tooo 500 Wavenumbers 2-7. IR spectrum of u-diazo ester wang resin (2.2 mmol/9 loading) made from route shown in Scheme 2-9. 114 Fig II 2‘8. Figure 2-8. 0 HN—/<=N 0'5 2 Resin 2-3 1°8I“"’”’ ’“ I“ * . * III WW I “KI/NFW/IIIIILI III ‘ III 2114.11 I I 95‘ I I I I I 700.14 I I I I I I I I 1.53.34 90 I I 1350.97 I .. I I I I I I I 80I 1107.32 I _--..- -_- - - H. fi- --_ __- -___-. ffig _-_I 4000 3500 3000 2500 2000 1500 1000 500 Wavenumbers I I I I I I I I .I I I I 2-8. IR spectrum of diazo amide tentagel made from route shown in Scheme 2- 9. 115 ,I,I||i (I‘I‘I‘ .AH/I I IIIIIHHIMV n4“ IIIIIII IHI II I III1\1. I.I.|I..I I I II “V .EIIIIIIHIMIIIIWIIIIHIIIII 5 IIIIWIV . . II -II- II- I m 0 2 A1”. I . 0 9 III III I 2 6 4 HMHHMuI 5 1 1| IIIUIIIHI 6 1 1500 1000 500 2000 a O 1% Resin 2-4 Wavenumbers 2500 116 Figure 2-9. (”Vb xww 2920.54 3000 3500 2-9. 4000 2-9. IR spectrum of diazo amide megabead made from route shown in Scheme Figure 2-10. “NJL —~2 Resin 2-5 100~r ‘“‘ _‘ *7 7 v , , 7,, —w——-_4Wr,ir ,, A If ”V I 2109.76 II 90I I I II I II 2871.09 I I 80‘ 1643.06 I 75* 1108.12 MEMHSEOE , ' 3000* “2500‘ 2000.- E00 w 1000 500' Wavenumbers 2-10. IR spectrum of PEGA diazo amide resin made from route shown in Scheme 2-9. 117 Fig 10! 95 85 75- 70 t / Figure 2-11 Resin 2-6 100177777777“ iiiiii Ave 4 ............ KflI’was- I\\ [I I 95‘ “1 fl II \\J 'N ,4." III IMMIN M\/ \I / 2111.68 ”I III I III I III . I I , ' I I I I. I I I 1 I 90* I I IIIIIIIII II III I I I '1‘. IIIIIII I II I II I I I II I I I I I 8" \I II I ,. ' I 2872.05 II I I 80 I 1644.99 I I I I II I 1107.73 70 II 4000 $500 3000 2500 2000 1500 1000 500 Wavenumbers 2-11. IR spectrum of PEGA HMPA diazo acetate resin made from route shown in Scheme 2-9. 118 Fig: Figure 2-12. 1001 “““““ 7* “7 7 ‘ 7 * ’ ' ” ”I II I I I 98I I1 /f\ II I II III III I 967 [WWII/I II IIIIIII I IIIII W 94 II I III I I II III , I I I ,I. ' II 92‘ 2115.76 II III/VIII I II II = 90* d I I 1 643.22 I I II I _ ____1 ___J _T~_... _-- ’ - _. v i *7 * *7 ‘— vw‘ fl .. _... 6000 2500 2000 1500 _1000" __ 500 Wavenumbers 4000 35:00 2-12. IR spectrum of diazo amide dendrimer made from route shown in Scheme 2-9. 119 Fig 70 66 62 Figure 2-13. Resin 2-8 "I II; I‘ I I INI II II I II 66I I/III If I I I I /"I I I III I I I, II I’III II “I / II II I I III I I fir , I' II II II I .I" // II II IIIIII I II I I I I I I. III I “I ' I III ‘IIIIII ‘ I IV I III III I II II I I I 2110.46 I IIIII III II I 62 I I 1509.72 II II III I I / III I 1449.43 I III II I I I II I III I 60I I I I II . I IN I I. II I “I___-__-___g_-2_ -__f_ _ M W ; # fl ,2} 4000 3500 3000 2500 2000 1500 1000 50 Wavenumbers 2-13. IR spectrum of 4-butyl diazo acetate polystyrene made from route shown in Scheme 2-9. 120 Figu 100 95 85 80 75 70 Figure 2-14. Resin 2-9 100I,-,r_2 + 2 + + + + - - fl~ #— 2 + ~~—+ ,J ~~~—-~II—- I I I I IVI MI IIIIIIIII I\ '1 . 95I ”VII M \ /I III IIIIIIIII III IIIIIIIIII IIIIII III I III IN I I I' 'II III III III II I II II IIIIII III II I 9“ I I I I'II 'III II ' I II I II I II I II II I I I I II I II II III I 'II I I 85'I ' I I II II II I III I aoI II II II I ‘ I I I ' I'II2849 27 lI694'2/4I49/2l1x I I I II ' I . 1451.14 ' I 75 I 3024.74 I 210835 I I I 2919.67 I "I 696.68 \I I 4000” _ ”9600““ flap—00* * {500 2000 [1600— 1000 .504 Wavenumbers 2-14. IR spectrum of diazo acetate polystyrene made from route shown in Scheme 2-9. 121 Figure . 100 9"." 95 85- 4900 f 245. IF Figure 2-15. C]D-— PEG; -~;3_4<==N2 wMM"’wu*a****/c~*N—/— o \ N2 Resin 2-14 “ mIkaI II I I I III I I /211747 I I 4° W 1625.71I I I 09.96 I I 1471.43 30' I I I II 20‘ I I ”086.28 10 _fll .- fl # _— #fl 4000 3500 3000 2500 2000 1500 1000 5017 Wavenumbers 2-19. IR spectrum of NHNMPTS diazo amide silica made from route shown in Scheme 2-9. 126 _.—.—- u:- Figu I 100‘ 7 95 85' 401 2-20. Figure 2-20. Resin 2-15 1000 2000 2600 95I 90“ 85‘ 1600 Wavenumbers 2-20. IR spectrum of the diazo ketone polystyrene (1.0 mmol/9 loading) made from route shown in Scheme 2-9. 127 Figure 2-2' 2-21. Figure 2-21. Resin 2-16 100 4 ' ' W 7 II " T ”III/1» III I I II I III «I IIIII I .f I 961 W" I I 2 I I . . I I II I II I I 1491.65 _ I / 92 II I 1622.22 I II 1599.64 III II II ”A II I Ii II I I I 2098.05 III aaifi___,_,_._+ __ F _.2 __ _ . . ,.1___ 22% _ f_I 4000 3500 3000 2500 2000 1500 1000 500 Wavenumbers 2-21. IR spectrum of the diazo ketone polystyrene (2.0 mmol/9 loading) made from route shown in Scheme 2-9. 128 Figure 2 100 95 2~22. Figure 2-22. 0 WW Resin 2-17 100_-,,,,,2-, *’*’”“#*“* ******“” I I I I WMAVJXWPJWMfi/\\VAWH I I 2112.01 I 95' 1749.1 _ I 1651.86‘ I i I I °°i I 851 I I 4300“"3500‘ ’ £00 _ 2500 é 531]: “"7530 7600 500 Wavenumbers 2-22. IR spectrum of diazo ketone silica (1.6 mmol/9 loading) made from route shown in Scheme 2-9. 129 Figur 75‘ 70- 55* 2-23. N2 Resin 2-18 ‘hulH InflIIIIII I II III IIIIIII I- III I IIIII. IIII' ‘IIII‘III I IIHI I IIIHIIHUIV III II..I I III .1 III I. I [III III AIIIuIII irIIIIIIII I l IIIIIIIIIHIIglII 5 [J Figure 2-23. 100~ 951 901 051 soI diazo methane polystyrene 2000 (StratoSpheres, 50-100 mesh) made from route shown in Scheme 2-9. Wavenumbers 1 30 2500 IR spectrum of benzyloxy phenyl 75 ‘ 70* 2-23. Figure 100 __ 95‘ 80 75*. Figure 2-24. O< >//N2 Resin 2-19 100 ' I /N I I / /" A III II II II I III I N W" I . I I’ IIIIIII' II , I 955 I In I III I III II” III III / IWII . 2054.87 III I I I I W I 6/ II I I II I 90« ”J I I I V I I III I I ‘ I 1672.91 I I 851 I II I I III I II I /I " I 1478.12 I 754 145325 1373.31 I 4000 3500 3000 2500 2000 1500 1000 500 I Wavenumbers 2—24. IR spectrum phenyl diazo methane polystyrene (100-200 mesh) made from route shown in Scheme 2-9. 131 Figure. 100*“ 70- Figure 2-25. 0 Q 0'04 Resin 2-20 100- M\ I /// \“x/ /fl’II/I III/III ”III II IW 9°‘ / I IV“ I I II II I I / 2051.67 I I I II I / ‘ I I I °°‘ I J I I I I I I I I I I I I I 70I \/ II I I I I I 60" I II I I I II I III I III II I 1480.19I: I I 40» 1453.79 1369.89 I I 4000 3500 3000 2500 2000 1000 1000 500 Wavenumbers 2-25. IR spectrum of benzyloxy phenyl diazo methane polystyrene (ZOO-400 mesh) made from route shown in Scheme 2-9. 132 Figure 2-1 Ce rban Abso ° Std 2.31, Figure 2-26. lfl‘ifl’ii ’ i "‘ A'IIKfJ ‘ ' ‘ _ ‘ ””"T””” R'” ' ' SD 1.81 1_...I ..... 4 , / 1_51_I....- .. . ........ ,,,..//n",.... I 1.411.... . , ._ // I 131.1. . / . I . ' I /( 111—L {/7 f I f I I f I . 7 . r T 0 0.5 1 1 .5 2 Concentration (pmol/mL) Linear Fit: y = A + Bx: A _B_ Q ' Std (Standards@Exp01: Conc vs MeanVaIue) 1.074 0.444 0.994 Absorbance 2.31. Fmoc-glycine standard curve Six concentrations of Fmoc glycine were subjected to Fmoc liberation in the presence of base (DBU). The amount of liberated Fmoc was measured in absorbance at a wavelength of 304 nm. Subsequent analysis to quantify the amount of liberated DBF present was determined by the above standard curve and its corresponding linear equation. 133 References Tao, W. A.; Wollscheid, B.; O'Brien, R.; Eng, J. K.; Li, X. J.; Bodenmiller, 8.; Watts, J. 0.; Hood, L.; Aebersold, R. Quantitative phosphoproteome analysis using a dendrimer conjugation chemistry and tandem mass spectrometry. Nature Methods 2005, 2, (8), 591-598. Zhou, H. L.; Watts, J. D.; Aebersold, R. A systematic approach to the analysis of protein phosphorylation. Nature Biotechnology 2001, 19, (4), 375-378. Warthaka, M.; Karwowska-Desaulniers, P.; Pflum, M. K. H. Phosphopeptide modification and enrichment by oxidation-reduction condensation. ACS Chemical Biology 2006, 1, (11), 697-701. Ye, T.; Mckervey, M. A. Organic-synthesis with d-diazocarbonyl compounds. Chemical Reviews 1994, 94, (4), 1091-1160. Zhang, 2. H.; Wang, J. B. Recent studies on the reactions of o- diazocarbonyl compounds. Tetrahedron 2008, 64, (28), 6577-6605. Iso, Y.; Shindo, H.; Hamana, H. Efficient synthesis of resin-bound a- TMSdiazoketones and their use in solid-phase organic synthesis. Tetrahedron 2000, 56, (30), 5353-5361. Lee, S. H.; Clapham, B.; Koch, G.; Zimmermann, J.; Janda, K. D. Solid- phase rhodium carbenoid N-H insertion reactions: The synthesis of a diverse array of indoles. Journal of Combinatorial Chemistry 2003, 5, (2), 188-196. Yamashita, M.; Lee, S. H.; Koch, G.; Zimmermann, J.; Clapham, B.; Janda, K. D. Solid-phase synthesis of oxazolones and other heterocycles via Wang resin-bound diazocarbonyls. Tetrahedron Letters 2005, 46, (33), 5495-5498. Cano, M.; Camps, F.; Joglar, J. Solid phase synthesis of cyclopropenes. Tetrahedron Letters 1998, 39, (52), 9819-9822. 134 10. 12. 17. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. Zaragoza, F.; Petersen, S. V. Generation of rhodium carbenoids on a polystyrene support and their OH-insertion reaction with alcohols. Tetrahedron 1996, 52, (16), 5999-6002. Clapham, B.; Spanka, C.; Janda, K. D. Solid-phase rhodium carbenoid reactions: An N-H insertion route to a diverse series of oxazoles. Organic Letters 2001, 3, (14), 2173-2176. Lee, S. H.; Clapham, B.; Koch, G.; Zimmermann, J.; Janda, K. D. Rhodium carbenoid N-H insertion reactions of primary ureas: Solution and solid-phase synthesis of imidazolones. Organic Letters 2003, 5, (4), 511- 514. Lee, S. H.; Yoshida, K.; Matsushita, H.; Clapham, B.; Koch, G.; Zimmermann, J.; Janda, K. D. N-H insertion reactions of primary ureas: The synthesis of highly substituted imidazolones and imidazoles from diazocarbonyls. Journal of Organic Chemistry 2004, 69, (25), 8829-8835. Matsushita, H.; Lee, S. H.; Yoshida, K.; Clapham, B.; Koch, G.; Zimmermann, J.; Janda, K. D. N-H insertion reactions of boc-amino acid amides: Solution- and solid-phase synthesis of pyrazinones and pyrazines. Organic Letters 2004, 6, (24), 4627-4629. Mckervey, M. A.; Osullivan, M. B.; Myers, P. L.; Green, R. H. Reductive acylation of d-keto azides derived from L-amino—acids using N-protected L-aminothiocarboxylic S-acids. Journal of the Chemical Society-Chemical Communications 1993, (1 ), 94-96. Garcia, B. A.; Shabanowitz, J.; Hunt, D. F. Analysis of protein phosphorylation by mass spectrometry. Methods 2005, 35, 256-264. Knight, 2. A.; Schilling, B.; Row, R. H.; Kenski, D. M.; Gibson, B. W.; Shokat, K. M. Phosphospecific proteolysis for mapping sites of protein phosphorylation. Nature Biotechnology 2003, 21, (9), 1047-1054. McLachlin, D. T.; Chait, B. T. Improved B—elimination-based affinity purification strategy for enrichment of phosphopeptides. Analytical Chemistry 2003, 75, (24), 6826-6836. Oda, Y.; Nagasu, T.; Chait, B. T. Enrichment analysis of phosphorylated proteins as a tool for probing the phosphoproteome. Nature Biotechnology 2001, 19, (4), 379-382. 135 20. 21. 23. 24. 25. 26. 27. 28. 29. 20. 21. 22 . 23. 24. 25. 26. 27. 28. 29. 30. 31. Bunin, B. A. The combinatorial index. Academic Press: London, 1998. Gordon, E. M.; Kerwin, J. F., Eds.; Combinatorial chemistry and molecular diversity in drug discovery. John Wiley and Sons: New York, 1998. Jung, G., Ed.; Combinatorial chemistry: Synthesis, analysis, screening;. Wiley-VCH: Weinheim, 1999. Obrecht, D.; Villalgordo, J. M. Solid-supported combinatorial and parallel synthesis of small-molecular-weight compound libraries. Elsevier Science: 1998. Czamik, A. W. Special issue on combinatorial chemistry. Accounts in Chemical Research 1996, 29, (3), 112-170. Feliu, L.; Vera-Luque, P.; Albericio, F.; Alvarez, M. Advances in solid- phase cycloadditions for heterocyclic synthesis. Journal of Combinatorial Chemistry 2007, 9, (4), 521-565. Gil, C.; Brase, S. Solid-phase synthesis of biologically active benzoannelated nitrogen heterocycles: An update. Journal of Combinatorial Chemistry 2009, 11, (2), 175—197. Gordon, E. M.; Barrett, R. W.; Dower, W. J.; Fodor, S. P. A.; Gallop, M. A. Applications of combinatorial technologies to drug discovery 2. Combinatorial organic-synthesis, library screening strategies, and future- directions. Journal of Medicinal Chemistry 1994, 37, (10), 1385-1401. Ley, S. V.; Baxendale, I. R.; Brusotti, G.; Caldarelli, M.; Massi, A.; Nesi, M. Solid-supported reagents for multi-step organic synthesis: Preparation and application. Fannaco 2002, 57, (4), 321 -30. Thompson, L. A.; Ellman, J. A. Synthesis and applications of small molecule libraries. Chemical Reviews 1996, 96, (1 ), 555—600. Ley, S. V.; Mynett, D. M.; Koot, W. J. Solid-phase synthesis of bicyclo[2.2.2]octane derivatives via tandem michael addition-reactions and subsequent reductive amination. Synlett1995, (10), 1017-1020. Venter, J. C.; Adams, M. D.; Myers, E. W.; Li, P. W.; Mural, R. J.; Sutton, G. G., et al. The sequence of the human genome. Science 2001, 291, (5507), 1304-1351. 136 32. 33. 35. 36. 37. 38. 39. 40. 41. 42. Storer, R. l.; Takemoto, T.; Jackson, P. S.; Brown, D. S.; Baxendale, I. R.; Ley, S. V. Multi-step application of immobilized reagents and scavengers: A total synthesis of Epothilone C. Chemistry 2004, 10, (10), 2529-2547. Hage, D. S. Survey of recent advances in analytical applications of immunoaffinity chromatography. Journal of Chromatography B-Analytical Technologies in the Biomedical and Life Sciences 1998, 715, (1 ), 3-28. Stevenson, D. lmmuno-affinity solid-phase extraction. Journal of Chromatography B-Analytical Technologies in the Biomedical and Life Sciences 2000, 745, (1 ), 39-48. Herrnanson, G. T.; Mallia, A. K.; Smith, P. K. Immobilized affinity ligand techniques. Academic Press: New York, 1992. Wilchek, M.; Bayer, E. A. The avidin biotin complex in bioanalytical applications. Analytical Biochemistry 1988, 171, (1 ), 1-32. Vocadlo, D. J.; Hang, H. C.; Kim, E. J.; Hanover, J. A.; Bertozzi, C. R. A chemical approach for identifying o-GIcnac-modified proteins in cells. Proceedings of the National Academy of Sciences of the United States of America 2003, 100, (16), 9116-9121. Baskin, J. M.; Bertozzi, C. R. Bioorthogonal click chemistry: Covalent labeling in living systems. Qsar & Combinatorial Science 2007, 26, (11- 12),1211-1219. Fonovic, M.; Verhelst, S. H. L.; Sorum, M. T.; Bogyo, M. Proteomics evaluation of chemically cleavable activity-based probes. Molecular and Cellular Proteomics 2007, 6, (10), 1761-1770. Camarero, J. A.; Kwon, Y. G. Traceless and site-specific attachment of proteins onto solid supports. International Journal of Peptide Research and Therapeutics 2008, 14, (4), 351-357. Lansdell, T. A.; Tepe, J. J. Isolation of phosphopeptides using solid phase enrichment. Tetrahedron Letters 2004, 45, 91-93. Ouihia, A.; Guilhem, J.; Pascard, C.; Badet, B. A new diazoacylating reagent: Preparation, structure, and use of succinimidyl diazoacetate Journal of Organic Chemistry 1993, 58, (7), 1642-1643. 137 43. 48. 50. 51. 43. 45. 46. 47. 48. 49. 50. 51. Blankley, C. J.; Sauter, F. J.; House, H. O. Crotyl diazoacetate. Organic Syntheses 1973, Collection Volume V, 258-263. Corey, E. J.; Myers, A. G. Efficient synthesis and intramolecular cyclopropanation of unsaturated diazoacetic esters. Tetrahedron Letters 1984, 25, (33), 3559-3562. Devos, A.; Remion, J.; Frisquehesbain, A. M.; Colens, A.; Ghosez, L. Synthesis of acyl halides under very mild conditions. Journal of the Chemical Society-Chemical Communications 1979, (24), 1180-1181. Gude, M.; Ryf, J.; White, P. D. An accurate method for the quantitation of frnoc-derivatized solid phase supports. Letters in Peptide Science 2002, 9, (4), 203-206. Bhalay, G.; Dunstan, A. R. Facile solid phase synthesis of an activated diazo linker. Tetrahedron Letters 1998, 39, (42), 7803-7806. Dudman, C. 0.; Reese, C. B. Preparation of aryldiazoalkanes from 2,4,6- triisopropylbenzenesuIphonyl hydrazones. Synthesis 1982, 5, 419-421. Kay, C.; Lorthioir, O. E.; Parr, N. J.; Congreve, M.; McKeown, S. C.; Scicinski, J. J.; Ley, S. V. Solid-phase reaction monitoring - chemical derivatization and off-bead analysis. Biotechnology and Bioengineering 2000, 71, (2), 110-118. Yan, B. Monitoring the progress and the yield of solid-phase organic reactions directly on resin supports. Accounts in Chemical Research 1998, 31, (10), 621-630. Varady, L.; Rajur, S. B.; Nicewonger, R. B.; Guo, M.; Ditto, L. Fast and quantitative high-performance liquid chromatography method for the determination of 9-fiuorenylmethoxycarbonyl release from solid-phase synthesis resins. Journal of ChromatographyA 2000, 869, 171-179. 138 139 AF 31 ofsoI vafiou dscus fifine Of prc Iflobe. Mfih; for pi enncr Gonna appro theh( Seen been Wfihn] by the DUC1e1 CHAPTER 3 APPLICATION OF DIAZO SOLID SUPPORTS IN THE ENRICHMENT OF PHOSPHOPEPTIDES 3 .1 Introduction to diazo solid support phosphopeptide enrichment As briefly mentioned in the previous chapter, the biological equivalent of solid phase enrichment in the field of proteomics has been illustrated by various covalent and non—covalent affinity chromatography techniques. As discussed earlier, some of the most commonly used solid-phase purification strategies include immunoaffinity chromatography“2 or synthetic modifications of proteins followed by subsequent covalent addition by a reactive affinity probe/solid support?"6 This approach has proven to be an effective tool in both protein labeling and protein identification. Covalent solid support purification is therefore an attractive approach for phosphopeptide enrichment. Briefly described in Chapter One was an enrichment method for phosphorylated serine/threonine and tyrosine containing substrates using a diazo-functionalized solid support. This approach first had the carboxylic acid residues and C-terminus protected via their conversion into methyl esters using methanolic hydrochloride as typically seen in IMAC based approaches.7 Once the carboxylic acid residues have been converted to their corresponding methyl esters, the most acidic proton within the peptide is on the phosphate. Upon protonation of the diazo moiety by the phosphate, the resin would become activated and susceptible towards nucleophile attack at the a carbon by the negatively charged oxygen on the 139 phl sol BIT] Sch Dho: phosphate. This material utilizes both the nucleophilicity and acidity of the phosphate group to selectively bind the phosphate to the diazo functionalized solid support, which is unique to any other published phosphopeptide enrichment strategy (Scheme 3-2). 9: I HO \ 40 HO’P\(') HC 0 H 2 M O N Writ Wit-W2 O H H O O— HO’P‘o 2 Ho’P‘o H C O H C O H 2 H 2 M O N M O N ° WY Ykn %%—~H2 ° WY fiukii-NHz O O O R 0 Scheme 3-1. General mechanism for the covalent enrichment of phosphopeptides using a diazo-functionalized solid support. Following trapping of the phosphorylated substrate, non-phosphorylated peptides can be removed by filtration. The phosphorylated substrates are 140 sub bon 3.2 prot subsequently liberated from the resin upon hydrolysis of the phospho-ester bond using aqueous acid. The collected eluent containing the phosphorylated peptides can then be analyzed and sequenced by tandem mass spectrometry. The development of the optimal enrichment conditions/solid support and its subsequent application to successfully enrich phosphorylated substrates from B-casein and ovalbumin digests, in low-picomolar quantities will be discussed herein. 3.2 Preliminary phosphopeptide enrichment from a peptide mixture and a protein digest Initial phosphopeptide enrichment studies used the diazo acetate Wang resin that had been prepared as previously described.8 The diazo- functionalized support was introduced to both a four peptide mix, which contained angiotensin II (a phosphotyrosine peptide), and a B-casein tryptic digest. The amounts of peptide mixture used for the enrichment was 5 nmol, however, only 1/200th of the sample was analyzed (25 pmol). The enrichment did prove successful at this level using both acidic and basic cleavage conditions (see Appendix, Figures 3-6, 3-7). Cleavage under basic conditions yielded the fully hydrolyzed angiotensin II (1127 mlz), while cleavage under acidic conditions kept the methyl ester protecting groups intact on angiotensin II (1155 m/z). Additionally, the B-casein enrichment also did prove successful, using 10 nmol of digest (50 pmol was analyzed). Initially, the digestion was performed on trypsin purchased from Sigma Aldrich. Although the enrichment 141 fill 091 nee he run. U89 and DMF ”DIS: 9003 @YCC IWWII CLam ‘denu did work, there were many ions that corresponded to the same phosphopeptide due to incomplete digestion, which further complicated the spectrum (see Appendix, Figure 3-8). This problem was circumnavigated by the use of sequencing-grade trypsin purchased from Promega. This change resulted in also successful enrichment, albeit, less complicated spectra (see Appendix, Figures 3-9, 3-10). While this approach was successful; in order to be competitive with other current enrichment however, the amount of phosphopeptide used needed to be reduced. This led to the optimization of the enrichment, both in the type of resin used as well as the conditions in which the enrichment was run. A variety of time-points for the enrichment were performed as well as the use of a variety of different solvent systems and cleavage conditions. All of the enriched samples were analyzed by a MALDI-TOF instrument and some samples illustrated successful enrichment (overnight with the use of DMF solvent) by the dominant phosphopeptide ions and the low baseline noise. However, other samples spectra looked poor, containing low phosphopeptide ion abundances and high signal to noise baseline (ethylene glycol enrichment). All of the various enrichment conditions and analysis, however, only provided qualitative data. It was then determined that the implementation of a quantitative enrichment approach would provide more conclusive data to identify the best enrichment conditions. 142 33 rateri phospI amour enliste Io qua the lat 3iia raciole dsadv DhOSp! of the ODfinie appro; Condh. Synthe phOSp 3.3 Evaluation of synthetic phosphopeptide enrichment by diazo supports There were two methods used to determine the optimal enrichment material. The first approach employed the synthesis of a 32P radiolabeled phosphopeptide followed by subsequent enrichment and determination of the amount of recovered 32P radiolabel post enrichment. The other approach enlisted the use of a deuterium-labeled phosphopeptide, which could be used to quantify the amount of unlabeled phosphopeptide in a similar manner as the labeling approach discussed in Chapter 1.9'10 3.3.a Evaluation of diazo supports for enrichment recovery using a radiolabeled phosphopeptide Both quantification approaches have their own advantages and disadvantages. The advantage in the employment of a radiolabeled phosphopeptide for enrichment quantification is its ability to monitor each step of the enrichment process. With this approach, we were able to identify optimal enrichment and cleavage conditions, while MS-based quantification approaches can only measure the “final enriched product”. To determine the phosphopeptide recovery for each of the enrichment conditions used, a 32P radiolabeled phosphopeptide was prepared. A synthetic peptide, CHKtide (KKKVSRSGLYRSpSMPENLNRPR) was phosphorylated with either unlabeled or radiolabeled ATP and checkpoint kinase 2 (CHK-2), using the standard assay protocol (Scheme 3-2).11 143 KKKVSI Schem using confirr to the thru 2 both h Mush; Confin Chlorr the SL paper radiol 32 KKKVSRSGLYRSPSMPENLNRPR 73%- KKKVSRSGLYRSPp3ZSMPENLNRPR Scheme 3-2. Formation of 32P labeled synthetic peptide. The phosphorylated peptide was separated from the remaining ATP using ion exchange chromatography and the phosphorylation status was confirmed by MALDI-ion trap MS analysis of the unlabeled peptide both prior to the phosphorylation assay as well as after (see Appendix, Figures 3-11 thru 3-13). The resulting spectra indicated the presence of the phosphate both in an 80 Da mass shift from 2701 to 2781 as well as MSIMS data which illustrated the -98 loss of H3PO4. A negative control was also performed to confirm the complete removal of the radiolabel during the ion exchange chromatography step, using 32P labeled ATP and CHK-2 enzyme but without the substrate. No radiation was detected by the Geiger counter on the ion exchange paper after the chromatography washes, indicating there was no remaining radiolabeled ATP. After the 32p labeled CHKtide was isolated from the ATP, the carboxylic acid residues and C-terrninus were converted to their corresponding methyl esters with methanolic hydrochloric acid. The peptide was then subjected to enrichment using the diazo acetate resin under a variety of different solvent conditions, DMF and THF were found to correspond to the highest phosphopeptide recovered. Additionally, the conditions in which the phosphopeptides were cleaved off of the solid support were also explored. The use of both NH4OH in water 144 and T The ft Afier most water solut'll hypot the h soluth condl matel thed IMAC FiQUrl peptic and TFA in water were attempted both at 4, 6, and 8 hrs as well as overnight. The resin and the cleavage solution were then surveyed for their radioactivity. After exposure of either of these two solutions to the resin, it revealed that most of the radiolabel was still left on the resin when exposed to NH4OH in water. It was also found that more radiolabel was recovered in the TFA solution, however, the solid support still contained radioactivity. It was then hypothesized that this could be due to a lack of the resin swelling, preventing the hydrolysis. As a result, dichloromethane was added to the cleavage solution, which was found to improve recovery of the radiolabel. These conditions were then subsequently used to compare different enrichment materials. The radiolabeled phosphopeptide was subjected to enrichment using the diazo acetate Wang resin (resin 2-2), diazo ketone resin (resin 2-17), IMAC, T102 or ZrOz pre-packed tips, using their reported protocols (Figure 3- 1). i2;- € I : . ' I. - ~ ; : _g remove ~ g cleave II; . I I _- .1 . I -" ~ I BE 2": ._ 'd 3 i .3 3 add 3 ,3 unbound .- peptl es ITI I. ‘I - Tl»: -’l‘=. 3 I resin ‘2 peptides '3 . .- from reSIn I: "3 ,, : . .- - . “ '.It , ' 3‘ i ' 1‘ ,;' ‘ I' ‘ i“: 3‘3- 2: a72‘ * L3: If a! 1"“ ."-"“"' ““‘"‘-‘¢ 331.; _-_--.;’:’ 11.2323. '- -.-T?§ E‘HNTT‘V’VS Figure 3-1. Enrichment recovery quantification procedure using 32P-labeled pepfide 145 per Tat ehr 9an Once each of the phosphopeptide enrichment techniques were performed, the amount of recovered 32F was determined by scintillation counting and then compared to that of the radiolabeled sample prior to enrichment. Triplicate analyses revealed that the enrichment efficiency was 15% for the diazo ketone resin, while the diazo acetate resin was poorer with only 5% enrichment efficiency. In comparison, the enrichment efficiencies obtained from the IMAC, Ti02 and Zl'02 pre-packed tips were 1%, 14%, and 15%, respectively (Table 3-1). Table 3-1. Enrichment resin % Radiolabeled peptide recovered Diazo ketone resin 2-16 15 (i5)% Diazo acetate wang resin 2-2 3.9 (iO.2)% IMAC Zip-tips 1.0 (i0.1)% Glygen TiOz tips 14 (i4)% Glygen ZI'Oz tips 15 (i0.5)% Table 3-1. Enrichment recovery results using a 32F labeled peptide. This data clearly suggests that the diazo ketone resin is competitive in enrichment capabilities to the commercially available affinity methods. While the use of a radiolabel enables quantification during each step of the enrichment process, there is a disadvantage of this approach. As a result of 146 imple 5am; ehhc ahah radic mass whhl DIOCI 3th D54; Ophr aDDr th05 One. ehnc phOE othEI implementing a radiolabel, no MS analysis is possible on these enrichment samples. This is an issue as the application of this phosphopeptide enrichment technique would be on samples which require MS and MSIMS analysis instead of scintillation counting. Since these samples are only measured for their amount of recovered radiolabel and not whether or not the phosphopeptide ion is observable in a mass spectrometer or purified from other unphosphorylated peptides present within the sample, it is important to obtain quantification of the enrichment procedure as it would take place upon the application of “real” samples. 3.3.b Evaluation of diazo supports for enrichment recovery using an H5 and D5-labeled synthetic phosphopeptide While the radiolabeled enrichment recovery experiment revealed the optimal enrichment conditions, the development of a MS-based quantification approach would be ideal as actual samples undergo the same protocol as those analyzed during the quantification. As discussed previously in Chapter One, various labeling approaches have been implemented in phosphopeptide enrichment both for quantification in enrichment as well as identifying phosphorylation changes in cells as a result of stimulation.9"°'12'18 3 we used In a similar quantification approach discussed previously,1 two synthetic phosphopeptides, one was an unlabeled peptide (H5) and the other was labeled with deuterium (D5). For our quantification study, one of the two peptides will undergo the enrichment and prior to MS analysis, a 147 km 9an ass the phc COL rec! phc sell est enr ml): phc on soII Coll dLF Ont enr San known amount of the other peptide can be added to determine the amount of enriched peptide. Each of the twenty synthesized diazo functionalized resins was assessed for their phosphopeptide enrichment ability in an effort to determine the most efficient enrichment solid support. This study used both a synthetic phosphopeptide, CH3CH2COLFTGHPEpSLEK (H5) and its isotopically labeled counterpart, CDachcOLFTGHPEpSLEK (D5) to determine enrichment recovery. The D5 labeled peptide (10 pmol) was added to a three non- phosphorylated peptide mixture (20 pmol) to additionally confirm phosphate selectivity. Following the conversion of the carboxylic acid moieties to methyl esters,7 the peptide mixture was subsequently exposed to each of the different enrichment materials (~20 mg) overnight in tetrahydrofuran. The enrichment mixture was filtered off to remove unbound peptides and the resin- phosphopeptide sample was subsequently exposed to 50% trifluoroacetic acid on 40% dichloromethane overnight to liberate the phosphopeptide from the solid support. The liberated phosphopeptide samples were subsequently collected and concentrated under nitrogen. The enrichment procedure for each material was carried out in duplicate, desalted and an aliquot (1/10th of the total peptide) was spotted onto a MALDI plate. Prior to ion trap MS analysis, an amount of the H5 methyl ester peptide corresponding to the theoretical 100% recovery of the enriched peptide (1 pmol) was added to the sample plate with the enrichment sample. A comparison of the ion abundances in the resulting mass spectra 148 beh~e pephc avera depu: ldaUI when SUppl differ. Whol enfic! S: a) U —r CD 9: m //T7 / Ff7a/ between the H5 and enriched 05 peptide determined the amount of D5 peptide that was recovered and the two recoveries for each material were averaged and reported in Table 3-2. The enrichment recovery values depicted at < 10% is a result of the amount of noise present in the spectra relative to the enriched ion (see Appendix, Figures 3-14 thru 3-37). Theoretically, the ionization ability of both peptides should be identical when using the same amount of material. However, to take signal suppression Into consideration, a standard curve was performed using five different amounts of 05 while maintaining the same amount of H5 peptide (1 pmol) (see Appendix, Figure 3-38). From the ratio of H5lD5 found in the enrichment spectra, the actual amount of D5 present was determined using the prepared standard curve . Table 3-2. Resin Solid support Recovery 1 Diazo wang resin (0.89 mmol/g) ~ 10 % 2 Diazo wang resin (2.2 mmol/9) < 10 % 3 Diazo amide tentagel 12 % 4 Diazo amide megabead < 10 % 5 Diazo amide PEGA resin < 10 % 6 Diazo amide HMPA PEGA resin 17 % 7 Diazo amide dendrimer 20 % 149 Ta Table 3-2 (cont’d). Resin Solid support Recovery 8 4-diazo acetate polystyrene < 10 % 9 Diazo acetate polystyrene < 10 % 10 Nova Syn TG diazo acetate < 10 % 11 Nova Syn TG HMP diazo acetate < 10 % 12 Nova Syn diazo anhydride < 10 % 13 Diazo acetate silica 15 % 14 Diazo amide silica a 10 % 15 Diazo ketone resin (1.0 mmol/9) 17 % 16 Diazo ketone resin (2.0 mmol/9) 24 % 17 Diazo ketone silica < 10 % 18 Diazomethylbenzene (50-100 mesh) < 10 % 19 Diazomethylbenzene (100-200 mesh) < 10 % 20 Diazomethylbenzene (200-400 mesh) < 10 % - Millipore ZiptipMc (IMAC) < 10 % - Glygen NuTip Ti02 z 10 % - Glygen NuTip ZrOz ~ 10 % Table 3-2. The average enrichment recovery for using each diazo- functionalized solid support. In addition to the quantification of the twenty different diazo- functionalized solid supports, three commercially available affinity enrichment 150 Ies en iOn Cor aCe Coil aVa MS materials; Millipore Ziptips, and Glygen Ti02 or Zl'Oz packed tips were also used for phosphopeptide enrichment following either the manufacturer’s protocol or an established procedure.19 The commercially available materials were found to have enrichment capabilities somewhat less than the most successful diazo resin used. The optimal material based on its phosphopeptide enrichment recovery was found to be the diazo ketone resin 16. Comparatively, the enrichment recovery of 16 and 15 correlates to their level of diazotization illustrated in Table 3-2. This trend was also consistent when comparing materials 1 and 2. The diazo dendrimer support, which was found to have the highest degree of diazo-functionalization, also exhibited a promising enrichment recovery. Unfortunately, the diazo dendrimer enrichment sample also contained additional polymer degradation ions present in the mass spectra as a result of the cleavage conditions making this material less desirable. The diazo amide PEGA HMPA also exhibited promising enrichment recovery as well, but this material also had polymer degradation ions present in the spectra. Additionally, it is interesting to note that the MS quantification data corresponds to the relative radiolabeled data when comparing the diazo acetate Wang resin and the diazo ketone solid support. Both sets of data also coincided with the relative extent of enrichment for each of the commercially available materials. The overall decrease in the enrichment efficiency for the MS quantification could be the result of either the affinity purification and/or the 151 vahabflhj OI enrichml likely du specificil commen they bul which ag E‘Jaluatel ova'buml variability of affinity enrichment depending on the composition of the peptide. Overall, the diazomethylbenzene supports were relatively inefficient in enrichment regardless of their degree of functionalization. This was most likely due to their higher degree of reactivity, which could result in a loss in specificity relative to their diazo carbonyl counterparts. Additionally, it is worth commenting that when these materials were exposed to the peptide mixture, they bubbled substantially, illustrating significant liberation of nitrogen gas, which again could illustrate their reactivity. 3.4 Scope of diazo ketone solid support The ultimate goal of this project was to implement a successful enrichment procedure on a complex peptide mixture, such as a proteolytic digest. By developing the optimal enrichment conditions, as well the most efficient enrichment diazo-functionalized material, the use of this data can be applied to the phosphopeptide enrichment of complex mixtures. 3.4.a Application of diazo ketone enrichment on a proteolytic digest The data in Table 3-2 identified the diazo ketone polystyrene resin 2-16 as one of the optimal enrichment solid support. This material was therefore evaluated for phosphopeptide enrichment from a proteolytic digest of ovalbumin. Ovalbumin protein was digested with trypsin and subjected to methanolic hydrochloride to form its corresponding methyl esters. An aliquot of the digest (40 pmol) was subjected to the enrichment procedure using the 152 diazo ketone resin overnight in tetrahydrofuran. The enrichment sample was subsequently filtered, cleaved off the solid support, desalted and an aliquot of the sample (4 pmol) was analyzed by MALDI-ion trap MS and compared with the original digest to highlight the phosphopeptide purification using the diazo resin (Figure 3-2). One of the two phosphopeptides of ovalbumin, containing residues 341- 360 (EWGpSAEAGVDAASVSEEFR), was the predominant peptide enriched by the diazo ketone resin, confirming the ability of the enrichment to be performed on a more complex mixture. There is also the loss of -98 (H3PO4) corresponding to ion 2075.6, indicating the loss of a phosphate moiety, most likely a result of post-source decay. The second phosphopeptide corresponding to residues 63-85 (LPGFGDpSlEAQCGTSVNVHSSLR) was also enriched. The only ion present in the enrichment sample that was not due to a phosphopeptide was ion at m/z 1816, which corresponds to residues 324-340 (lSQAVHAAHAEINEAGR), an abundant ion present in the digest. 153 Fig 1 o 41(Fuf —\lullf- (I airmic—flqg 1 O aria-C—\a-lr- «I a!.u.-C~n«& Fig f0 rn Figure 3-2. fl 0 O l phosphopepfide A I * I phosphopeptide * Relative Abundance ° 0 1200 1600 2000 2400 2800 3200 3600 4000 2173.7 100- * o B O o 2 E- E 2075.6 :3 'H3P04 .9 4 1816.7 * 0' 2553.9 E ‘U a: 1200 1600 2000 2400 2800 3200 3600 4000 m-‘z Figure 3-2. Enrichment of ovalbumin. Depicted in Figure 3-2 is A) MALDI mass spectrum of 1 picomole of the formed methyl ester tryptic digest of ovalbumin. (B) MALDI mass spectrum of 154 4 pl corre the it (reslc and - tbnm was mefil hope HIPC 3.39, 2-16 8000 pmol MALl 4 picomole enriched ovalbumin tryptic digest. The ion at mlz 2173.7 corresponds to methyl ester form of phosphopeptide (residues 341-360) and the ion at mlz 2553.9 corresponds to the methyl ester form of phosphopeptide (residues 63-85). The ions at mlz 2093.7 and 2075.6 corresponds to the -80 and -98 loss of HPO3 and H3PO4 from the 2173.7 ion, respectively. To further confirm the identification of the two phosphorylated peptides, MSIMS analysis was performed for each ion. The MSIMS of 2173 ion showed a loss of methanol (2140.5 ion) and the loss of 98, corresponding to H3PO4 (see Appendix, Figure 3-39). The MS/MS of the ion at 2553.9 also indicated a H3PO4 loss of 98 (2455.6 ion) which corresponds to (see Appendix, Figures 3-39, 3-40). Additionally, phosphopeptide enrichment using the diazo ketone resin 2-16 was performed on a B-casein tryptic digest (40 pmol). The sample enrichment protocol was performed as described above and an aliquot (4 pmol) of the desalted enrichment sample was subsequently analyzed by MALDI-ion trap MS (Figure 3-3). The enrichment results primarily consisted of the monophosphorylated peptide containing residues 48-63 (FQpSEEQQQTEDELQDK) and the tetraphosphorylated peptide containg residues 16-40 (RELEELNVPGElVEpSLpSpSpSEESlTR). The ion corresponding to the multiply phosphorylated peptide is smaller, however, and this could be due to either ion suppression or a sub-stoichiometric amount of tetraphosphorylated peptide in the digest.20 The additional ion at m/z at 2244.7, is salt adduct of a 155 Sf‘ me 4 l the 32: (re: of t HIF illus Gm 302. peptide containing residues 199-217 (DMPIQAFLLYQEPVLGPVR), which was the only ion that did not correspond to a phosphopeptide. The remaining small ions present all corresponded to either de-methylation or salt adducts of the phosphorylated peptide ions. Illustrated in Figure 3-3 is A) MALDI mass spectrum of 1 picomole of methyl ester converted tryptic digest of B-casein. (B) MALDI mass spectrum of 4 picomole enriched B-casein digest. The ion at mlz 2160.6 corresponds to the methyl ester form of phosphopeptide (residues 48-63) and the ion at mlz 3234.8 corresponds to the methyl ester form of tetraphosphorylated peptide (residues 16-40). Each of these phosphorylated peptides were also analyzed by MALDI-ion trap MSIMS (see Appendix, Figures 340, 3-41). The MSIMS of the 2160.6 ion contained a dominant -98 Da loss, again corresponding to H3PO4. The MS/MS of the multiply phosphorylated peptide ion 3234 illustrates a dominant ion at 3122, corresponding to both the loss of a methyl group and H3PO4. There is also another loss of H3PO4 from the 3122 ion at 3024, confirming that it is a multiply phosphorylated peptide. 156 Flgl Figure 3-3. 100‘ A phosphopepfides ’1‘ K i 1200 1600 2000 2400 2800 3200 36m 4000 Relative Abundance ° 0 2150.5 100 - 3234.8 Relative Abundance °o 1200 1600 2000 2400 2800 3200 3500 4000 Ill-"Z Figure 3-3. Enrichment of B-casein phosphopeptides. 157 Dfl {F I Dr: 30 It is important to make note of the different type of phosphorylated peptides enriched in the two different phosphoprotein digests. The two phosphopeptides enriched in the ovalbumin sample contained more non-polar residues than the two phosphopeptides enriched from the B-casein digest. Additionally, this enrichment approach successfully enriched both monophosphorylated peptides and multiply phosphorylated peptides as well. While all of the phosphopeptides present in each of the digests were phosphoserines, it was hypothesized that this enrichment approach would be compatible with each of the different types of phosphorylated residues. 3.4.b Application of diazo ketone enrichment on various phosphorylated residues To confirm that the diazo enrichment technique was compatible with all the different phosphoresidues, two synthetic phosphopeptides, one containing a phosphotyrosine (DRVleHPF) and the other a phosphothreonine residue (LFTGHPEpTLEK), were each independently spiked into an unphosphorylated protein digest of bovine serum albumin, esterase and phosphorylase b (Figures 3-4, 3-5). The mixtures were then subjected to the enrichment procedure using diazo ketone resin 2-16. Each phosphopeptide was successfully isolated from the mixture, illustrating the ability of the diazo ketone resin to indiscriminately enrich each of the different phosphorylated residues. 158 Figure 3-4. DRVpYIHPF (Angioleusinm I 1154.3 8 Relativealbuudame'o ... 1000 1500 2000 2500 3000 3500 4000 111-12 1154.2 Relativealblmdmce'o 8 ‘ ‘.A_-_J I llllllilillllll‘lllI‘Ilf1llTlfil 1000 1500 2000 2500 3000 3500 4000 11152 Figure 3-4. Enrichment of phosphtyrosine peptide angiotensin II. 159 The spectra shown in Figure 3-4 illustrate (A) Spectra of angiotensin II (1 picomole) spiked into a three non-phosphorylated protein digest (0.1 pg of each). 8) Spectra of an aliquot (25 picomole) of the enrichment of angioteinsin lI (DRVpYIHPF). 100 picomole of angiotensin II was spiked into a digest containing 10 pg each of three unphosphorylated proteins and exposed to the diazo ketone 2-16 solid support for enrichment. This indicates the success of the solid-phase technique for phosphotyrosine peptides. 160 Figure 3-5. LFTGHPEIDTIFK 100- ° ‘ O .3. 5 1393.9 2 * =3. 3’ .2: ‘5 '3 at 1200 1600 2000 2400 2800 3200 3600 4&30 m-‘z B 1393.9 100 * O 0 § 0: E .5 "I 1” .fi ‘5 '8 m 1200 1600 2000 2400 2800 3200 3600 4000 Ill-‘2 Figure 3-5. Enrichment of a synthetic phosphothreonine peptide. The spectral data shown in Figure 3-5. illustrates: A) Spectra of LFTGHPEpTLEK. (1 picomole) spiked into a three non-phosphorylated protein digest (0.1 pg of each). B) Spectra of and aliquot (15 picomole) of the 161 enrichment of synthetic peptide, LFTGHPEpTLEK. 75 picomole of the synthetic peptide was spiked into a digest containing 7.5 pg each of three unphosphorylated proteins and exposed to the diazo ketone 16 solid support for enrichment. This indicates the success of the solid-phase technique for phosphothreonine peptides. 3.5 Experimental procedures Materials. Sequencing grade trypsin was purchased from Promega. 2,5 dihydroxybenzoic acid was provided from Laser Biolabs. C18 desalting ZipTips were provided by Millipore. The NuTip Ti02 and Zl‘Oz were purchased from Glygen Co. Bovine B-casein, ovalbumin, and all organic chemical reagents were obtained from Sigma Chemical Co. Polystyrene resins were purchased from either Novabiochem, Sigma Chemical Co., or Advanced Chemtech. Carboxylic acid silica was purchased from Silicycle. The H5 and D5 synthetic peptides were generously provided by the Reid lab. Two functionalized silica materials were generously provided by Prof. T. Pinnavaia (Michigan State University). Preparation of ovalbumin digests. Ovalbumin (100 pg) was dissolved in 6 M urea-50 mM Tris-HCI (20 pL). A 10 mM solution of dithiothreitol (5 pL) was added to the protein solution and heated at 65°C for 1 hr. The solution was cooled to room temperature and 50 mM ammonium bicarbonate (160 pL) and 100 mM iodoacetamide (10 pL) was added then incubated at room temperature in the absence of light for 1 hr. 20 pg of trypsin was dissolved in 50 mM acetic 162 acid (40 pL) and an aliquot of the trypsin solution (10 pL) was added to the protein solution then incubated for 16 hr. at 37°C. The reaction was quenched with glacial acetic acid (11 pL). The digest was dried down under nitrogen and subsequently stored at -80°C until their use. Preparation of unphosphorylated protein digests. Bovine serum albumin (100 pg), esterase (100 pg), and phosphorylase b (100 pg), were dissolved in 6 M urea-50 mM Tris-HCI (60 pL). A 10 mM solution of dithiothreitol (15 pL) was added to the protein solution and heated at 65°C for 1 hr. The solution was cooled to room temperature and 50 mM ammonium bicarbonate (480 pL) and 100 mM iodoacetamide (30 pL) was added then incubated at room temperature in the absence of light for 1 hr. 20 pg of trypsin was dissolved in 50 mM acetic acid (40 pL) and an aliquot of the trypsin solution (30 pL) was added to the protein solution then incubated for 16 hr. at 37°C. The reaction was quenched with glacial acetic acid (33 pL). The digest was dried down under nitrogen and subsequently stored at -80°C until their use. Preparation of B-casein digests. B-casein (10 nmol) was dissolved in 1 M ammonium bicarbonate (10 pL) and water (14 pL) and incubated for 5 minutes at 37°C. The solution was sonicated for 1 minute. 20 pg of trypsin was dissolved in 50 mM acetic acid (40 pL) and an aliquot of the trypsin solution (10 pL) was added to the protein solution then incubated for 16 hr. at 37°C. The digests were 163 divided into 10 pg aliquots, lyophilized under a steady stream of nitrogen gas and stored at -80°C until their use. Preparation of methyl ester converted digests. Methanolic hydrochloric acid was prepared by the addition of acetyl chloride (320 pL) to anhydrous methanol (2 mL). An aliquot (200 pL) of this solution was then added to 10 pg dried digests and incubated at room temperature for 2 hr. The methyl ester converted digest was then dried down under nitrogen, dissolved in methanol and stored in 100 pmol aliquots at -20°C. For the unphosphorylated protein digest, the 100 pg digest was incubated in an aliquot of methanolic hydrochloric acid (500 pL) for 3 hr. and then dried down under nitrogen and stored at -20°C. Enrichment of phosphorylated proteins using diazo-functionalized resins. Briefly, the diazo functionalized material (20 mg) and tetrahydrofuran (1 mL) were added to an aliquot of either a methyl ester converted protein digest (40 pmol) or a methyl ester converted purified phosphopeptide (70-100 pmol) spiked into a unphosphorylated protein digest (7.5-10 pg of each protein). The mixture was rocked gently overnight at room temperature. The resin was recovered and washed with tetrahydrofuran (5x 500 pL), acetonitrile (4x 500 pL) and then methanol (1x 500 pL). Cleavage of the captured species from the resin was completed by incubation in a solution 50% trifluoroacetic acid, 40% DCM, 10% water (500 pL). The mixture was rocked gently overnight at room temperature, allowing for full interaction between the resin and the cleavage solution. The 164 resin was filtered and the elution fraction was collected. The resin was washed once with methanol (500 pL) and added to the original fraction. The combined eluents were concentrated under nitrogen. Prior to analysis, the samples were diluted in water (20 pL) and desalted using Millipore C18 Zip Tips following the manufacturer’s protocol. Briefly, the zip tip was moistened with acetonitrile (3x10 pL), then equilibrated with 0.1% trifluoroacetic acid (3x10 pL) and introduced to the enrichment sample (18x10 pL). The bound peptides were liberated from the zip tip with 50:50 0.1% trifluoroacetic acidzacetonitrile (5 pL). All desalted samples were subsequently stored at -20°C. Quantification of enrichment for diazo-functionalized resins. The enrichment procedure was performed as described above in duplicate, with each diazo solid support using the D5 labeled synthetic peptide (10 pmol) and a non- phosphorylated peptide mixture (20 pmol of 3 peptides). The samples were dissolved in 0.1% trifluoroacetic acid (20 pL) and subsequently desalted using Millipore C18 ZipTips. Briefly, the zip tip was moistened with acetonitrile (3x10 pL), then equilibrated with 0.1% trifluoroacetic acid (3x10 pL) and introduced to the enrichment sample (18x10 pL). The bound peptides were liberated from the zip tip with 50:50 0.1% trifluoroacetic acidzacetonitrile (5 pL). An aliquot of the desalted enrichment product was spotted on the MALDI plate (0.5 pL) and the H5 peptide (1 pmol) was spotted directly on top of the enrichment sample to determine enrichment recovery. 165 Phosphorylation assay of CHKtide with P32 ATP. A 2mM aqueous solution of CHKtide (2.5pL) was added to a 50mM MOPS, pH 7.0, 2.5 mM EDTA solution (5pL). To the CHKtide solution, an aliquot of 25 nglpL solution of CHK-2 in 20mM MOPS, pH 7.5, 1 mL EDTA, 0.03% Tritton-X-100, 5% glycerol, 0.1 % B- mercaptoethanol, 1 mg/mL bovine serum albumin (2.5pL) and sterile water (5pL) was added. A solution (90pL) of 37.5mM magnesium chloride and 250pM ATP in 20mM MOPS, pH 7.2, 25mM B-glycerol phosphate, 5mM EGTA, 1mM sodium orthovanadate, 1mM dithiothreitol was added to 10pCi/pL solution of SOOOCi/mmol [y-32P]ATP (mph. The 9:1 ATP-[y-32P1ATP (10pL) was added to the assay and incubated at 30°C for 10 minutes. 20pL of the assay was spotted on a 2 cm circle of P81 ion exchange chromatography paper, allowed to dry and washed three times for five minutes in 0.75% phosphoric acid (40mL) and then once in acetone (40mL) for five minutes. The peptide was removed from the paper by adding 10% ammonium hydroxide (1.5mL). The filtered peptide was dried down under nitrogen. Methanolic hydrochloric acid was prepared by adding acetyl chloride (320pL) to anhydrous methanol (2mL) and subsequently added (100pL) to the phosphorylated CHKtide and incubated at room temperature for 2hr. The methyl esterified, phosphorylated peptide stock was made into aliquots and dried under nitrogen. Enrichment of radiolabeled CHKtide using diazo ketone functionalized material. To a dried methyl esterified radiolabeled CHKtide aliquot (10pL), N,N- dimethylfomtamide or tetrahydrofuran, (1mL) was added along with the diazo 166 ketone-functionalized solid-phase material (~10 mg). The mixture was shaken overnight at 25°C. The material was washed with N,N-dimethylformamide (2.5mL), acetonitrile (2.5mL) and methanol (2.5mL) and each washing was removed. 90% trifluoroacetic acid (500pL) or 10% trifluoroacetic acid in dichloromethane (500pL) was added and the resin and shaken for 6.0 hr. The resin was filtered off and the eluent was collected and dried under nitrogen. Dried enrichment sample was diluted in water (50pL) and an aliquot (10pL) was added to a solution of HiSafe 3 scintillation cocktail (800pL) and water (190pL). Radioactivity was measured by a Wallac 1414 Spectral Scintillation Counter. Quantification of enrichment for diazo-functionalized resins. The enrichment procedure was performed as described above using the D5 labeled synthetic peptide (10 pmol). The enrichment samples were pulled up in 0.1% trifluoroacetic acid (20 pL) and subsequently desalted using Millipore C18 ZipTips. Briefly, the zip tip was moistened with acetonitrile (3x10 pL), then equilibrated with 0.1% trifluoroacetic acid (3x10 pL) and introduced to the enrichment sample (18x10 pL). The bound peptides were liberated from the zip tip with 50:50 0.1% trifluoroacetic acidzacetonitrile (5 pL). An aliquot of the desalted enrichment product was spotted on the MALDI plate (0.5 pL) and the H5 peptide (1 pmol) was spotted directly on top of the enrichment sample to determine enrichment recovery. 167 w gppfifl lineal quad“ Laser 0‘3: 156% Mass Spectrometry. MS and CID-MSIMS analyses were performed using a linear quadrupole ion trap mass spectrometer equipped with a Matrix Assisted Laser Desorption Ionization (MALDI) source (model vMALDl-LTQ, Thermo, San Jose, CA). Desalted enrichment samples were spotted on the MALDI plate (0.5 pL) and then an aliquot of 50 mglmL DHB matrix in a solution of 50:50 acetonitrile:0.1 % trifluoroacetic acid (0.5 pL). 168 Figure 34 Appendix Figure 3-6. 1155.2 100— 8 l Relaivelntenslty l ’9. ii; ; f 'I . . Wt. 2'. 7' ' Aw‘ 4‘“ L“ 4 ALL... __ - A A ‘_ 0‘1" ’il * 'I‘ ‘l l l 500.0 1400.2 2303.4 3200.8 4100 8 51010 Figure 3-6. MALDI-TOF MS of the enrichment of 25 pmol of angiotensin II, using 90% TFA for the cleavage conditions. (methyl ester converted angiotensin II peptide mlz = 1155). 169 Figure 3-7. 1127.1 100— a U) E I g 50 _ i .2 I E f 0 f a: 1 I "1,111 .13 “a”: " 2...... 0‘l l l ’ l l ' ‘ I 500.0 1400.2 2300.4 3200.3 41005 51010 mlz Figure 3-7. MALDI-TOF MS of the enrichment of 25 pmol of angiotensin II, using 30% NH4OH for the cleavage conditions. Under basic conditions, the methyl esters are hydrolyzed, resulting in the recovery of the unmodified peptide (mlz 1127). 170 Figure 3-8. 100- 2551.3 4257.3 I I I l ! Rolalveinlensiy 8 l l l l l l | 490.0 1300.4 2209.8 3200.2 41 (X16 5101.0 Figure 3-8. MALDI-TOF MS of the enrichment of 100 pmol of methyl ester converted B-casein non-sequencing grade trypsin digest. 171 Figure 39. 2145.4 100— Relatlve in ten slty 8 | ..._ {J «D 5" nun-n..— It 1 l I III. . It If} ‘ fiT, . M. ’ 1‘1 1 An” , KM 3,13 _ . 3 l‘ L 499.0 1399.4 22%.8 3200.2 4100 6 5101.0 Figure 3-9. MALDI-TOF MS of the enrichment of 100 pmol of methyl ester converted B-casein non-sequencing grade trypsin digest, under 90% TFA cleavage conditions. 172 Figure 3-10. 2058.1 100— 3 1980.3 1 \" _a» U c . 2 '3 5° --; .2 ' 33 O m 1 3140.7 41m.0 5101.0 Figure 3-10. MALDI-TOF MS of the enrichment of 250 pmol of methyl ester converted B-casein non-sequencing grade trypsin digest, under 30% NH4OH cleavage conditions. 173 Figure 3-11. Relativeintensity _I- o 8 8 g 1111111121 0 A O C.) O A ‘o” O to D 8 3 M 01 8 100 NI SEE 3'18- w o: o o C) _n 0 9° 2 o: ‘9’ 9,, 'de a, NJ 0 h o Q 0 Figure 3-11. MALDI-ion trap MS of CHKtide standard 174 Figure 3-12. Relative intensity 009 09 *-00l WI» 8‘ 0 § 0 3 38‘ g? "I E g 8:3 3. 0099 #917999 Figure 3-12. MALDI-ion trap MS of the CHKtide phosphorylation assay 175 Figure 3-13. Relative inten sity 9 DOI- 008 l -, r r a, PQ'POSL 0003 9118M 91.1.261- M 45 8 88 :30) 23.0- o-3 - oo 31 '2. $23 3 .. o .l-o No (I) s» M ~d Figure 3-13. The MALDI-ion trap MSIMS spectra of the CHKtide phosphorylated ion at mlz 2781. 176 Figure 3-14. 1-05 05le Calibration Curve 0.95 0.85 0.75 0.55 0.55 0.45 0.35 0.25 0.15 0.05 "0‘05 0 0.2 0.4 0.5 0.8 1 Amount of D5 peptide (pmol) y 1' 1.062x - 0.027 R2 = 0.987 I... . . r... . .... . .-... _ . T" .. ...-.. I Relative intensity ratio of 05 to H5 pepflde Figure 3-14. H5lD5 MS calibration curve. Various amounts of the synthetic labeled D5 peptide (50, 100, 250, 500, 750, 1000 fmol) were spotted with 1000 fmol of H5 peptide and the calculated ratio between the two peptides were plotted against the actual relative abundance in the spectra. The standard curve was then used to determine the amount of D5 present within the enrichment sample by comparing the relative abundance of H5lD5 in the sample. 177 Figure 3-15. H5 100_ 14335.5 .3 '- m —l g _ s 50... 5 - a '_' 05 o: _ 1440.5 0 I l l l I 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 100— n '5 - l 2 .— s 50— g " U 05 or: - 1440.5 0— 1425 1430 1435 1440 1445 1450 mlz Figure 3-15. MALDI-ion trap MS of D5 enrichment using diazo acetate Wang resin (Resin 2-1). 178 Figure 3-16. H5 1435.5 100— n g _ E 50-‘ 3 - § _ g - 05 .. 1440.5 0_ I 1 r1 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 .g' __ n g _ g - a " U 05 ‘r 1 1440.5 0_"1'9'¢F7*l| f1? IT! II 1425 1430 1435 1440 1445 1450 ml? Figure 3-16. MALDI-ion trap MS of D5 enrichment using diazo acetate Wang resin (Resin 2-2). 179 Figure 3-17. H5 1435.5 .4? ‘ m I: g _ E 50— '5 : 05 c: _ 1440.5 O—d 5.. A A - A. ‘AMAM 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 Relative intensity 1425 1430 1435 1440 1445 1450 Figure 3-17. MALDI-ion trap MS of D5 enrichment using diazo amide tentagel resin (Resin 2-3) 180 Figure 3-18. H5 1435.5 100-— “ .3‘ - a _ ‘5 - .5 50— % - '6 : 05 n: g 1 1440.5 0"_T‘T‘T_I ll 7 I II I I I 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 100-m 4 1 Relative inten sity 0| 0 L . _1 D5 4 1440.5 o“'fi‘§‘*‘7_hII I I I] I I I 1425 1430 1435 1440 1445 1450 mlz Figure 3-18. MALDI-ion trap MS of D5 enrichment using diazo amide megabead resin (Resin 2-4). 181 Figure 3-19. H5 1435.5 2' .. .5 .1 § - .s 50— ¢ .> ‘4 8 _ g .. 05 .. 1440.5 0 II I I I 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 100-— Relative inten sity OI O I 1425 1430 1435 1440 1445 1450 mi: Figure 3-19. MALDI-ion trap MS of D5 enrichment using diazo amide PEGA resin (Resin 2-5) 182 Figure 3-20. H5 1435.5 4 8 IILI :3- Relative intensity 01 O I Z 05 i _ 1440.5 2 0 _..AAA . _.. - -, _ , - - ‘L 1425 1430 1435 1440 1445 1450 mlz i H5 100‘ 14F55 z~ _ .5 _ g .. g 50.. 15 .. 0: 1 0 l l 1425 1430 1435 1440 1445 1450 mlz Figure 3-20. MALDI-ion trap MS of D5 enrichment using diazo acetate PEGA HMPA resin (Resin 2-6). 183 Figure 3-21. H5 1435.5 .2‘ ‘ a; .. C 2 s 50.. O -I § _ D5 3 1440.5 m -l 0 I I I r I T 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 100% n 5 : § 4 5 50— ° - 5 _ 05 g _ 1440.5 1 0 AAA - - _ L -‘A A .. _ A 1425 1430 1435 1440 1445 1450 mlz Figure 3-21. MALDI-ion trap MS of DS enrichment using diazo amide dendrimer resin (Resin 2-7) 184 Figure 3-22. H5 1435.5 .3' - a - 1 g .. .E 50— .9 H 5 - U ° 5 05 [I _ 1440.5 0%.? II? I'T" I FTfi 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 100— 2? -. '6 d g .. E 50— % 4 g d D5 .. 1440.5 0— 1425 1430 1435 1440 1445 1450 mlz Figure 3-22. MALDI-ion trap MS of D5 enrichment using 4-diazo acetate polystyrene (Resin 2-8) 185 Figure 3-23. H5 1435.5 100-1 .. 1 .2- ‘ 65 .. g _, .3 50-1 e> - g _ .. 05 a: ‘ 1440.5 0— II I I I FI 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 100— " 2‘ .. '55 .. c s _- .E 50... g " 05 0: ‘ 1440.5 0.... 1425 1430 1435 1440 1445 1450 mlz Figure 3-23. MALDI-ion trap MS of D5 enrichment using diazo acetate polystyrene (Resin 2-9). 186 Figure 3-24. H5 1435.5 100— . 1 .03.; g n g .4 s 50— ° - ‘5. - u 3 § - 05 3+ _ 1440.5 _ o 1425 1430 1435 1440 1445 1450 3 mlz H5 1435.5 100— 1 .g' _ H g .. '5 - U 6? H 05 _ 1440.5 0 I‘I‘I I [Fl I 1425 1430 1435 1440 1445 1450 mlz Figure 3-24. MALDI-ion trap MS of D5 enrichment using NovaSyn diazo acetate resin (Resin 2-10). 187 Figure 3-25. H5 1435.5 100! 4 1 .33 _ 3 '1 s - E 50— 3 - E .. o m -1 0 .1 {+1 I I I I? I I I I 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 100— ‘ 1 Relative intensity at O I .1 D5 1 1440.5 0—‘f r T I 1 Fl 1 'f‘ I I I I 1425 1430 1435 1440 1445 1450 mlz Figure 3-25. MALDI-ion trap MS of D5 enrichment using NovaSyn TG HMP diazo acetate resin (Resin 2-11). 188 Figure 3-26. 100% l 50— 0— 1425 é [III I 1 Relative intensity 0| 0 I O 1L1 1425 H5 1435.5 U 1 D5 1440.5 I l I I 1430 1435 1440 1445 1450 mlz H5 1435.5 U D5 1440.5 II I I I 1430 1435 1440 1445 1450 Figure 3-26. MALDI-ion trap MS of D5 enrichment using NovaSyn diazo anhydride resin (Resin 2-12). 189 Figure 3-27. H5 1435.5 100— a .g' _ é _ g . 05 C! ‘ 1449-5- 0 -.‘AAA - -- -, A _A,A _ - 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 100— n 2' _ .5 .. g _ .5 50... '5 4 _ - D5 5'? 1 1440.5 0... 1425 1430 1435 1440 1445 1450 mlz Figure 3-27. MALDI-ion trap MS of D5 enrichment using vicinal diazo acetate silica (Resin 2-13). 190 1' .{I- .1993 q Figure 3-28. H5 1435.5 1007 11 g .. E ‘50—; h g - s: E! "' ; ' 0 1425 1430 1435 1440 1445 1450 11 mlz 9 H5 1435.5 100- n g I .E 50— % - a " 05 0‘ " 1440.5 fi 0 A‘A-. ,- .. _A‘AA“- 1425 1430 1435 1440 1445 1450 mlz Figure 3-28. MALDI-ion trap MS of DS enrichment using diazo amide silica (Resin 2-14). 191 Figure 3-29. H5 1435.5 100— 1 .3' " £3 .. é _. "33 so: '5 - a) _ 05 a: _ 1440.5 0— [I I I I [f 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 100— n .E' ' a, — g _ 5 50— 3 g _ D5 Ct: _ . 1440.5 0; AA -. . .- __A.A - - 1425 1430 1435 1440 1445 1450 mlz Figure 3-29. MALDI-ion trap MS of D5 enrichment using diazo ketone resin (Resin 2-15). 192 Figure 3-30. H5 1435.5 100— 1 .5" ' a; .. § _ .s 50_ 3 " r g 05 i 0: 1440.5 0— 1425 1430 1435 1440 1445 1450 4: mlz ; H5 100- 147155 g _ 1 C 2 .— .s 50.. o _ D5 5 _ U 1440.5 6 m — 0 I] l T l l T 1425 1430 1435 1440 1445 1450 mlz Figure 3-30. MALDI-ion trap MS of D5 enrichment using diazo ketone resin (Resin 2-16). 193 Figure 3-31. H5 1435.5 100— 1 ‘3‘ a g _ E 50— 2:. 5 05 0: 1440.5 3 o l I I F F 1425 1430 1435 1440 1445 1450 "V2 1 H5 1435.5 100—- n {-3 I n g _ c '3 ,> 5 0 a: 1425 1430 1435 1440 1445 1450 mlz Figure 3-31. MALDI-ion trap MS of D5 enrichment using diazo ketone silica (Resin 2- 17). 194 Figure 3-32. H5 1435.5 100— h .2." as 5 s 50- .> - § - g - D5 - 1440.5 0— 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 100— n 4%“ I n g _ .s O ,> 5 O a: 1425 1430 1435 1440 1445 1450 mlz Figure 3-32. MALDI-ion trap MS of DS enrichment using StratoSpheres phenyl diazo methane (Resin 2-18). 195 Figure 3-33. H5 1435.5 100— n g .. s 50— .g - 3 : 05 _ 1440.5 0— T l I I 1425 1430 1435 1440 1445 1450 ml! H5 1435.5 100— n .E‘ '- 0) .— g .. .5 50— .> "' 5 _ g - D5 .. 1440.5 °‘7 1425 1430 1435 1440 1445 1450 mlz Figure 3-33. MALDI-ion trap MS of D5 enrichment using phenyl diazo methane polystyrene (Resin 2-19). 196 Figure 3-34. H5 1435.5 100— n l l Relative inten sity 0'! C) l °1 1425 1430 1435 1440 1445 1450 miz H5 100__ 1415.5 Relative intensity 0! O l 1425 1430 Figure 3-34. MALDI-ion trap MS of DS enrichment using phenyl diazo methane polystyrene (Resin 2-20). 197 Figure 3-35. H5 1435.5 ‘3‘ a; _ c 2 — E 50.. o .> - 8 _ g - D5 _ J 1440.5 oTfilrlIII711Irl 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 100—- n é" I n c: 2 — S 50..— -§ - l E - D5 .. 1440.5 o—IIIIII113'ITIvafii—‘F—rfi 1425 1430 1435 1440 1445 1450 mlz Figure 3-35. MALDI-ion trap MS of D5 enrichment using Millipore IMAC Ziptips. 198 Figure 3-36. H5 100— 14:65.5 2‘ m — § _ s 50.. g - ‘6 ' . 05 0‘ : 1440.5 0——,%¢; [111—r leI l 1425 1430 1435 1440 1445 1450 mlz H5 1435.5 100— R g _ ” g _ .5 50— '> - g - l 05 0: " J 1440.5 °—-flir°: [Ill—7 IIrTl Ill 1425 1430 1435 1440 1445 1450 mi: Figure 3-36. MALDI-ion trap MS of DS enrichment using Glygen TlOz NuTips. 199 Figure 3-37. H5 1435.50 100— n E a) .- C s - .5 50— .> 5 ‘ 05 O a: : 1440.5 0"" I I I I 'I I 1425 1430 1435 1440 1445 1450 mfz H5 1435.50 100—' n .23 - m - c 8 - .s so— '3 - Tn : 05 0‘ _ 1440.5 0— I I I I II I 1425 1430 1435 1440 1445 1450 mlz Figure 3-37. MALDI-ion trap MS of 05 enrichment using Glygen ZrOz NuTips. 200 Figure 3-38. Relativeinlensitv _II OI O O O O on O 11L L Li 1 l [J O ---I A A- O P o— —l' A— g__ O_"_as M LC!) “‘0! ”'4 “’Q P " a: A b 8- w 0} M O O O a; M o 2 0 NM. fic’I Q 9‘0 ~- 89 3% a 53° C) 011 O Figure 3-38. MALDI-ion trap CID MSIMS spectra of ion at m/z 2173 from the ovalbumin enrichment. 201 Figure 3-39. Relative tnten slty O as —ml DOLL WI nu mu m \ Him 2995?! 659862 372793 ’oa‘- l- essesz C‘H— 3 Figure 3-39. MALDI-ion trap CID MSIMS spectra of ion at mlz 2553 from the ovalbumin enrichment. 202 Figure 340. Relativeintensity 0 d o o 8 8 I I I I I I I I ’ o _L D 0* o “—I 33_ o o‘— ’—h _“Nl P33 _. ,‘cn 8—-'°" A 0‘”? 113m —01§ _ ' I *— 3 3...: ..._' NI 3'0 '- 31 3%.? _——-~ M NO 34 an o 0‘ Figure 3-40. MALDI-ion trap CID MS/MS spectra of ion at mlz 2160 from the B- casein enrichment. 203 -ZoOI .159 80 II 33.8 -2601 .150“ .. -quo. I 88.8 I www.— .mw was: .mn mocha Relative inten sity 01 O - ‘iIII )I'Illln . ..',iIP It-" Pl . I I'I.l.l|l‘lltlll’lri'»l ,I II. 'E'IIIII.III|'I’IIU . . li'ln II. III? 08 Ammo m 30 ~30 ago 580 :5 Figure 3-41. Figure 3-41. MALDI-ion trap CID MSIMS spectra of ion at m/z 3234 from the B- casein enrichment. 204 References Hage, D. S. Survey of recent advances in analytical applications of immunoaffinity chromatography. Journal of Chromatography B-Analytical Technologies in the Biomedical and Life Sciences 1998, 715, (1 ), 3-28. Stevenson, D. lmmuno-affinity solid-phase extraction. Journal of Chromatography B-Analytica/ Technologies in the Biomedical and Life Sciences 2000, 745, (1 ), 39-48. Vocadlo, D. J.; Hang, H. C.; Kim, E. J.; Hanover, J. A.; Bertozzi, C. R. A chemical approach for identifying o-Glcnac-modified proteins in cells. Proceedings of the National Academy of Sciences of the United States of America 2003, 100, (16), 9116-9121. Baskin, J. M.; Bertozzi, C. R. Bioorthogonal click chemistry: Covalent labeling in living systems. Qsar & Combinatorial Science 2007, 26, (11- 12),1211-1219. Fonovic, M.; Verhelst, S. H. L.; Scrum, M. T.; Bogyo, M. Proteomics evaluation of chemically cleavable activity-based probes. Molecular and Cellular Proteomics 2007, 6, (1 O), 1761 -1 770. Camarero, J. A.; Kwon, Y. G. Traceless and site-specific attachment of proteins onto solid supports. lntemational Journal of Peptide Research and Therapeutics 2008, 14, (4), 351-357. Ficarro, S. B.; McCleland, M. L.; Stukenberg, P. T.; Burke, D. J.; Ross, M. M.; Shabanowitz, J.; Hunt, D. F.; White, F. M. Phosphoproteome analysis by mass spectrometry and its application to saccharomyces cerevisiae. Nature Biotechnology 2002, 20, (3), 301-305. Lansdell, T. A.; Tepe, J. J. Isolation of phosphopeptides using solid phase enrichment. Tetrahedron Letters 2004, 45, (1 ), 91-93. Goshe, M. B.; Conrads, T. P.; Panisko, E. A.; Angell, N. H.; Veenstra, T. D.; Smith, R. D. Phosphoprotein isotope-coded affinity tag approach for isolating and quantitating phosphopeptides in proteome-wide analyses. Analytical Chemistry 2001, 73, (11), 2578-2586. 205 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. Tao, W. A.; Wollscheid, B.; O'Brien, R.; Eng, J. K.; Li, X. J.; Bodenmiller, 8.; Watts, J. D.; Hood, L.; Aebersold, R. Quantitative phosphoproteome analysis using a dendn'mer conjugation chemistry and tandem mass spectrometry. Nature Methods 2005, 2, (8), 591-598. Furnari, B.; Rhind, N.; Russell, P. Cdc25 mitotic inducer targeted by Chk1 DNA damage checkpoint kinase. Science 1997, 277, 1495-1497. Olsen, J. V.; Blagoev, B.; Gnad, F.; Macek, B.; Kumar, C.; Mortensen, P.; Mann, M. Global, in vivo, and site-specific phosphorylation dynamics in signaling networks. Cell 2006, 127, (3), 635-648. Zhou, H. L.; Ranish, J. A.; Watts, J. D.; Aebersold, R. Quantitative proteome analysis by solid-phase isotope tagging and mass spectrometry. Nature Biotechnology 2002, 20, (5), 512-515. Ong, S. E.; Blagoev, 8.; Kratchmarova, l.; Kristensen, D. B.; Steen, H.; Pandey, A.; Mann, M. Stable isotope labeling by amino acids in cell culture, silac, as a simple and accurate approach to expression proteomics. Molecular & Cellular Proteomics 2002, 1, (5), 376-386. Zhu, H. N.; Pan, S. Q.; Gu, S.; Bradbury, E. M.; Chen, X. Amino acid residue specific stable isotope labeling for quantitative proteomics. Rapid Communications in Mass Spectrometry 2002, 16, (22), 2115-2123. Chen, X.; Smith, L. M.; Bradbury, E. M. Site-specific mass tagging with stable isotopes in proteins for accurate and efficient protein identification. Analytical Chemistry 2000, 72, (6), 1134-1143. Spellman, D. S.; Deinhardt, K.; Darie, C. C.; Chao, M. V.; Neubert, T. A. Stable isotopic labeling by amino acids in cultured primary neurons. Molecular & Cellular Proteomics 2008, 7, (6), 1067-1076. Dunn, J. D.; Igrisan, E. A.; Palumbo, A. M.; Reid, G. E.; Bruening, M. L. Phosphopeptide enrichment using MALDI plates modified with high- capacity polymer brushes. Analytical Chemistry 2008, 80, (15), 5727- 5735. Kweon, H. K.; Hakansson, K. Selective zirconium dioxide-based enrichment of phosphorylated peptides for mass spectrometric analysis. Analytical Chemistry 2006, 78, (6), 1743-1749. 206 20. Steen, H.; Jebanathirajah, J. A.; Rush, J.; Monica, N.; Kirschner, M. W. Phosphorylation analysis by mass spectrometry - myths, facts, and the consequences for qualitative and quantitative measurements. Molecular and Cellular Proteomics 2006, 5, (1 ), 172-181. 207 CHAPTER4 APPLICATION OF PHOSPHOPEPTIDE ENRICHMENT ON A BIOLOGICAL SAMPLE CONTAINING NF-KB PROTEIN-PRELIMINARY WORK AND FUTURE PROJECT DIRECTIONS 4.1 Introduction In the previous chapter, application of the enrichment approach was illustrated using commercially available proteins. Upon the successful enrichment of commercially available proteins, the next logical step is probing for phosphopeptides from biologically important samples. The application of the enrichment approach on a more complex matrix is important for a few reasons; (1) successful enrichment illustrates the capabilities of this approach and (2) enables the identification of potentially new phosphorylation sites. As mentioned in chapter one, phosphoproteome analysis of a dynamic system (i.e. cellular extracts) could identify relevant phosphorylation events linked to the pathology of a specific disease through protein (mis-)regulation. Our objective was to implement our enrichment strategy in an effort to probe the cancer phosphoproteome. Identification of these phosphorylation events could provide insight towards the identification of possible cancer biomarkers. The importance and impact that biomarker identification has on cancer treatment can be considered a three-faceted approach involving prognostic, predictive and diagnostic markers.1 Prognostic biomarkers indicate the severity of the type of tumor and the logical outcome for the patient. To date, prognostic-type biomarkers are probed through DNA or gene-expression signatures in the form of microarrays, rather than proteomic studies, so our 208 enrichment strategy is not applicable to the identification of prognostic biomarkers. The other two types of biomarkers, however, include proteomic analysis. As a result, both are avenues to explore the phosphoproteome and to identify biomarkers using our enrichment strategy. The first of the two is predictive biomarkers, which can indicate how a patient will respond to treatment or which type of treatment they should receive. Utility of these biomarkers includes the observation that breast cancer patients who have an up—regulated ERBBZ gene benefit better from the drug trastuzumab, where other patients with over-expressed estrogen receptor are given tamoxifen to treat the disease.1 These identified genomic over- expressions do have proteomic ramifications, which can lead to further insights into the potential of proteomic detection and treatment strategies. For example, it have been found that the lung tumor drug, gefitinib, can more effectively treat cancer when there are mutations in DNA for the kinase domain of the epidermal growth factor receptor (EGFR).2 Based on the cancer’s resulting phenotype caused by the genetic mutation, this study clearly has opportunities to identify more specific biomarkers by probing the phosphoproteome. Furthermore, characterization of the tumor phosphoproteome cannot just include the presence (or absence) of a protein but also requires analysis of changes in protein levels, which can also become a significant biomarker as well. Diagnostic biomarkers could lead to much earlier detection of specific types of cancer, which would result in earlier treatment and greater rate of 209 survival. Statistically, cancer patients who are diagnosed at an advanced stage of the disease have a five-year survival rate of less than 20%, depending on the type of cancer. This number changes to almost 90% for patients who are diagnosed with cancer at the early stage. In early detection patients, the ten year survival rate is 80%.3 Clearly, this illustrates both the importance in early detection and determining the vehicle to allow for early detection, namely disease markers. The concept of biomarkers originated from early studies which involved probing for tumor-suppressor genes and other genomic molecules to study the proteomic impact of the corresponding genomic profile. The limitation of genomics in the realm of diagnosis and prognosis has been shown in some cancer cell lines by the lack of correlation between mRNA abundance and protein levels.4 Studying specific oncogenic pathways and the regulation of those pathways through signal transduction, however, could provide valuable insight on the disease's specific molecular signature. Understanding the molecular impact of the disease and how it varies from healthy tissue would allow looking, not just at how to diagnose the patient at an early stage, but also at potential drug targets that would be specific for only the tumorgenic and not the healthy tissue. There are many pathways that have been researched for their role in the pathogenesis of cancer, but discussion of all relevant pathways exceeds the specified objective of this project, as I will discuss the relevant pathway l pursued for phosphopeptide enrichment. As mentioned before, the phosphoproteome in 210 cancer cells is of interest because of the impact that phosphorylation has on mis- regulation of signal transduction pathways within tumorgenic cells. Using our enrichment strategy will allow for the monitoring of how and where these pathways become activated (i.e. de/phosphorylated) or unactivated within the tumor provide valuable information to identify both potential biomarkers and drug targets. 4.2 Introduction to NF-KB The transcription factor protein, nuclear factor-KB (NF-KB), has been thoroughly studied since it was first discovered in 1986.5 It is now known to consist of a group of similar proteins RelA (p65), RelB, c-Rel, p50 and p52, all containing a Rel homology domain, which is responsible for both DNA-binding and homo/hetero dimerization. The NF-KB proteins are responsible for transcribing a variety of different genes involved in metastasis, cell proliferation, inflammation, cell survival and suppression of apoptosis (reviewed extensively in 6'7). This protein has been widely researched in an attempt to understand how this one transcription factor is involved with so many intracellular signals and such a wide variety of stimuli.2 It is believed that the ability of NF-KB to take part in the regulation of so many different processes is due to post-translational modifications, such as phosphorylation.3 Although there have been a few of the phosphorylation sites on the p65 subunit which have been determined, to date, it is hypothesized that all the protein’s phosphorylations have not fully been characterized.4 These modifications are of much interest to help understand how 211 this protein is involved in so many different pathways and in response to so many different stimuli. As previously mentioned, NF-KB is composed of homo— and heterodimers of Rel proteins, the most common being the p50/p65 heterodimer.5 Typically NF- KB exists in its inactive form in the cytoplasm bound to the inhibitory protein, IKB, until activated by outside stimuli. Activation of the NF-KB pathway can occur in a variety of different ways including the introduction of a chemotherapeutic agent such as camptothecin or a cytokine like TNF-q, etc. It is these activators that initiate the classical NF-KB pathway and are believed to be involved in both cancer pathology and inflammatory diseases. Activation of the classical NF-KB pathway is initiated within the cell, it mediates a series of phosphorylations on I- KB by IKK kinases. The phosphorylations target l-KB for ubiquitinylation where l- KB is subsequently degraded by the 26S proteasome. Upon the degradation of I- KB, NF-KB (p50, p65 subunits) is translocated into the nucleus (Figure 4-1).1'7 212 Cytokines (TNF-a) Extracellular Cytoplasm - Senne Chemotherapeutlcs T kinases IKK Kinases _ - ®® ‘P657 ,A» ‘ P65_ j.-- * "“3 P50 , ' ' "“3. P50, tr ubiquitinylation _ .@ (,9 P65 " _ proteasome . ' I I-KB ' .P65- degradation ~P50 ,. Nucleus NF-KB translocation Inhibition of apoptosis .ifp65' Gene 5‘ p50, transcription Figure 4-1. The classical NF-kB signaling pathway. 213 Apoptosis regulates the number of cells as well as limiting the number of mutated cells,8 however, this programmed cell death system is down-regulated in cancer cells, resulting in tumor cell growth and chemotherapeutic resistance.9 The role that the NF-KB pathway plays in apoptosis is of particular interest to provide some insight into regulating this process as a means of adjuvant therapy. 4.3 General signaling pathway of NF-KB The inactive form of NF-xB is sequestered in the cytoplasm by a complex of inhibitory-KB proteins (l-KB). Upon activation of the “classical” pathway, l-KB becomes phosphorylated by l-KB kinases (IKKs) and the resulting phosphorylations on l-KB target it for ubiquitinylation, followed by degradation by 8‘10). Upon the 26S proteasome (comprehensive NF-KB pathway reviews degradation of I-KB, the nuclear localization sequence of NF-KB is accessible and subsequently translocated into the nucleus, where it initiates the transcription of multiple genes. Depending on the origin of the activation, the pathway does vary on how and in what form NF-KB is translocated into the nucleus, which would be expected from the variety of genes NF-KB is responsible for transcribing. The known phosphorylation sites of each of the subunits of NF-KB are illustrated in Figure 4-2. The number of different phosphorylation sites and the corresponding kinases responsible for these events illustrate the dynamic response to stimuli by NF-KB. The exposure of different stimuli results in phosphorylation events at different sites of NF-KB and the variance of phosphorylation events invoke the transcriptions of different genes. An overview of the known phosphorylation sites 214 of each of the NF-KB subunits shown in Figure 4-2, and their corresponding kinase, if known, is also listed. [24] [25] [26] PKAc GSKBB lKKb I / \ / \ $337 $903S907 89278932 Cl? p105 (p50) [Rel Homology Domain Ankyrin repeats Death Domain phosphorylation sites [27- -28] [27-28] IK / I/KK8\\ / ‘(a\ 899 810881158123 8866 8870 8872 ! ®<€®® @9999] p100 (p52) Eel Homology Domain Ankyrin repeats Death Domain phosphorylation sites [35-37] [41-44] [31-32] IKKb, IKKb, [29] [30] PKAc [29] [33] [34] IKKe, [38-39] [40] IKKa, ? ’l? M8K1/2 ’l? PKCE, I? GSIK3b Cl|1k1 Cll<2 RSKI 8%)5 $4 8§6 86318311 T435 8468 8505 S529 S536 phospizerylatioJ Rel Homology Domain Transactivation Domain (TAD) sites [45] [46] [45] r r i T84 8368 8552 (l9 C? 9'9 phospfirzlrglatioJ Leucine zipper Rel Homology Domain Transactivation Domain (T AD)] sites [50-51] [47481 I491 PKCx, PFA ? ‘|? NIIK 8267 $4 8460 S471 phosghsrijlation[ Rel Homology Domain Transactivation Domain (TAD) ] sites Figure 4-2. Some of the known phosphorylation sites of the NF-KB subunits (adapted from a figure in 9 and phosphorylation sites identified in “'38 ). 215 The different kinases responsible for the phosphorylation events shown in Figure 4-2 illustrate how different stimuli can evoke an NF-KB response leading to the transcription of a variety of genes.39 Specific types of activation of NF-KB, such as the chemotherapeutic drug camptothecin, can also result in the subsequent transcription of pro-survival signals and is an important contributor to the tumorgenic properties involved in many kinds of cancer including pancreatic, breast, gastric, and prostate.“°“13 Understanding the signal transduction pathways that NF-KB is involved in would provide valuable information in determining its impact and role in the pathology of the disease. Additionally, it allows for the potential to identify biomarkers associated with the specific types of cancer to help in early diagnosis, prognosis and subsequent treatment. Many chemotherapeutic drugs also induce NF-KB activation,“ which in turn can result in resistance to chemotherapy by NF-KB mediated anti-apoptotic 4547 Since the NF-KB pathway is activated by gene transcription. chemotherapeutic drugs and up-regulated in cancer cells, this provides therapeutic utility to develop additional drugs to inhibit NF-KB, which would 45'48'51 Many research programs have sensitize cancer cells to chemotherapy. developed small molecule inhibitors of NF-KB as adjuvant drugs in an effort to treat cancer more effectively (reviewed in 52). Additionally, the development of these drugs and determination of their mode of inhibition provides a higher understanding on how the regulatory process of NF-KB works, specifically identifying how and where NF-KB is becoming modified and by which proteins. 216 4.4 Phosphorylation events in NF-KB pathway and their impact in cancer therapy The importance and impact that biomarker identification has on cancer treatment has already been addressed earlier in this chapter. Many post- translational modifications of NF-kB have been characterized and been extensively reviewed 3'9 and it is at these sites of modification where many drug targets and biomarkers lie. It has already been illustrated that different modification sites along the activation pathway of NF-KB have been utilized to prevent the subsequent transcription of anti-apoptotic genes. Nonspecific inhibitors of IKK, such as curcumin and quercetin, have been developed to prevent activation of NF-KB by preventing the phosphorylation of I-KB, which indicate that this adjuvant treatment with chemotherapeutics could effectively sensitize tumors.”55 Additional drugs have been developed to inhibit other proteins involved in the NF-KB pathway, preventing the transcription of NF-KB regulated anti- apoptotic genes,56 such as bortezomib, which is clinically used in the treatment of multiple myeloma.57 Other NF-KB pathway inhibitors have been shown to exhibit chemotherapeutic efficacy such as ALLN,58'59 MG-132,6°'61 lactacystin,62 and salinosporamide.63 Clearly, this pathway and its respective proteins play a role in drug discovery and can also play a role as cancer biomarkers as well. The phosphorylation events which lead to activation and subsequent phosphorylation 217 of NF-kB, are critical for the cancer cells oncogenicity, which has been illustrated by a study on NF-KB activated mouse epidermal cells 54 and on tumor growth of mouse colitis-associated cancer.65 4.5.a Previous work to determine NF-KB phosphorylation sites As illustrated in the previous section, the impact in identifying phosphorylation events can prove to be important in the development of chemotherapeutics. As such, the relevance of probing the phosphoproteome in both the diagnosis and treatment of cancer is clear. Extensive research towards the identification of phosphorylation sites of NF-KB has been performed (reviewed in 7'9'10'6%9 ) and is continuing to be an avenue of study. One such study performed phosphorylation analysis of the NF- KB pathway using tissue from patients with either stage I or stage II non-small cell lung cancer (NSCLC).70 In this study, 88 patients with NSCLC had either a biopsy or surgical removal of the tumorgenic tissue, which was used in the data analysis. Along with the tumor tissue, normal lung tissue was also collected as a control. The collected tissues were lysed and the subsequent nuclear extracts were collected. Through the use of both staining methods and electromobility shift assays (EMSA), as would be expected, there was an increase of NF-KB p65 subunit in the tumor cells when compared to that of the normal cells. It was also found that the expression of phosphorylated l-kBa was increased in the tumor tissues compared to the normal lung tissues, even though the total amount of l-KB within the cytoplasm was consistent for both normal and 218 tumor lung tissues. It was also found that 46.6% of patients had NF-KB over- expressed in their tissue, which was also shown in correlation with the severity of the cancer. Additionally, poor survival ratings were linked to both NF-KB and pl- kBq, which coincides with the results obtained in previous studies."72 The use of NF-kB as a biomarker in prognosis was found in other studies to correlate with poor prognosis of other types of cancer as well, including pancreatic, gastric, and breast. “4373 Using a different approach, another study performed quantitative phosphoproteomic analysis in cells observing the tumor necrosis factor (TNF) pathway.74 This pathway has long been identified to work synergistically with the NF-kB pathway. Since NF-KB proteins are responsible for transcribing a variety of different genes involved in metastasis, cell proliferation, inflammation, cell survival and suppression of apoptosis, NF-ch has been implicated in cancer (reviewed extensively in 6'7). The impact TNF-a has on cancer cell survival cooperatively with NF-KB has been a topic of increasing interest. Costas and co- workers found that TNF-q could, in part, be controlling tumor growth by the activation and complex formation between NF-KB and estrogen receptor (ER).75 In this quantitative study, Yates and co-workers grew HeLa cells in either 14N or 15N amino acid mix to isotopically label the proteins, allowing for quantification.74 The whole cell extracts were obtained for both TNF-a treated and untreated cellular environments. The collected whole cell proteins were proteolytically digested with trypsin and subsequently separated on a gel-free isoelectric focusing instrument, which separated the peptides by their pl. 219 Additionally, another fraction containing undigested whole cell proteins, was subjected to IEF prior to digestion. IEF is a tool of choice based on the general idea that upon phosphorylation the isoelectric point (pl) of a protein or peptide will shift from a higher to lower value based on the additional negative charge from the phosphate moiety. An aliquot of each of the IEF fractions were subjected to LC/LC-MS/MS analysis referred to as multidimensional protein identification technology (MudPlT).76 The corresponding results obtained from the IEF fractions for both the intact proteins and the peptides are provided in Tables 4-1 and 4-2. As one would expect, the data from the digested then IEF sample suggests that some enrichment is occurring as the number of phosphorylated peptides goes down with the increasing pH. The sample that first undenrvent IEF and then was digested resulted in no real enrichment of the phosphopeptides. 220 Table 4-1. Table 4-1. unmodified (unmod.) and phosphorylated (phos.) peptides identified in MudPlT IEF unmod. phos. phos./ fractions peptides peptides unmod. pH 3 555 12 1.00 pH 4.6 592 13 1.02 pH 5.4 1867 13 0.32 pH 6.2 1445 0.00 pH 7 1350 0.17 Total non- redundant 5242 41 IEF of digested whole cell protein extract, a comparison between Table 4-2. IEF unmod. phos. fractions proteins peptides peptides pH 3 388 801 3 pH 4.6 327 811 1 pH 5.4 403 1241 7 pH 6.2 566 1723 5 pH 7 516 1085 8 Total non- redundant 1714 5435 23 Table 4-2. IEF of whole cell protein extract, comparing the unmodified and phosphorylated peptides and their corresponding proteins by MudPlT. 221 To further enrich the complex mixture of peptides, the IEF fractions from pH 3 and 4.6 were subjected to IMAC purification followed by LC-MS, since they indicated the highest percentage of phosphopeptides. The enriched samples were subsequently pooled together and analyzed by LC-MS/MS/MS. The results identified a total of 701 phosphopeptides between the triplicate analyses, characterizing phosphoserine, phosphothreonine and phosphotyrosine form both the 14N and 15N isotopically labeled fractions. From the MSIMS analysis, the comparison between the two labeled fractions led to the quantification of 223 of the phosphopeptides. Even more interesting was the identification of 33 phosphopeptides (containing 56 phosphorylation sites) which were up-regulated by at least two-fold and 15 phosphopeptides (containing 23 sites) were down- regulated by two-fold or more as a result of TNF-q activation.74 To eliminate the possibility that some of the results were due to biological isotope effect, the reciprocal experiment was also performed (i.e. the 14N cells were activated with TNF-d while the 15N labeled cells were left untreated). While the reciprocal experiment did enrich many of the same phosphopeptides, none of the up or down regulated phosphopeptides were quantified. The authors hypothesize a few rationales for this observation; including reduced ability to identify the 15N peptides, variability in sample handling, or experimental variation. Even though a large number of the phosphopeptides could not be quantitatively determined due to low signal to noise ratio of the precursor ion peak, they still could be identified and could be compared to the other data sets qualitatively. 222 This led to the identification of 79 unique phosphopeptides that were present in the treated samples but not in the untreated, therefore deeming those as TNF-a regulated.74 Among those phosphopeptides characterized from protein kinase alpha (PKA) which is known to phosphorylate and activate NF-r10% of the untreated and treated IP digests looked the same as the p65 antibody negative control digest (Figures 4- 8 thru 4-10). Not only do the main ions present in the digests not correspond to p65 for either the stimulated or un-stimulated samples, they are all the same ions that are present in the p65 antibody digest. This is most likely due to the significant excess of p65 antibody present in the sample since the antibody:p65 ratio is probably close to 20:1 respectively, which results in those dominant ions. To confirm the hypothesis that all the ions are in fact the same, subsequent sequence analysis of all of the dominant ions present in the digests was performed and it was found that all the ions coincided with the p65 antibody ions. Additionally, none of the sequenced ions corresponded to any phosphopeptide ions. This is not necessarily bad if the enrichment proved successful, as this would illustrate the importance of performing phosphopeptide enrichment, since no ions corresponding to phosphopeptides were found in the pre-enrichment digest. Regardless of the fact that the p65 antibody, the untreated IP and treated lP sample digests all appeared the same and there was no indication of any phosphopeptides, i went on to try the enrichment using the diazo ketone solid support, theorizing that the phosphopeptides present in the 237 digest are most likely to be in sub-stoicheometric levels relative to the p65 anfibody. An aliquot of these methyl ester converted digests were pulled up in tetrahydrofuran and ~20 mg of our optimal enrichment material, the diazo ketone functionalized polystyrene, was added. The covalent coupling took place overnight while being rocked gently at room temperature, the same enrichment conditions as described for the commercially available phosphoprotein digests. The resin was filtered and washed with THF, acetonitrile and methanol to ensure removal of all unbound, non-phosphorylated peptides. After filtration, the resin was exposed to 50% trifluoroacetic acid, 40% dichloromethane overnight to hydrolyze the phosphoester-resin bond, liberating the phosphopeptide. Upon exposure to the cleavage conditions, the eluent (containing the liberated phosphopeptides) was collected by filtration, and dried down under nitrogen. The dried enriched phosphopeptides were desalted with C18 zip tips and subsequently analyzed by MALDI-ion trap MS. The enrichment of the stimulated THP-1 nuclear extract lP proved to be successful, as there were a few phosphopeptides present in the MS spectrum (Figure 4-11). There was no enrichment identified in the un-stimulated nuclear extract IP, which coincides with both the western and silver stain data as the amount of NF-KB is significantly less in the un-stimulated lP. Additionally, there was not any enriched phosphopeptides for the p65 antibody standard digest either, which illustrates that the enriched peptides were from a nuclear protein and not a result of an enriched peptide from a p65 antibody digest. The two 238 confirmed phosphopeptides were the 1738.1 and the 2302.3 ions. The MS/MS data for the 1738.1 ion indicated a dominant loss of ammonia (1720.8 ion) and the loss of methanol (1705.8). There was a smaller ion at 1640.1, indicating a loss of -98, corresponding to -H3PO4 (Figure 4-12). Further confirmation that the 1738 ion was a phosphopeptide was obtained by the MS3 data. The 1705.8 ion was further fragmented and the dominant ion was a -98 loss of -H3PO4 (1607.8), confirming the peptide was phosphorylated (Figure 4-13). Additionally, the ion at m/z 2302.3 was also subjected to MSIMS analysis and the resulting spectra contained a dominant ion at 2204, indicating a -98 loss of -H3PO4 (Figure 4-14). The subsequent MS3 analysis did provide some additional sequence information, as the peptide contained a C-terrninal arginine (Figure 4-15). The other ions present in the enrichment did not exhibit a -98 loss when fragmented, thus they cannot be confirmed (or ruled out) as being phosphopepfides. Although, this data is very promising, as we were able to successfully enrich a few phosphopeptides from the stimulated nuclear extract lP, however, the phosphopeptide ions present in the enrichment spectra, did not correspond to the predicted p65 phosphopeptide ions from a tryptic digest (Table 4-3). There are a few possible reasons for this outcome. These enriched phosphopeptides could belong to one of the 15+ additional proteins that were visualized in the silver stain of the IP sample. Additionally, the protein could be modified in a plethora of different ways, which would also shift the monoisotopic mass, resulting is a lack of lineage for the predicted peptide masses. 239 The subsequent MSIMS analysis then would prove to be a valuable tool in determination if the phosphopeptide ions were in fact from the p65 subunit. Sequence analysis proved to be difficult on the enriched samples, however, due to the dominant H3PO4, and MeOH (since peptides were converted into methyl esters) loss. As indicated previously, not much sequence information, other than determining that the peptide was in fact a phosphopeptide, was determined from MSIMS analysis (Figures 4-12, 4-14). There were a few amino acids that were believed to be present given the MS/MS data, but the partial sequence data did not coincide with any of the p65 protein sequence. The collection of MS3 data did not aid in providing much more sequence information (Figures 4-14, 4-16). Because if this difficultly, the enrichment samples were subjected to hydrolysis under basic conditions, to remove the methyl esters, which could reduce some of the complexity of the MSIMS and MS3 data. Additionally, the fragmentation in the MS/MS and MS3 could be different for the hydrolyzed form of the peptide, providing additional sequence information. I was able to obtain the fully hydrolyzed form of the enrichment sample (the 1738.1 and 2303.0 correspond to the 1649.6 and 2231.1 respectively), but now the baseline appears more noisy, potentially creating more difficulty in interpreting the sequence (Figure 4-16). The ion at mlz 2232.09 corresponds to the methyl ester converted peptide at m/z 2302, which indicates that there are at least four carboxylic acid residues + the C-terrninus present in the peptide, as the hydrolysis led to a 70 Da loss. The MS/MS analysis had an ion present at 2133.6, indicating a loss of -98 (H3PO4) and provided a few sequence ions. 240 Hc 58 0C er I!" However, the ions present in the MSIMS data did not provide any protein sequence hits in the database (Protein Prospector) (Figure 4-17). The ion at m/z 1649.6 is believed to be the hydrolyzed version of the methyl ester converted ion at 1738.1. While the difference in the masses does not add up exactly to equal a number of methyl groups, this could be the result of an additional elimination of water + hydrolysis of the methyl esters. If that is the case, then there are at least four carboxylic acid residues present in this peptide as well. The MSIMS analysis of the ion at m/z 1649.6 confirmed that this peptide is also phosphorylated by the dominant ion at 1551.5 (-H3PO4) (Figure 4-18). The additional sequence ions present in the MSIMS were also input into an MS database to determine the sequence and the protein it is from. Unfortunately no hits matched up well to either the methyl ester converted or fully hydrolyzed sequence ions that were present in the enriched sample. I changed a variety of parameters to see if I could obtain better sequence coverage in the database, but I was never able to get any prospector hits with MSIMS ions that l obtained from any of the enriched peaks. In an effort to obtain part of the peptide sequence, l broadened the search by entering only the main MSIMS ions to, which also proved unsuccessful. This is most likely due to the absence of the protein of interest being in the database. The small sequence information I did obtain from the MS data as well as the determination of the number of acidic residues present, I was able to rule out the possibility that either of the two enriched phosphopeptides belonged to the p65 protein. 241 Further confirmation that the enriched phosphopeptides were from a nuclear extract protein and not from lgG or from the p65 antibody used, was obtained by the enrichment of the negative control digests and there were no phosphopeptides enriched in either of these two controls. Even when l analyzed both the digests and the enriched samples in SIM (selected ion monitoring) for the two phosphopeptides that were enriched in the treated samples, I was not able to observe anything at the m/z value. This led me to conclude that the enriched peptides were not a result of either the antibody or lgG, but from some protein pulled out in the immunoprecipitation experiment. While the success of the enrichment illustrates promise of this approach (and the chosen enrichment material) in its application towards nuclear extract phosphopeptide enrichment, the limitations/challenges that have been identified raise some questions on how to modify this approach to effectively perform the task it was designed for; to identify the phosphopeptides present in NF-KB as a result of activation of its” pathway. 4.8.a Future project directions towards the application of enrichment on cellular proteins The problems that have arisen as a result of using this technique to purify and enrich phosphopeptides from p65 are as follows: (1) the digests are saturated with the p65 antibody, making it very difficult to confirm that p65 peptides are present in the digest, (2) the immunoprecipitation purification still 242 contains many additional proteins whose identity is largely unknown, (3) the enriched phosphopeptides’ protein origin cannot be identified. While these problems are evident there are options to circumnavigate around them. The continuation of this project and its application in nuclear protein enrichment is plausible, by implementing a few modifications. Elimination of the significant excess of p65 antibody in the IP digest could provide very valuable MS data, which would contain peptide ions that were purified by the IF. Using this data, we would gain a better idea of which proteins were present in the IP, provided the proteins were in the database. This could be accomplished one of two ways, either by covalently coupling the antibody to the immobilized support (ref), which could allow for liberation of the affinity bound protein/s by changing the stringency of the buffer used without liberating the antibody as well. Additionally, the immunoprecipitated sample could be separated by gel electrophoresis and the bands of interest could be excised and subjected to in- gel digestion, however, preparatory gel electrophoresis would be required to obtain enough protein to use for phosphopeptide enrichment. The use of gel electrophoresis to purify the proteins in the IP sample, could also address the problem in the identification of the proteins present within the sample. To reduce the tedious extraction, digestion and analysis of every band present, the use of a phosphospecific stain, such as Pro-Diamond Q, could be used to identify the phosphoproteins.89"‘35 The subsequent extraction, digestion and analysis of only the detected phosphoproteins would reduce the sample’s complexity. 243 Q) n\ An additional potential complication that should not be overfooked is the sequence coverage of phosphorylated peptides in a tryptic digest of p65. While it is hypothesized that there are phosphorylation sites on NF-KB that are unknown, which is why we are pursuing to enrich and analyze this protein, there are already some known phosphorylation sites of p65 as a result of TNF-a activation."'10 The known phosphorylation sites were included in the theoretical tryptic digest of p65, however, additional post-translational modifications were not taken into account. As illustrated in Table 4-3, the resulting known phosphopeptides present in a tryptic digest would be challenging albeit impossible to enrich and then observe by MALDI-ion trap MS. Either the monoisotopic mass is too small to be observed relative to the matrix adducts present in the spectrum, or the peptides are too large to even be observed by MALDI-ion trap M8. Implementation of other proteases (chymotrypsin, GIuC, and pepsin) could somewhat improve the sequence coverage in the digestion. 244 Table 4-3. “an 'e.mzra".'1 Ref. Residues Sequence Missed Monoisotopic cleavages mass 311-315 pSlMKK 1 686.4 20 18 274-278 RPpSDR 0 710.3 20 305-314 TYETFKpSIMK 1 1327.6 13 268-278 VSMQLRRPpSDR 1 1424.7 503-551 LVTGAQRPPDPAPAPLGAPGL 0 5052.4 27,30 PNGLLpSGDEDFSpSlADMDFS ALLSQISS 337-502 SSASVPKPAPQPYPFTSSLSTI 0 17141.6 NYDEFPTMVFPSGQISQASALA PAPPQVLPQAPAPAPAPAMVS ALAQAPAPVPVLAPGPPQAVA 22-24 PPAPKPTQAGEGTLSEALLQL QFDDEDLGALLGNSTDPAVFT DLApSVDNSEFQQLLNQGlPVA PH'ITEPMLMEYPEAITR Table 4-3. Predicted masses for the known phosphorylated peptides of a tryptic digest of the p65 subunit of NF-KB 245 4.8 SU of ti‘ 4.8.b Future project direction towards the improvement in enrichment efficiency While we have illustrated that the this solid-phase enrichment approach is successful, further improvements have been hypothesized to increase the level of enrichment using the diazo-phosphate covalent binding including the development of a solution-phase enrichment and a diazo-functionalized support that stabilizes the phosphate-reactive form. 4.8.b.i Solution-phase approach to improved efficiency While solid supports clearly are advantageous for their ease in purification, the reaction can be compromised when performing a solid-phase reaction on ‘in- solution” materials. Additionally, solid supports are very solvent dependent as their swelling properties vary significantly depending on the solvent used. in an effort to improve the percentage of recovery of phosphorylated peptides, looking to change to a solution-phase reaction might prove to be more efficient. By utilizing the same diazo moiety that we know is reactive towards phosphate and placing it on a ‘support’ that is soluble and then purifying the ‘support’ in another manner (i.e. affinity). With that in mind, multiple groups have previously used biotin to react with their modified peptide and then isolated the biotin-peptide by manipulating biotin’s affinity for avidin, using an immobilized avidin column. Specifically, diazo functionalizing a biotin-containing moiety to bind to phosphopeptides and then utilizing its affinity for avidin is an approach worth pursuing. in the introduction chapter l briefly mentioned a few groups who used the biotin-avidin interaction to react with the phosphopeptidesaf”87 However, in 246 these cases the phosphate had been eliminated and the covalent coupling takes place upon the Michael addition of the modified phosphopeptide and the biotin denvafive. In an effort to covalently label the phosphate backbone in nucleotides, Bourget and co-workers used a diazo functionalized biotin material.88 The diazo species was prepared following the synthetic route outline in Scheme 4-1. First, biotin was coupled to 2-amino acetophenone in the presence of isobutyl chloroformate and N-methyl morpholine to form compound 4-1. The coupled biotin-amide was then subjected to conversion of the ketone moiety to its corresponding hydrazone in the presence of hydrazine monohydrate under refluxing conditions to obtain product 4-2. The prepared hydrazone was then oxidized to form its corresponding diazo moiety upon the introduction of manganese dioxide to complete the synthesis of 4-3. The manganese dioxide was removed and the resulting pink solution was removed of solvent under vacuum evaporation. Confirmation of the diazo-biotin product was achieved by both NMR and lR. 247 NH2 0 JUL 1.©/u\ j: HN NH HN NH O 2 S o 2 H "I OH ‘1” S r/V\Ir A S /\/\n/ O Y0 CI 0 4-1 N-methyl morpholine, in DMF JHZN—NHZ 0 it NH2 HN NH HN NH ' N2 Mnoz N \ n = H n S "11W 8 "Ir/\/\"/ 0 O 4-3 4-2 Scheme 4-1. The formation of the diazo-functionalized biotin derivative Upon the successful preparation of the diazo biotin material, a phosphopeptide, angiotensin II (MW 1154.5 when converted to corresponding methyl ester peptide), was introduced to the diazo biotin reagent and the coupling was accomplished using the optimized conditions cited by Bouget and co-workers; heating the mixture in a 1:3:1 solution of DMSO:ACN:H20 in 2mM Boric acid, pH 7.3 (Scheme 4-2). An aliquot from the coupling vessel was removed, dried and desalted with a C18 zip tip prior to MALDI-ion trap MS analysis. Confirmation of the success of the coupling was ascertained by the presence of a molecular weight shift by 346 Da in the spectra (Figure 4-19), corresponding to angiotensin ll-biotin coupled product. While the reaction did not go to completion, as some unmodified angiotensin II peptide is present, the result 248 does illustrate that the coupling is occurring and as such, could be manipulated under modified conditions to yield a higher percent conversion. H o R o H2N_§\ll’N\(lkfi/k§§AOMe o 0 CH2 HNJLNH N2 ’ U “ .,,I N 8 /\/\ll/ 0‘ ,OH O o’/P‘0H 1:3:1 DMSO:ACN:H20 Boric Acid 60°C I H O R O Him-gm” NJ\§§JLOMe H 0 CH2 0‘ ,OH o”P‘o 0 HM \>~NH HN #0 \ 8 Scheme 4-2. The coupling of angiotensin II to diazo biotin The proof of concept of the diazo biotin coupling leads to potential other avenues of study including the application of this approach followed by phosphopeptide purification through the introduction of an immobilized avidin column. Additionally, the phosphopeptide could be liberated from the avidin 249 nu column under acidic conditions to eliminate the potential disassociation problems associated with the avidin-biotin chromatography, which have already been previously addressed. While biotin affinity chromatography is an attractive approach because of its availability and well documented protocols, there are a few limitations using biotin as an affinity tag. The biotin-avidin interaction is quite strong, making elution of the biotin-peptide species sometimes difficult. As a result, other researchers have utilized this established enrichment strategy but developed a solution to the current issues associated with biotin affinity tags. The incorporation of both the ethanedithiol and the maleimide was performed, however, the maleimide was bound to an acid cleavable solid support instead of biotin, thereby eliminating the problems associated with biotin.89 Additional work maintained the use of biotin as an affinity label, but was first coupled to an acid cleavable linker prior to its introduction to the modified peptide and then subsequently liberated the modified peptide from the affinity column under acidic conditions.87 It would also be of interest to compare the solution-phase phosphate coupling of the phenylmethyldiazo biotin with a biotin derivative containing the diazo ketone moiety which has shown to be the more efficient enrichment material in the solid-phase reaction. The preparation of these types of materials can be proposed by the application of the previously used synthetic route for diazo ketone supports. The coupling of an 0- or p- amino substituted benzoic acid in the same manner as illustrated in Scheme 4-2, followed by the conversion for the acid to 250 de VI the acid chloride using thionyl chloride. Upon formation of the acid chloride biotin derivative, the conversion into the diazo ketone can be obtained by the addition of TMS-diazomethane. Thorough washing and subsequent solvent removal in vacuo should remove the un-reacted TMS-diazo methane. Once the material is in hand, it would be important to determine its relative enrichment efficiency compared to the diazo ketone solid support as well as the other in-solution reagent, the phenylmethyldiazo-biotin. O O o HNJLNH DAG” HNJKNH 8 Scheme 4-3. Synthetic route to prepare diazo ketone functionalized biotin denvafive 4.8.b.ii Preparation of a new diazo solid support to improve efficiency Additional diazo-functionalized solid supports could be theorized as being potential phosphopeptide enrichment reagents. As mentioned previously, the 251 basis of the reactivity of the diazo carbonyl moiety towards phosphopeptides is the result of the interchanging resonance forms of the support (Scheme 4-4). ..._ O H O O HO’P‘o 2 2% H H2C o MeO N I \r 11*” aé-NHZ R 0 Scheme 44. Mechanism for enrichment of phosphopeptides. It can be hypothesized that implementation of an additional group to stabilize the formation of the resonance form containing the carbanion, could shift the equilibrium to favor that form. Since the reactivity of the diazo ketone species towards phosphopeptides is a result of that resonance structure, the incorporation of an additional carbonyl would help shift the equilibrium to the 252 negatively charged carbon resonance form, which is phosphopeptide-reactive (Scheme 4-5). VS. 0 O O O W .__——-- W 4. c3_ mi Scheme 4-5. Comparison between diazo ketone solid support and a diazo dicarbonyl solid support Due to the stabilization of the diazo diketone moiety and subsequent application of these materials to perform synthetic transformations, there are numerous publications highlighting the synthesis and use of these types of diazo diketone structures, particularly on solid supports.”93 The seemingly straight- fonrvard preparation of these materials on solid supports also makes these materials good candidates for pursuing their potential as phosphopeptide enrichment supports. As we have already illustrated in Chapter 2, the various reactivities of these diazo-functionalized species, we have not yet pursued manipulating the structure to favor the desired resonance form. 253 4.9 Conclusions The preliminary data illustrating the success of phosphopeptide enrichment from a nuclear extract using our optimal solid support is encouraging for the potential of this technique. The aforementioned problems in both the purification of the protein and its corresponding analysis, limited the approach in its current form. However, there are possible solutions to some of the issues discussed previously and I believe the implementation of these solutions can provide the ability to isolate, enrich and characterize the phosphorylation sites of p65 (or one of its carrier proteins). Additional to the implementation of modifications from the nuclear extract/protein purification end, is the development of a more efficient enrichment material, which could also improved the enrichment of phosphopeptides from a nuclear extract IP sample. The concept of developing a solution-phase enrichment support using a functionalized biotin is a possible approach, which has shown promise in it preliminary data. The advantages in both reaction time and efficiency by implementing solution-phase chemistry, instead of mediating the enrichment through a solid-support, is an avenue worth pursuing. The use of developing a material, which shifts the equilibrium to the reactive ‘carbanion’ resonance form, could also prove to be an additional material (or group of materials) to analyze for the enrichment capabilities. Lastly, i would like to briefly highlight the impact that phosphoproteomic characterization and technologies have had on the identification of cancer biomarkers and drug targets. The sheer number of publications that are relevant 254 to this area illustrate its importance and depth of research ongoing in the field. While our phosphoproteomic technique and the other enrichment approaches discussed herein, indicate that much progress in this area have been made in the last two decades, there are still limitations with each technique and no ‘gold- standard’ has yet to be found for phosphoprotein enrichment and characterization. The merging of technologies seems to be on the rise, leading toward a more encompassing approach that utilizes multiple methods, as the limitations of one approach may not be limitations in another. This type of cooperative analysis yields more complete phosphoproteomic analysis and results. While complete global phosphorylation site elucidation is not yet routine, publications everyday are identifying novel modification sites and including these sites on protein databases, enabling more complete sequence information for database analysis. Additionally, new MS sequencing parameters are being renovated to provide more complete phosphopeptide identification and sequence analysis/interpretation. improving the MS automated capabilities allows for larger datasets to be analyzed with a higher confidence and fewer ‘false positive’ results. All of the technologies developed for phosphoprotein identification and quantification have increased the capabilities of these phosphoproteins to be used as biomarkers. Specific elucidation of signal transduction pathways pertaining to cancer improves the breadth of use of phosphoproteins as predictive or diagnostic markers, and potential drug targets. 255 4.10 Experimental procedures Materials Sequencing-grade trypsin was purchased from Promega. Cell culture materials were all purchased from Gibco. Biotin and additional synthetic reagents were all purchased from Sigma Aldrich Chemical Co. All primary and secondary antibodies and the antibody-agarose conjugates were all purchased from Santa Cruz Biotechnology. Nuclear Extraction of CEM cells THP-1, CEM, L363, RPMI 8336 cells were all grown in RPMI-1640 media supplemented with 10% fetal bovine serum, penicillin (614 ng/mL), streptomycin (10 pg/mL), and HEPES buffer (pH 7.2) at 37°C, 5% C02. The activated cells (~ 1.0 x 106 cells/mL) were subsequently stimulated with TNF-q (0.05 pg/mL) for 30 min. The cells (~ 4.0 x 107 total cells) were harvested by centrifugation, washed in ice-cold PBS (1 mL), and the cellular pellets were lysed on ice in 1 mL cytoplasmic extraction buffer (10 mM HEPES buffer, pH 7.9, 10 mM potassium chloride, 10 mM sodium fluoride, 1.5 mM magnesium chloride, 1.0% Nonidet p- 40, 1 mM sodium orthovanadate, 1 mM B—glycerophosphate, 0.5 mM dithiothreitol, 0.5 mM phenylmethylsulfonyl fluoride, 5pL leupeptin, 5pL aprotinin). Nuclei were pelleted (10,000 RPM, 4°C, 1 minute), and maintained on ice and the cytosolic extracts were extracted off using a pipet and stored at -80 °C . The nuclei were resuspended by vortexing at 4°C for 20 minutes in 120 pL of nuclear extraction buffer (20 pM HEPES buffer, pH 7.9, 0.42 M sodium chloride, 10 mM 256 sodium fluoride, 1.5 mM magnesium chloride, 25% glycerol, 1 mM sodium orthovanadate, 1 mM B-glycerophosphate, 0.5 mM dithiothreitol, 0.5 mM phenylmethylsulfonyl fluoride, 0.2 mM EDTA, 5 pL leupeptin, 5pL aprotinin). The nuclear extracts were cleared (14,000 RPM, 4 °C. 10 minutes), transferred to fresh tubes and pulled up in 240 pL of nuclear protein storage buffer (20 mM HEPES buffer, pH 7.9, 0.1 M potassium chloride, 10 mM sodium fluoride, 20% glycerol, 1 mM sodium orthovanadate, 1 mM B-glycerophosphate, 0.5 mM dithiothreitol, 0.5 mM phenylmethylsulfonyl fluoride, 0.2 mM EDTA, 5 pL leupeptin, 5 pL aprotinin). Protein concentrations were determined by the Bradford assay. The nuclear extracts were all stored at -80 °C and thawed on ice prior to their use for either the western blot analysis or for immunoprecipitation punficafion. Bradford Assay of Nuclear Extracts A five point standard curve was obtained using lgG (0 pg/pL, 0.1 pg/pL, 0.2 pg/pL, 0.4 pg/pL, 0.8 pg/pL) in a 1:4 dilution of nuclear storage buffer (20 mM HEPES buffer, pH 7.9, 0.1 M potassium chloride, 10 mM sodium fluoride, 20% glycerol, 1 mM sodium orthovanadate, 1 mM B-glycerophosphate, 0.5 mM dithiothreitol, 0.5 mM phenylmethylsulfonyl fluoride, 0.2 mM EDTA, 5 pL leupeptin, 5 pL aprotinin) in sterile water (40 pL total volume for each standard). The standards were then pulled up in 500 pL Bradford Reagent Dye (diluted 1:4 in deionized water), and their absorbance was measured at a wavelength of 595 nm. Nuclear extracts were prepared as outlined above and an aliquot of each 257 nuclear extract sample (1 pL) was diluted in deionized water (39 pL) and then further diluted in 500 pL Bradford Reagent Dye (diluted 1:4 in deionized water), the absorbance measured at a wavelength of 595 nm and its corresponding protein concentration was determined by comparison to the prepared standard curve. p65 Western Blot Analysis of Nuclear Extracts Each nuclear extract samples (20 pg) diluted in deionized water (15 pL total volume) and further diluted in 15pL of 2x sample buffer (10 mM tris pH 6.8, 0.2 M sodium dodecyl sulfate, 20% glycerol, 0.01% bromophenol blue, 2% 2- mercaptoethanol). The samples were boiled for 3 min, briefly centrifuged and an aliquot (20 pL) of each sample was loaded onto a 10% Tris-SDS gel and run at 200 const. V until tracking dye ran off gel. The proteins were electroblotted to a PVDF membrane in Towbin buffer (25 mM tris base, 192 mM glycine, 5% methanol) at 60 V for 75 minutes. The membrane was blocked in 4% non-fat dairy milk for 1 hour at room temperature and then washed for 10 minutes in TBS-T buffer (50 mM tris base pH 7.6, 154 mM sodium chloride, 0.2% Tween- 20) and probed overnight at 4 °C with p65 antibody (Santa Cruz sc-372) that was diluted 1:1000 in 4% non-fat dry milk (10 mL). The membrane was washed 3x10 minutes in TBS-T buffer and probed for 1 hr. with the 2° antibody (anti-rabbit conjugated to horseradish peroxidase) that was diluted 1:3000 in 4% non-fat dry milk (10 mL). The membrane was washed again 3x10 minutes in TBS-T buffer and the detection reagent (GE Healthcare) was introduced for 1 min (2 mL) and 258 then exposed the film for detection. To confirm the same amount of protein was loaded for each sample, the membrane was probed for actin for three hours at room temperature with actin antibody that was diluted to 1:10000 in 4% non-fat dry milk (10 mL). The membrane was washed 3x10 minutes in TBS-T buffer and probed for 1 hr. with the 2° antibody (anti-mouse conjugated to horseradish peroxidase) that was diluted 1:3000 in 4% non-fat dry milk (10 mL). The membrane was washed again 3x10 minutes in TBS-T buffer and the detection reagent (GE Healthcare) was introduced for 1 min (2 mL) and then exposed the film for detection. immunoprecipitation of p65 from Nuclear Extracts Nuclear extract samples (200-1000 pg) were pre-cleared by adding them to lgG (1 pg) and Protein A/G Plus Agarose (20 pL) and incubated at 4 °C for 30 min. The samples were centrifuged at 10,000 RPM for 1 min at 4 °C. The eluent was removed and placed in an eppendorf tube containing p65-antibody agarose conjugate (Santa Cruz sc-372) (20 pL), pulled up in cytoplasmic extraction buffer (10 mM HEPES buffer, pH 7.9, 10 mM potassium chloride, 10 mM sodium fluoride, 1.5 mM magnesium chloride, 1.0% Nonidet p-40, 1 mM sodium orthovanadate, 1 mM B-glycerophosphate, 0.5 mM dithiothreitol, 0.5 mM phenylmethylsulfonyl fluoride, 5pL leupeptin, 5pL aprotinin) (200 pL) and incubated by rocking overnight at 4 °C. The immunoprecipitate was centrifuged 10,000 RPM for 1 min and the eluent was removed by a pipet. The protein- antibody-agarose pellet was washed 2x in cytoplasmic extraction buffer (500 pL) 259 and 2x PBS (500 pL) and placed on ice until further use in either proteolytic digestion or gel electrophoresis. p65 Western Blot Analysis of lmmunoprecipitated Samples The immuoprecipitatied sample pellet (200 pg total protein) was diluted in 30 pL of 2x electrophoresis sample buffer (10 mM tris pH 6.8, 0.2 M sodium dodecyl sulfate, 20% glycerol, 0.01 % bromophenol blue, 2% 2-mercaptoethanol) and boiled at 100 °C for 3 minutes. An aliquot (20 pL) of sample was loaded onto a 10% Tris-SDS gel and run at 200 const. V until tracking dye ran off gel. The proteins were electroblotted to a PVDF membrane in Towbin buffer (25 mM tris base, 192 mM glycine, 5% methanol) at 60 V for 75 minutes. The membrane was then blocked in 4% non-fat dairy milk for 1 hour at room temperature and then washed for 10 minutes in TBS-T buffer (50 mM tris base pH 7.6, 154 mM sodium chloride, 0.2% Tween-20) and probed overnight at 4°C with p65 antibody (Santa Cruz sc-8008) diluted 1:250 in 4% non-fat dry milk (10 mL). The membrane was washed 3x10 minutes in TBS-T buffer and probed for 1 hr. with the 2° antibody (anti-mouse horseradish peroxidase conjugate) diluted 1:3000 in 4% non-fat dry milk (10 mL). The membrane was washed again 3x10 minutes in TBS-T buffer and the detection reagent (GE Healthcare) was introduced for 1 min (2 mL) and then exposed to film for detection. 260 Tryptic Digestion of lmmunoprecipitated Samples and the Conversion of the Peptides to Methyl Esters The immuoprecipitatied sample pellet (1000 pg total protein) 20 pL of a denaturing solution (6 M urea, 50 mM Tris-HCI) was added along with 5 pL of dithiolthreitol (10 mM) and the sample was incubated at 65 °C for 1 hour. The sample was cooled to room temperature and 160 pL of ammonium bicarbonate (50 mM) and 10 pL of iodoacetamide (100 mM) was added and the sample was incubated at room temperature in the dark for 1 hour. A 10 pL aliquot of sequencing-grade trypsin (purchased from Promega) was then added (0.5 pg/ pL) and the sample was incubated overnight at 37 °C. The digested sample was then dried down under nitrogen gas, pulled up in a solution of 30% acetonitrile in water (200 pL), aliquoted out into 50 pL aliquots, then dried down under nitrogen again. The dried digests were converted to their methyl esters upon the addition of methanolic hydrochloric acid, which was prepared by the addition of acetyl chloride (320 pL) to anhydrous methanol (2 mL). An aliquot (40 pL) of this solution was added to dried digests and incubated at room temperature for 2 hr. The methyl ester converted digest was then dried down under nitrogen and subsequently stored at -20 °C until their use. Enrichment of phosphorylated peptides from a digest of an immunoprecipitated protein using diazo functionalized resins Diazo functionalized material (~20 mg) and tetrahydrofuran (1 mL) was added to dried methyl ester converted immunoprecipitated protein digest. The mixture 261 man: .I l was rockedg filtration and and then met was complete 10% water I temperature, solution. Th combined 6 samples we following ti acetonitrile and inlrod liberatedt In a ima\ MALDI-i SV“~ht Teas cook; iSObu SOlut ”“0c and was rocked gently overnight at room temperature. The resin was recovered by filtration and washed with tetrahydrofuran (5x500 pL), acetonitrile (3x 500 pL) and then methanol (3x 500 pL). Cleavage of the captured species from the resin was completed by incubation in a solution 50% trifluoroacetic acid, 40% DCM, 10% water (500 pL). The mixture was rocked gently overnight at room temperature, allowing for full interaction between the resin and the cleavage solution. The resin was filtered and washed once with methanol (500 pL). The combined eluents were concentrated under nitrogen. Prior to analysis, the samples were diluted in water (20 pL) and desalted using Millipore C18 Zip Tips, following the manufacturers protocol. Briefly, the zip tip was moistened with acetonitrile (3x10 pL), then equilibrated with 0.1% trifluoroacetic acid (3x10 pL) and introduced to the enrichment sample (18x10 pL). The bound peptides were liberated from the zip tip with 50:50 0.1% trifluoroacetic acidzacetonitrile, resulting in a final volume of 6 pL. All samples were subsequently stored at -20°C prior to MALDI-ion trap MS analysis. Synthesis of 4-1a. 2-(N-Biotinoylamino)acetophenone To a solution of U-biotin (1.0 g, 4.1 mmol) in dry N,N—dimethylformamide (45 mL) coole dat 0 °C under argon, N-methylmorpholine (590 pL, 5.33 mmol) and isobutyl chloroformate (840 pL, 6.60 mmol) were added successively. The solution was stirred for 30 min, and then 2-aminoacetophenone (824 mg, 6.10 mmol) was added. The solution was then stirred at room temperature for 3.5 h and then the solvent was removed in vacuo. The residue was triturated with cold 262 water (50 mL) and the resulting precipitate was filtered, washed with water and recrystallized from ethanol to obtain the coupled product 4-1a (0.8 mg, 53%). lR (NaCl plate): 3238, 2920, 1699, 1653, 1450, 1247 cm". 1HNMR (500 MHz, DMSO-d5): 6 = 1.38-1.66 (m, 6H), 2.38 (t, J = 7.0 Hz, 2H), 2.58 (d, J = 7.5, 1H), 2.61 (s, 3H), 2.82 (dd, J: 5.0, 12.5 Hz, 1H), 3.11 (m, 1H), 4.14 (m, 1H), 4.31 (m, 1H), 6.35 (br s, 1H), 6.44 (br s, 1H), 7.19 (t, J = 7.0 Hz, 1H), 7.58 (t, J = 7.5 Hz, 1H), 7.97 (d, J = 7.5 Hz, 1H), 8.33 (d, J = 8.5 Hz, 1H), 11.25 (br s, 1 H). 13cme (500 MHz, DMSO-d6): 6 = 24.87, 28.03, 28.06, 28.69, 37.12, 55.33, 59.18, 61.00, 120.54, 122.78, 124.20, 131.48, 134.06, 139.03, 162.70, 171.49, 202.59. MS (calc. for M+ = 361.15 ), found: 362.1 (M-H“). Synthesis of 4-2. 2-(N-Biotinoylamino)acetophenone hydrazone To a solution of the ketone 4-1a (0.40 g, 1.11 mmol) in absolute ethanol (8 mL) and hydrazine monohydrate (570 pL, 11.0 mmol) was added. The solution was refluxed for 50 min and then solvents were evaporated. The residue was suspended in water, filtered, washed with water and with ether and dried in vacuo to afford hydrazone 4-1b (0.30 g, 73 %). IR (NaCl plate): 3249 (br), 2921, 1697, 1608, 1530, 1446 cm". 1HNMR (500 MHz, DMSO-d6): 5 = 1.31-1.63 (m, 6H), 2.10 (s, 3H), 2.32 (t, J = 7.4 Hz, 2H), 2.57 (d, J = 12.5, 1H), 2.82 (dd, J = 5.0, 12.5 Hz, 1H), 3.10 (m, 1H), 4.14 (m, 1H), 4.30 (m, 1H), 6.36 (br s, 1H), 6.43 (br s, 1H), 6.61 (br s, 2H), 7.04 (t, J = 7.5 Hz, 1H), 7.20 (t, J = 8.0 Hz, 1H), 7.45 (d, J = 7.5 Hz, 1H), 8.36 (d, J = 8.0 Hz, 1H), 11.98 (br s, 1 H). ”CNMR (500 MHZ, DMSO-d6): 5 = 13.19, 13.23, 25.15, 28.06, 28.16, 37.41, 55.43, 59.18, 263 61.06, 120.11, 122.56, 125.55, 127.40, 127.63, 136.71, 145.36, 162.71, 170.90. MS (calc. for M+ = 375.17), found: 375.2 (M*). Synthesis of 4-3. 2-(N-Biotinoylamino)phenylmethyldiazomethane To a solution of hydrazone (0.30 g, 0.80 mmol) in 10 mL N,N-dimethylformamide, Mi’IOz (800 mg) was added and the suspension was stirred for 15 min at room temperature. The mixture was suction filtered through Celite (~1 cm thickness). The filtrate was placed into a clean 25 mL round bottom flask and the solvent was evaporated in vacuo. The residue was then triturated with ether to obtain product 4-1, as a pink powder (0.18 g, 60%). IR (NaCl plate): 3250, 2039, 1700, 1685 cm". 1HNMR (500 MHz, DMSO-d6): 5 = 1.39-1.64 (m, 6H), 2.13 (s, 3H), 2.26 (t, J = 7.0 Hz, 2H), 2.59 (d, J = 12.5, 1H), 2.83 (dd, J = 4.0, 12.5 Hz, 1H), 3.13 (m, 1H), 4.15 (m, 1H), 4.32 (m, 1H), 6.36 (br s, 1H), 6.44 (br s, 1H), 7.02- 7.21 (m, 4H), 9.37 (brs, 1 H). ”CNMR (500 MHz, DMSO- d6): 5 = 12.26, 24.96, 28.08, 28.28, 35.10, 55.39, 59.20, 61.03, 124.14, 124.80, 126.33, 127.63, 128.20, 132.76, 162.70, 171.09. MS (calc. for M+ = 373.47 ), found: 345.2 (M‘- N2). Biotin-phenylmethyldiazomethane-phosphopeptide coupling In an eppendorf tube containing a dried aliquot of angiotensin II converted to its methyl esters (100 pmol), a small amount of the compound 4-1 (~5 mg) was added. The mixture was dissolved in 1:3:1 DMSO:acetonitrile:water solution (100 pL total) containing 2 mM H3B03 (pH 7.3) and incubated at 60 °C for 2 264 hours. An aliquot of the solution (10 pL) was removed from the mixture, and was dried under nitrogen. Prior to analysis, the samples were diluted in water (20 pL) and desalted using Millipore C18 Zip Tips, following the manufacturers protocol. Briefly, the zip tip was moistened with acetonitrile (3x10 pL), then equilibrated with 0.1% trifluoroacetic acid (3x10 pL) and introduced to the enrichment sample (18x10 pL). The bound peptides were liberated from the zip tip with 50:50 0.1% trifluoroacetic acidzacetonitrile, resulting in a final volume of 6 pL. The samples were subsequently stored at -20°C prior to MALDI-ion trap MS analysis. MALDI-ion trap MS: ion at m/z 1154.5 (angiotensin II) and ion at m/z 1500.6 (angiotensin ll-coupled to biotin derivative) Mass Spectrometry M8 and CID-MSIMS analyses were performed using a linear quadrupole ion trap mass spectrometer equipped with a Matrix Assisted Laser Desorption Ionization (MALDI) source (model vMALDl-LTQ, Thermo, San Jose, CA). Desalted enrichment samples were spotted on MALDI plate (0.5 pL) and an aliquot of DHB matrix (10 mg/mL in a solution of 50:50 acetonitrile:1 % phosphoric acid) was spotted over the sample. Additionally, another sample was spotted on the MALDI plate (0.5 pL) and then overlaid with 1 pL a-cyano matrix (10 mglmL in a solution of 50:50 acetonitrile: 0.1 % trifluoroacetic acid). 265 .f’iimmltt! I _ - _ , Appendix Figure 4-8. Reiatlveintensiw 8 8 l l 1 J L l I I I q 8 0) (O 0) i9. :3 ... r3 2‘3 (.0 O 99' SCSI SS'W‘QI SP'SQQ l- 9}. 89:2 #98898 £31909 9L'ZZLS 60m :'9 s 60' A Figure 4-8. MALDI-ion trap MS of the p65 antibody standard tryptic digest. 266 Figure 4-9. RelalveinMnsity 8 8 l L J l L l l l l q 0 9 N w (0 8 o) o _L [0 ‘ .8 — _. 8 (It N 9° _. 3 8 _. in Q (It .2 5 0| N) 3 s N Q 00 SL'VLSZ 388893 "$909 if: 0 awesome; -' fi—‘-—"‘vv-wv—vr'—'-V—-v'vv———va ‘1” £312 8 Figure 4-9. MALDI-ion trap MS of the THP-1 untreated nuclear extract immunoprecipitation tryptic digest. 267 Figure 4-10. Relative intensity _L o 8 8 l I I l I I I [J ‘4 .3 N ”it b) G _e 3 2° A (.0 g 0 9" _m A 0'! O .3 0| 0| SV'WQI 91.1935 5. b '5. “E 88 .00 9° ~59 o... .8 at“. ... 9 0' L. 2 on Figure 4-10. MALDI-ion trap MS of the THP-1 treated nuclear extract immunoprecipitation tryptic digest. 268 Figure 4-11. Relative intensity 009 003i OO'OQSI 60 891i 0002: 1.28063 000$ (1)59 00017 Figure 4-11. MALDI-ion trap MS of the THP-1 treated nuclear extract immunoprecipitation tryptic digest enrichment. 269 Figure 4-12. Relative intensity o 8 8 8 l I I L l L l I i o .0 l o—w' g o .1 —t I”: e- -2 MH-. 8 2 _r l _l 23 Zn‘ 8" .8 m M 25': 93 .5 I o“ a; 83 SI 0031 L819“ 58 OCLII Figure 4-12. MALDI-ion trap CID MSIMS of ion 1738 from the THP-1 treated nuclear extract IP enrichment 270 Figure 4-13. Relative intensity m .4 o o 8 8 llllIJJlf 0 fl .1 *0) 8 8“ r _.L. 58— A 0 __§ 5” Ur —:m —_3 A 0 TN “at w _l 01 °— °:- L gal: “3 a 3. a) 00 9° to .I ... Q o 0 Figure 4-13. MALDI-ion trap CID MS3 of ion 1738, 1705 from the THP-1 treated nuclear extract lP enrichment 271 Figure 4-14. Relative inten sity _. o. 8 C) O lLl'lllJ 008 003i 3 009i 0003 COWZZ ’0ch- 00% 0098.71. ‘Hls- Figure 4-14. MALDI-ion trap CID MS/MS of ion 2302 from the THP—1 treated lP tryptic digest enrichment. 272 Figure 4-15. Relative intensity OH O 00L lllllJl‘ll1 4w 09H 0031 096 001.1. 9!}.le lG'CLQI 096i 396303 009113 ‘HN- 0018M 0033 Figure 4-15. MALDI-ion trap CID MS3 of the 2302, 2204 ion from the THP-1 treated enrichment 273 ‘4___-2—. an?" _ I‘ ‘— Figure 4-16. Relativeinhensily 0| 0 O l '—00L 1 I I l g l 009 V9'899L W'GVQL 0003 SL'3303 603833 an: 0008 0093 0099 I 000i? Figure 4-16. MALDI-ion trap MS of the THP-1 treated nuclear extract IP tryptic digest enrichment, NH4OH hydrolysis 274 Figure 4-1 7. Relative intensity .3 (II D O o Q g lLllllillJ O —_4 8.— 0— 003i 4w OOSL 008L SS'SLGI OOLZ W'SHZ 99 re L3 'Od‘H- oovz: LL! I Figure 4-17. MALDI-ion trap CID MSIMS of ion 2232 from THP-1 treated nuclear extract IP tryptic digest enrichment, NH4OH hydrolysis 275 Figure 4-18. Relative Intensity 006 099 8L'6178 L — 0 ~— 00l 09” mm; 0017i LZ'PLEI 099i £368” 9v ISSI 'oa‘H- 97' ESQI 0 H- (1)61. Figure 4-18. MALDI-ion trap CID MSIMS of ion 1649 from THP-1 treated nuclear extract IP tryptic digest enrichment, NH4OH hydrolysis 276 Figure 4-19. Relative in ten sity o 8 8 <5 I I L! 1 1 J i I O C.) ,2; )9 f. o 01 O 01 fl I 11 I; O D a 6‘. O .0 _. 2 O) 0.. L.) a 8 M O 8 Figure 4-19. MALDI-ion trap MS angiotensin II-diazo biotin binding. 277 10. References Sawyers, C. L. The cancer biomarker problem. Nature 2008, 452, (7187), 548-552. Sharma, S. V.; Bell, D. W.; Settleman, J.; Haber, D. A. Epidermal growth factor receptor mutations in lung cancer. Nature Reviews Cancer 2007, 7, (3), 169-181. Etzioni, R.; Urban, N.; Ramsey, S.; McIntosh, M.; Schwartz, 8.; Reid, 8.; Radich, J.; Anderson, 6.; Hartwell, L. The case for early detection. Nature Reviews Cancer 2003, 3, (4), 243-252. Broxterman, H. J.; Lankelma, J.; Hoekman, K. Resistance to cytotoxic and anti-angiogenic anticancer agents: Similarities and differences. Drug Resistance Updates 2003, 6, (3), 111-127. Sen, R.; Baltimore, D. Inducibility of kappa immunoglobulin enhancer- binding protein NF-kappa B by a posttranslational mechanism. Cell 1986, 47, 921-928. Aggarwal, B. B. S., G.; Nair, A.; Ichikawa H. Nuclear factor-kappa B: A holy grail in cancer prevention and therapy. Current Signal Transduction Therapy 2006, 1, 25—52. lnoue, J.; Gohda, J.; Akiyama, T.; Semba, K. NF-kappa B activation in development and progression of cancer. Cancer Science 2007, 98, (3), 268-274. Sebban, H.; Yamaoka, S.; Courtois, G. Posttranslational modifications of NEMO and its partners in NF-kappa B signaling. Trends in Cell Biology 2006, 16, (11), 569-577. Perkins, N. D. Post-translational modifications regulating the activity and function of the nuclear factor kappa B pathway. Oncogene 2006, 25, (51), 6717-6730. Viatour, P.; Merville, M. P.; Bours, V.; Chariot, A. Phosphorylation of NF- kappa B and l kappa B proteins: Implications in cancer and inflammation. Trends in Biochemical Sciences 2005, 30, (1 ), 43-52. 278 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. Hou, S. H.; Guan, H. C.; Ricciardi, R. P. Phosphorylation of serine 337 of NF-kappa B p50 is critical for DNA binding. Journal of Biological Chemistry 2003, 278, (46), 45994-45998. Demarchi, F.; Bertoli, C.; Sandy, P.; Schneider, C. Glycogen synthase kinase-3 beta regulates NF-kappa B1 /p105 stability. Joumal of Biological Chemistry 2003, 278, (41 ), 39583-39590. Lang, V.; Janzen, J.; Fischer, G. 2.; Soneji, Y.; Beinke, S.; Salmeron, A., et al. Beta TRCP-mediated proteolysis of NF-kappa B1 p105 requires phosphorylation of p105 serines 927 and 932. Molecular and Cellular Biology 2003, 23, (1 ), 402-413. Senftleben, U.; Cao, Y. X.; Xiao, G. T.; Greten, F. R.; Krahn, G.; Bonizzi, G., et al. Activation by lKKa of a second, evolutionary conserved, NF- kappa B signaling pathway. Science 2001, 293, (5534), 1495-1499. Xiao, G. T.; Harhaj, E. W.; Sun, S. C. NF-kappa B-inducing kinase regulates the processing of NF-kappa 82 p100. Molecular Cell 2001, 7, (2),401-409. Anrather, J.; Racchumi, G.; ladecola, C. Cis-acting, element-specific transcriptional activity of differentially phosphorylated nuclear factor-kappa B. Journal of Biological Chemistry 2005, 280, (1 ), 244-252. Ryo, A.; Suizu, F.; Yoshida, Y.; Perrem, K.; Liou, Y. C.; Wulf, G.; Rottapel, R.; Yamaoka, 8.; Lu, K. P. Regulation of NF-kappa B signaling by pin1- dependent prolyl isomerization and ubiquitin-mediated proteolysis of p65/relA. Molecular Cell 2003, 12, (6), 1413-1426. Venneulen, L.; De Wilde, G.; Van Damme, P.; Vanden Berghe, W.; Haegeman, G. Transcriptional activation of the NF-kappa B p65 subunit by mitogen- and stress-activated protein kinase-1 (MSK1). EMBO Journal 2003, 22, (6), 1313-1324. Zhong, H. H.; Voll, R. E.; Ghosh, 8., Phosphorylation of nf-kappa b p65 by pka stimulates transcriptional activity by promoting a novel bivalent interaction with the coactivator cbp/p300. Molecular Cell 1998, 1, (5), 661- 671. Duran, A.; Diaz—Meco, M. T.; Moscat, J. Essential role of relA ser311 phosphorylation by zeta PKC in NF-kappa B transcriptional activation. EMBO Journal 2003, 22, (15), 3910-3918. 279 21. 22. 23. 24. 25. 26. 27. 28. 29. Yeh, P. Y.; Yeh, K. H.; Chuang, S. E.; Song, Y. C.; Cheng, A. L. Suppression of MEK/ERK signaling pathway enhances Cisplatin-induced nf-kappa b activation by protein phosphatase 4-mediated NF-kappa B p65 THR dephosphorylation. Journal of Biological Chemistry 2004, 279, (25), 26143-26148. Buss, H.; Dorrie, A.; Schmitz, M. L.; Frank, R.; Livingstone, M.; Resch, K.; Kracht, M. Phosphorylation of serine 468 by GSK-3B negatively regulates basal p65 NF-kappa B activity. Journal of Biological Chemistry 2004, 279, (48), 49571 -49574. Mattioli, l.; Geng, H.; Sebald, A.; Hodel, M.; Bucher, C.; Kracht, M.; Schmitz, M. L. Inducible phosphorylation of NF-kappa B p65 at serine 468 by T cell costimulation is mediated by IKK epsilon. Journal of Biological Chemistry 2006, 281 , (10), 6175-6183. Schwabe, R. F.; Sakurai, H. IKKB phosphorylates p65 at $468 in transactivaton domain 2. FASEB Journal 2005, 19, (9), 1758-1760. Campbell, K. J.; Witty, J. M.; Rocha, S.; Perkins, N. D. Cisplatin mimics ARF tumor suppressor regulation of relA (p65) nuclear factor-kappa B transactivation. Cancer Research 2006, 66, (2), 929-935. Rocha, 8.; Garrett, M. D.; Campbell, K. J.; Schumm, K.; Perkins, N. D. Regulation of NF-kappa B and p53 through activation of ATR and CHK1 by the ARF tumour suppressor. EMBO Journal 2005, 24, (6), 1157-1169. Wang, D.; Westerheide, S. 0.; Hanson, J. L.; Baldwin, A. S. Tumor necrosis factor alpha-induced phosphorylation of reIA/p65 on ser(529) is controlled by casein kinase II. Journal of Biological Chemistry 2000, 275, (42), 32592-32597. Buss, H.; Dorrie, A.; Schmitz, M. L.; Hoffmann, E.; Resch, K.; Kracht, M. Constitutive and interleukin-1-inducible phosphorylation of p65 NF-kappa B at serine 536 is mediated by multiple protein kinases including l kappa B kinase (IKK)-d, IKKB, IKK epsilon, TRAF family member-associated (TANK)-binding kinase 1 (T BK1 ), and an unknown kinase and couples p65 to tata-binding protein-associated factor |l31-mediated interleukin-8 transcription. Journal of Biological Chemistry 2004, 279, (53), 55633- 55643. Lawrence, T.; Bebien, M.; Liu, G. Y.; Nizet, V.; Karin, M. IKKa limits macrophage NF-kappa B activation and contributes to the resolution of inflammation. Nature 2005, 434, (7037), 1138-1143. 280 30. 31. 32. 33. 34. 35. 36. 37. 38. Sakurai, H.; Chiba, H.; Miyoshi, H.; Sugita, T.; Toriumi, W. I kappa B kinases phosphorylate NF-kappa B p65 subunit on serine 536 in the transactivation domain. Journal of Biological Chemistry 1999, 274, (43), 30353-30356. Sizemore, N.; Lerner, N.; Dombrowski, N.; Sakurai, H.; Stark, G. R. Distinct roles of the l kappa B kinase C: and [3 subunits in liberating nuclear factor kappa b (NF kappa B) from i kappa b and in phosphorylating the p65 subunit of NF-kappa B. Journal of Biological Chemistry 2002, 277, (6), 3863-3869. Marienfeld, R.; Berberich-Siebelt, F.; Berberich, l.; Denk, A.; Serfling, E.; Neumann, M. Signal-specific and phosphorylation-dependent reIB degradation: A potential mechanism of NF-kappa B control. Oncogene 2001, 20, (56), 8142-8147. Maier, H. J.; Marienfeld, R.; Wirth, T.; Baumann, B. Critical role of relB serine 368 for dimerization and p100 stabilization. Journal of Biological Chemistry 2003, 278, (40), 39242-39250. Mosialos, G.; Hamer, P.; Capobianco, A. J.; Laursen, R. A.; Gilmore, T. D. A protein kinase-a recognition sequence is structurally linked to transformation by p59v-rel and cytoplasmic retention of p680-rel. Molecular and Cellular Biology 1 991 , 1 1 , (12), 5867-5877. Yu, S. H.; Chiang, W. 0.; Shih, H. M.; Wu, K. J. Stimulation of c-rel transcriptional activity by PKA catalytic subunit beta. Journal of Molecular Medicine-[mm 2004, 82, (9), 621-628. Starczynowski, D. T.; Reynolds, J. G.; Gilmore, T. D. Mutations of tumor necrosis factor alpha-responsive serine residues within the c-terminal transactivation domain of human transcription factor rel enhance its in vitro transforming ability. Oncogene 2005, 24, (49), 7355-7368. Martin, A. G.; Fresno, M. Tumor necrosis factor-alpha activation of NF- kappa B requires the phosphorylation of ser—471 in the transactivation domain of c-rel. Joumal of Biological Chemistry 2000, 275, (32), 24383- 24391. Sanchez-Valdepenas, C.; Martin, A. G.; Ramakrishnan, P.; Wallach, D.; Fresno, M. NF-kappa B-inducing kinase is involved in the activation of the cd28 responsive element through phosphorylation of c-rel and regulation of its transactivating activity. Journal of Immunology 2006, 176, (8), 4666- 4674. 281 39. 40. 41. 42. 43. 44. 45. 46. 47. Vermeulen, L.; De Wilde, G.; Notebaert, S.; Vanden Berghe, W.; Haegeman, G. Regulation of the transcriptional activity of the nuclear factor-kappa B p65 subunit. Biochemical Pharmacology 2002, 64, (5-6), 963-70. Huber, M. A.; Azoitei, N.; Baumann, B.; Grunert, S.; Sommer, A.; Pehamberger, H.; Kraut, N.; Beug, H.; Wirth, T. NF-kappa B is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. Journal of Clinical Investigation 2004, 114, (4), 569- 581. El-Rayes, B. F.; Ali, 8.; Ali, I. F.; Philip, P. A.; Abbruzzese, J.; Sarkar, F. H. Potentiation of the effect of erlotinib by genistein in pancreatic cancer: The role of AKT and nuclear factor-kappa B. Cancer Research 2006, 66, (21), 10553-10559. Ross, J. S.; Kallakury, B. V. S.; Sheehan, C. E.; Fisher, H. A. G.; Kaufman, R. P.; Kaur, P.; Gray, K.; Stringer, B. Expression of nuclear factor-kappa B and l kappa Ba proteins in prostatic adenocarcinomas: Correlation of nuclear factor-kappa b immunoreactivity with disease recurrence. Clinical Cancer Research 2004, 10, (7), 2466-2472. Sasaki, N.; Morisaki, T.; Hashizume, K.; Yao, T.; Tsuneyoshi, M.; Noshiro, H., et al. Nuclear factor-kappa B p65 (reIA) transcription factor is constitutively activated in human gastric carcinoma tissue. Clinical Cancer Research 2001, 7, (12), 4136-4142. Karin, M.; Lin, A. NF-kappa B at the crossroads of life and death. Nature Immunology 2002, 3, (3), 221-227. Wang, C. Y.; Mayo, M. W.; Baldwin, A. S. TNF- and cancer therapy- induced apoptosis: Potentiation by inhibition of NF-kappa B. Science 1996, 274, (5288), 784-787. Mayo, M. W.; Baldwin, A. S. The transcription factor NF-kappa B: Control of oncogenesis and cancer therapy resistance. Biochimica Et Biophysica Acta-Reviews on Cancer 2000, 1470, (2), M55-M62. Wang, C. Y.; Mayo, M. W.; Komeluk, R. G.; Goeddel, D. V.; Baldwin, A. S. NF-kappa B antiapoptosis: Induction of traf1 and traf2 and c-iap1 and c- iap2 to suppress caspase-8 activation. Science 1998, 281, (5383), 1680- 1683. 282 48. 49. 50. 51. 52. 53. 55. 56. Cusack, J. C.; Liu, R.; Baldwin, A. S. NF-kappa B and chemoresistance: Potentiation of cancer chemotherapy via inhibition of nf-kappa b. Drug Resistance Updates 1999, 2, (4), 271-273. Sharma, V.; Peddibhotla, S.; Tepe, J. J. Sensitization of cancer cells to DNA damaging agents by imidazolines. Journal of the American Chemical Society 2006, 128, (28), 9137-9143. Jones, D. R.; Broad, R. M.; Madrid, L. V.; Baldwin, A. 8.; Mayo, M. W. Inhibition of NF-kappa B sensitizes non-small cell lung cancer cells to chemotherapy-induced apoptosis. Annals of Thoracic Surgery 2000, 70, (3), 930-936. Cusack, J. C.; Liu, R.; Baldwin, A. S. Inducible chemoresistance to 7- ethyl-10-[4-(1-piperidino)-1-piperidlno]carbonyloxycamptothecin (opt-11) in colorectal cancer cells and a xenograft model is overcome by inhibition of nuclear factor-kappa B activation. Cancer Research 2000, 60, (9), 2323- 2330. Sharma, V.; Hqu, C. D.; Tepe, J. J. Enhancement of chemotherapeutic efficacy by small molecule inhibition of NF-kappa B and checkpoint kinases. Current Medicinal Chemistry 2007, 14, (10), 1061-1074. Chan, M. M.; Fong, D.; Soprano, K. J.; Holmes, W. F.; Heverling, H. Inhibition of growth and sensitization to Cisplatin-mediated killing of ovarian cancer cells by polyphenolic chemopreventive agents. Journal of Cellular Physiology 2003, 194, (1 ), 63-70. Bava, S. V.; Puliappadamba, V. T.; Deepti, A.; Nair, A.; Karunagaran, D.; Anto, R. J. Sensitization of taxol-induced apoptosis by curcumin involves down-regulation of nuclear factor-kappa b and the serine/threonine kinase AKT and is independent of tubulin polymerization. Joumal of Biological Chemistry 2005, 280, (8), 6301-6308. Deeb, D.; Jiang, H.; Gao, X. H.; Hafner, M. S.; Wong, H.; Divine, G.; Chapman, R. A.; Dulchavsky, S. A.; Gautam, S. C. Curcumin sensitizes prostate cancer cells to tumor necrosis factor-related apoptosis-inducing ligand/ap02l by inhibiting nuclear factor-kappa b through suppression of | kappa Ba phosphorylation. Molecular Cancer Therapeutics 2004, 3, (7), 803-812. Richardson, P. G.; Mitsiades, C.; Hideshima, T.; Anderson, K. C. Proteasome inhibition in the treatment of cancer. Cell Cycle 2005, 4, (2), 290-296. 283 57. 58. 59. 60. 61. 62. 63. 65. Montagut, C.; Rovira, A.; Mellado, B.; Gascon, P.; Ross, J. S.; Albanell, J. Preclinical and clinical development of the proteasome inhibitor bortezomib in cancer treatment. Drugs of Today 2005, 41, (5), 299-315. Sors, A.; Jean-Louis, F.; Pellet, C.; Laroche, L.; Dubertret, L.; Courtois, G.; Bachelez, H.; Michel, L. Down-regulating constitutive activation of the NF- kappa B canonical pathway overcomes the resistance of cutaneous T-cell lymphoma to apoptosis. Blood 2006, 107, (6), 2354-2363. Yunmbam, M. K.; Li, O. D. 0.; Mimnaugh, E. G.; Kayastha, G. L.; Yu, J. J.; Jones, L. N.; Neckers, L.; Reed, E. Effect of the proteasome inhibitor ALLN on cisplatin sensitivity in human ovarian tumor cells. lntemational Journal of Oncology 2001, 19, (4), 741 -748. Shirley, R. B.; Kaddour—Djebbar, l.; Patel, D. M.; Lakshmikanthan, V.; Lewis, R. W.; Kumar, M. V. Combination of proteasomal inhibitors lactacystin and MG132 induced synergistic apoptosis in prostate cancer cells. Neoplasia 2005, 7, (12), 1104-1111. Munshi, A.; Kurland, J. F.; Nishikawa, T.; Chiao, P. J.; Andreeff, M.; Meyn, R. E. Inhibition of constitutively activated nuclear factor-kappa b radiosensitizes human melanoma cells. Molecular Cancer Therapeutics 2004, 3, (8), 985-992. Cory, A. H.; Cory, J. G. Lactacystin, a proteasome inhibitor, potentiates the apoptotic effect of parthenolide, an inhibitor of NF kappa B activation, on drug-resistant mouse leukemia I1210 cells. Anticancer Research 2002, 22, (6C), 3805-3809. Mimnaugh, E. G.; Yunmbam, M. K.; Li, O. D.; Bonvini, P.; Hwang, S. G.; Trepel, J.; Reed, E.; Neckers, L. Prevention of cisplatin-DNA adduct repair and potentiation of cisplatin-induced apoptosis in ovarian carcinoma cells by proteasome inhibitors. Biochemical Pharmacology 2000, 60, (9), 1343- 1354. Hu, J.; Nakano, H.; Sakurai, H.; Colbum, N. H. Insufficient p65 phosphorylation at $536 specifically contributes to the lack of NF-kappa B activation and transformation in resistant jb6 cells. Carcinogenesis 2004, 25, (10), 1991-2003. Greten, F. R.; Eckmann, L.; Greten, T. F.; Park, J. M.; Li, Z. W.; Egan, L. J.; Kagnoff, M. F.; Karin, M. IKK beta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 2004, 118, (3), 285- 296. 284 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. Agganrval, B. B.; Sethi, G.; Nair, A.; Ichikawa, H. Nuclear factor-kappa b: A holy grail in cancer prevention and therapy. Current Signal Transduction Therapy 2006, 1, 25-52. Schmitz, M. L.; Mattioli, I.; Buss, H.; Kracht, M. NF-kB: A multifaceted transcription factor regulated at several levels. Chembiochem 2004, 5, 1348-1358. Verstrepen, L.; Bekaert, T.; Chau, T.-L.; Tavemier, J.; Chariot, A.; Beyaert, R. TrI-4, ll-1r and TNF-r signaling to NF-kB: Variations on a common theme. Cell and Molecular Life Sciences 2008, 65, 2964-2978. Brown, K. D.; Claudio, E.; Siebenlist, U. The roles of the classical and alternative nuclear factor-kB pathways: Potential implications for autoimmunity and rheumatoid arthritis. Arthritis Research & Therapy 2008, 10, (4), 212-226. Jin, X. 0.; Wang, Z. M.; Qiu, L.; Zhang, D. L.; Guo, Z. H.; Gao, Z. W., et al. Potential biomarkers involving lKK/relA signal in early stage non-small cell lung cancer. Cancer Science 2008, 99, (3), 582-589. Zhang, 2. F.; Ma, J. 0.; Li, N.; Sun, M; Wang, C. L. Expression of nuclear factor-kappa b and its clinical significance in nonsmalI-cell lung cancer. Annals of Thoracic Surgery 2006, 82, (1 ), 243-248. Tang, X. M.; Liu, D. N.; Shishodia, S.; Ozbum, N.; Behrens, C.; Lee, J. J.; Hong, W. K.; Aggarwal, B. B.; Wistuba, l. l. Nuclear factor-kappa b (NF- kappa b) is frequently expressed in lung cancer and preneoplastic lesions. Cancer 2006, 107, (1 1), 2637-2646. Van Waes, C. Nuclear factor-kappa b in development, prevention, and therapy of cancer. Clinical Cancer Research 2007, 13, (4), 1076-1082. Cantin, G. T.; Venable, J. D.; Cociorva, D.; Yates, J. R. Quantitative phosphoproteomic analysis of the tumor necrosis factor pathway. Journal of Proteome Research 2006, 5, (1 ), 127-134. Rubio, M. F.; Werbajh, S.; Cafferata, E. G. A.; Quaglino, A.; Colo, G. P.; Nojek, I. M.; Kordon, E. C.; Nahmod, V. E.; Costas, M. A. TNF-a enhances estrogen-induced cell proliferation of estrogen-dependent breast tumor cells through a complex containing nuclear factor-kappa b. Oncogene 2006, 25, (9), 1367-1377. 285 76. 77. 78. 79. 80. 81. 82. 83. 84. Link, A. J.; Eng, J.; Schieltz, D. M.; Carrnack, E.; Mize, G. J.; Morris, D. R.; Garvik, B. M.; Yates, J. R. Direct analysis of protein complexes using mass spectrometry. Nature Biotechnology 1999, 17, (7), 676-682. Zhong, H. H.; SuYang, H.; ErdjumentBromage, H.; Tempst, P.; Ghosh, S. The transcriptional activity of NF-kappa B is regulated by the l kappa B- associated PKAc subunit through a cyclic amp-independent mechanism. Cal/1997, 89, (3), 413-424. Bohuslav, J.; Chen, L. F.; Kwon, H.; Mu, Y. J.; Greene, W. C. P53 induces NF-kappa B activation by an I kappa B kinase-independent mechanism involving phosphorylation of p65 by ribosomal s6 kinase 1. Journal of Biological Chemistry 2004, 279, (25), 26115-26125. Guan, H. C.; Hou, S. H.; Ricciardi, R. P. DNA binding of repressor nuclear factor-kappa b p50/p50 depends on phosphorylation of ser(337) by the protein kinase a catalytic subunit. Journal of Biological Chemistry 2005, 280, (11), 9957-9962. Chen, Z. J.; Parent, L.; Maniatis, T. Site-specific phosphorylation of | kappa B a by a novel ubiquitination—dependent protein kinase activity. Cell 1996, 84, (6), 853-862. Perfettini, J.-L.; Roumier, T.; Castego, M.; Larochette, N.; Boya, P.; Raynal, B., et al. Nf-kB and p53 are the dominant apoptosis-inducing transcription factors elicited by the hiv-1 envelope. Journal of Experimental Medicine 2004, 199, (5), 629-640. Ficarro, S. B.; McCleland, M. L.; Stukenberg, P. T.; Burke, D. J.; Ross, M. M.; Shabanowitz, J.; Hunt, D. F.; White, F. M. Phosphoproteome analysis by mass spectrometry and its application to saccharomyces cerevisiae. Nature Biotechnology 2002, 20, (3), 301-305. Steinberg, T. H.; Agnew, B. J.; Gee, K. R.; Leung, W. Y.; Goodman, T.; Schulenberg, B., et al. Global quantitative phosphoprotein analysis using multiplexed proteomics technology. Proteomics 2003, 3, (7), 1128—1144. Stasyk, T.; Morandell, S.; Bakry, R.; Feuerstein, l.; Huck, C. W.; Stecher, G.; Bonn, G. K.; Huber, L. A. Quantitative detection of phosphoproteins by combination of two-dimensional difference gel electrophoresis and phosphospecific fluorescent staining. Electrophoresis 2005, 26, (14), 2850-2854. 286 85. 86. 87. 88. 89. 90. 91. 92. 93. Schulenberg, B.; Aggeler, R.; Beechem, J. M.; Capaldi, R. A.; Patton, W. F. Analysis of steady-state protein phosphorylation in mitochondria using a novel fluorescent phosphosensor dye. Journal of Biological Chemistry 2003, 278, (29), 27251-27255. Oda, Y.; Nagasu, T.; Chait, B. T. Enrichment analysis of phosphorylated proteins as a tool for probing the phosphoproteome. Nature Biotechnology 2001, 19, (4), 379—382. van der Veken, P.; Dirksen, E. H. C.; Ruijter, E.; Elgersma, R. C.; Heck, A. J. R.; Rijkers, D. T. S.; Slijper, M.; Liskamp, R. M. J. Development of a novel chemical probe for the selective enrichment of phosphorylated serine- and threonine-containing peptides. Chembiochem 2005, 6, (12), 2271-2280. Bourget, C.; Trevisiol, E.; Bridon, l.; Kotera, M.; Lhomme, J.; Laayoun, A. Biotin-phenyldiazomethane conjugates as labeling reagents at phosphate in mono and polynucleotides. Bioorganic & Medicinal Chemistry 2005, 13, (5), 1453-1461. Chowdhury, S. M.; Munske, G. R.; Siems, W. F.; Bruce, J. E. A new maleimide-bound acid-cleavable solid-support reagent for profiling phosphorylation. Rapid Communications in Mass Spectrometry 2005, 19, (7), 899-909. Lee, S. H.; Clapham, B.; Koch, G.; Zimmermann, J.; Janda, K. D. Rhodium carbenoid N-H insertion reactions of primary ureas: Solution and solid-phase synthesis of imidazolones. Organic Letters 2003, 5, (4), 511- 514. Paez, J. G.; Janne, P. A.; Lee, J. C.; Tracy, S.; Greulich, H.; Gabriel, 8., et al. EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science 2004, 304, (5676), 1497-1500. Matsushita, H.; Lee, S. H.; Yoshida, K.; Clapham, B.; Koch, G.; Zimmermann, J.; Janda, K. D. N-H insertion reactions of Boo-amino acid amides: Solution— and solid-phase synthesis of pyrazinones and pyrazines. Organic Letters 2004, 6, (24), 4627-4629. Yamashita, M.; Lee, S. H.; Koch, G.; Zimmermann, J.; Clapham, B.; Janda, K. D. Solid-phase synthesis of oxazolones and other heterocycles via Wang resin-bound diazocarbonyls. Tetrahedron Letters 2005, 46, (33), 5495-5498. 287 IIIIIII'I'I'IIII I I ' I «I