Post-translational Modifications in the Cardiovascular Proteome

The analysis of post-translational modifications is critical for understanding the regulation of protein function in the heart. These small, often charged, groups are added to a protein’s structure to modulate its activity, localization or associations. The development of proteomic technologies has greatly improved the identification and subsequent characterization of these modifications. However, due to the complex nature of the cardiovascular proteome, a particular post-translational modification may represent only a tiny fraction of the milieu. Additionally, some modifications are too labile for mass spectrometry analysis. To address these limitations, a variety of techniques and strategies have been developed to specifically target and improve the detection of these modifications. In the following chapter, we review the challenges and solutions to identifying several prominent post-translational modifications in the cardiovascular system.

[1]  M. Mann,et al.  Proteomic analysis of post-translational modifications , 2003, Nature Biotechnology.

[2]  Chang Chen,et al.  An ascorbate-dependent artifact that interferes with the interpretation of the biotin switch assay. , 2006, Free Radical Biology & Medicine.

[3]  S. Rhee,et al.  Regulation of reactive oxygen species generation in cell signaling , 2011, Molecules and cells.

[4]  M. Mann,et al.  Global and site-specific quantitative phosphoproteomics: principles and applications. , 2009, Annual review of pharmacology and toxicology.

[5]  Steven P Gygi,et al.  The SCX/IMAC enrichment approach for global phosphorylation analysis by mass spectrometry , 2008, Nature Protocols.

[6]  Lance Wells,et al.  Mapping Sites of O-GlcNAc Modification Using Affinity Tags for Serine and Threonine Post-translational Modifications* , 2002, Molecular & Cellular Proteomics.

[7]  Yingming Zhao,et al.  Modification‐specific proteomics: Strategies for characterization of post‐translational modifications using enrichment techniques , 2009, Proteomics.

[8]  M. Ferlito,et al.  S-nitrosylation of TRIM72 at cysteine 144 is critical for protection against oxidation-induced protein degradation and cell death. , 2014, Journal of molecular and cellular cardiology.

[9]  Zhike Lu,et al.  Identification of 67 Histone Marks and Histone Lysine Crotonylation as a New Type of Histone Modification , 2011, Cell.

[10]  S. Gygi,et al.  Quantitative analysis of complex protein mixtures using isotope-coded affinity tags , 1999, Nature Biotechnology.

[11]  Goedele Roos,et al.  Protein sulfenic acid formation: from cellular damage to redox regulation. , 2011, Free radical biology & medicine.

[12]  Pierre Thibault,et al.  The Coming of Age of Phosphoproteomics—from Large Data Sets to Inference of Protein Functions* , 2013, Molecular & Cellular Proteomics.

[13]  T. Finkel,et al.  Cellular mechanisms and physiological consequences of redox-dependent signalling , 2014, Nature Reviews Molecular Cell Biology.

[14]  K. Mechtler,et al.  Application of different fragmentation techniques for the analysis of phosphopeptides using a hybrid linear ion trap-FTICR mass spectrometer. , 2006, Biochimica et biophysica acta.

[15]  Matthew E Monroe,et al.  Tandem mass spectrometry identifies many mouse brain O-GlcNAcylated proteins including EGF domain-specific O-GlcNAc transferase targets , 2012, Proceedings of the National Academy of Sciences.

[16]  R. Annan,et al.  Hydrophilic Interaction Chromatography Reduces the Complexity of the Phosphoproteome and Improves Global Phosphopeptide Isolation and Detection*S , 2008, Molecular & Cellular Proteomics.

[17]  A. Ross,et al.  Enrichment and analysis of phosphopeptides under different experimental conditions using titanium dioxide affinity chromatography and mass spectrometry. , 2010, Rapid communications in mass spectrometry : RCM.

[18]  Tong Liu,et al.  A strategy for direct identification of protein S-nitrosylation sites by quadrupole time-of-flight mass spectrometry , 2008, Journal of the American Society for Mass Spectrometry.

[19]  J. Stamler,et al.  NO/redox disequilibrium in the failing heart and cardiovascular system. , 2005, The Journal of clinical investigation.

[20]  M. Collins,et al.  Analysis of protein phosphorylation on a proteome‐scale , 2007, Proteomics.

[21]  Lance Wells,et al.  Combining high-energy C-trap dissociation and electron transfer dissociation for protein O-GlcNAc modification site assignment. , 2011, Journal of proteome research.

[22]  Antonio Malgaroli,et al.  A novel approach to identify proteins modified by nitric oxide: the HIS-TAG switch method. , 2007, Journal of proteome research.

[23]  A. van Dorsselaer,et al.  A method for detection of overoxidation of cysteines: peroxiredoxins are oxidized in vivo at the active-site cysteine during oxidative stress. , 2002, The Biochemical journal.

[24]  L. Landino,et al.  Ascorbic acid reduction of microtubule protein disulfides and its relevance to protein S-nitrosylation assays. , 2006, Biochemical and biophysical research communications.

[25]  S. Snyder,et al.  Hydrogen sulfide-linked sulfhydration of NF-κB mediates its antiapoptotic actions. , 2012, Molecular cell.

[26]  G. Hart,et al.  Detection of O-GlcNAc modifications on cardiac myofilament proteins. , 2013, Methods in molecular biology.

[27]  Xinning Jiang,et al.  Classification filtering strategy to improve the coverage and sensitivity of phosphoproteome analysis. , 2010, Analytical chemistry.

[28]  Jason D Hoffert,et al.  Taking aim at shotgun phosphoproteomics. , 2008, Analytical biochemistry.

[29]  R. Wait,et al.  Protein Sulfenation as a Redox Sensor , 2007, Molecular & Cellular Proteomics.

[30]  B K Hayes,et al.  Selective detection and site-analysis of O-GlcNAc-modified glycopeptides by beta-elimination and tandem electrospray mass spectrometry. , 1996, Analytical biochemistry.

[31]  G. McAlister,et al.  Performance Characteristics of Electron Transfer Dissociation Mass Spectrometry*S , 2007, Molecular & Cellular Proteomics.

[32]  R. Cole,et al.  Identification and Quantification of S-Nitrosylation by Cysteine Reactive Tandem Mass Tag Switch Assay* , 2011, Molecular & Cellular Proteomics.

[33]  A. Alpert Hydrophilic-interaction chromatography for the separation of peptides, nucleic acids and other polar compounds. , 1990, Journal of chromatography.

[34]  Kate S. Carroll,et al.  A chemical approach for detecting sulfenic acid-modified proteins in living cells. , 2008, Molecular bioSystems.

[35]  Jonathan D. Barber,et al.  Prophossi: automating expert validation of phosphopeptide–spectrum matches from tandem mass spectrometry , 2010, Bioinform..

[36]  Jonathan C Trinidad,et al.  O-Linked N-Acetylglucosamine Proteomics of Postsynaptic Density Preparations Using Lectin Weak Affinity Chromatography and Mass Spectrometry*S , 2006, Molecular & Cellular Proteomics.

[37]  G. Hart,et al.  Topography and polypeptide distribution of terminal N-acetylglucosamine residues on the surfaces of intact lymphocytes. Evidence for O-linked GlcNAc. , 1984, The Journal of biological chemistry.

[38]  Chunaram Choudhary,et al.  Proteome-wide Analysis of Lysine Acetylation Suggests its Broad Regulatory Scope in Saccharomyces cerevisiae* , 2012, Molecular & Cellular Proteomics.

[39]  J. Stamler,et al.  Detection of protein S-nitrosylation with the biotin-switch technique. , 2009, Free radical biology & medicine.

[40]  Leina Ma,et al.  MAPK/ERK signaling pathway-induced hyper-O-GlcNAcylation enhances cancer malignancy , 2015, Molecular and Cellular Biochemistry.

[41]  Lance Wells,et al.  Quantitative analysis of both protein expression and serine / threonine post‐translational modifications through stable isotope labeling with dithiothreitol , 2005, Proteomics.

[42]  A. Burlingame,et al.  Electron transfer dissociation (ETD): The mass spectrometric breakthrough essential for O‐GlcNAc protein site assignments—a study of the O‐GlcNAcylated protein Host Cell Factor C1 , 2013, Proteomics.

[43]  Kenneth F Reardon,et al.  Phosphoproteomics and molecular cardiology: techniques, applications and challenges. , 2012, Journal of molecular and cellular cardiology.

[44]  Guo-Ping Zhao,et al.  Acetylation of Metabolic Enzymes Coordinates Carbon Source Utilization and Metabolic Flux , 2010, Science.

[45]  Brian O'Rourke,et al.  Redox Regulation of Mitochondrial ATP Synthase: Implications for Cardiac Resynchronization Therapy , 2011, Circulation Research.

[46]  N. Dhalla,et al.  Evidence for the role of oxidative stress in acute ischemic heart disease: a brief review. , 1999, The Canadian journal of cardiology.

[47]  J. Gorman,et al.  Protein disulfide bond determination by mass spectrometry. , 2002, Mass spectrometry reviews.

[48]  Gerald W. Hart,et al.  O-GlcNAc signalling: implications for cancer cell biology , 2011, Nature Reviews Cancer.

[49]  Yingming Zhao,et al.  Lysine glutarylation is a protein posttranslational modification regulated by SIRT5. , 2014, Cell metabolism.

[50]  J. V. Van Eyk,et al.  Evidence of myofibrillar protein oxidation induced by postischemic reperfusion in isolated rat hearts. , 2004, American journal of physiology. Heart and circulatory physiology.

[51]  P. Roepstorff,et al.  Highly Selective Enrichment of Phosphorylated Peptides from Peptide Mixtures Using Titanium Dioxide Microcolumns* , 2005, Molecular & Cellular Proteomics.

[52]  T. Issad,et al.  O-GlcNAc modification, insulin signaling and diabetic complications. , 2010, Diabetes & metabolism.

[53]  R. Cohen,et al.  Isotope-coded affinity tag (ICAT) approach to redox proteomics: identification and quantitation of oxidant-sensitive cysteine thiols in complex protein mixtures. , 2004, Journal of proteome research.

[54]  Sarah B. Scruggs,et al.  Myocardial infarction in mice alters sarcomeric function via post-translational protein modification , 2012, Molecular and Cellular Biochemistry.

[55]  A. Burlingame,et al.  Identification of Novel Sites of O-N-Acetylglucosamine Modification of Serum Response Factor Using Quadrupole Time-of-flight Mass Spectrometry* , 2003, Molecular & Cellular Proteomics.

[56]  A. Burlingame,et al.  Global Identification and Characterization of Both O-GlcNAcylation and Phosphorylation at the Murine Synapse* , 2012, Molecular & Cellular Proteomics.

[57]  R. Traut,et al.  Diagonal polyacrylamide-dodecyl sulfate gel electrophoresis for the identification of ribosomal proteins crosslinked with methyl-4-mercaptobutyrimidate. , 1974, Proceedings of the National Academy of Sciences of the United States of America.

[58]  T. Pawson,et al.  Reading protein modifications with interaction domains , 2006, Nature Reviews Molecular Cell Biology.

[59]  Forest M White,et al.  Quantitative phosphoproteomics by mass spectrometry: Past, present, and future , 2008, Proteomics.

[60]  A. Heck,et al.  Selective isolation at the femtomole level of phosphopeptides from proteolytic digests using 2D-NanoLC-ESI-MS/MS and titanium oxide precolumns. , 2004, Analytical chemistry.

[61]  M. Wolfert,et al.  Glycopeptide-specific monoclonal antibodies suggest new roles for O-GlcNAc. , 2010, Nature chemical biology.

[62]  M. Gucek,et al.  Disruption of caveolae blocks ischemic preconditioning-mediated S-nitrosylation of mitochondrial proteins. , 2012, Antioxidants & redox signaling.

[63]  Solomon H. Snyder,et al.  The Biotin Switch Method for the Detection of S-Nitrosylated Proteins , 2001, Science's STKE.

[64]  J. Stamler,et al.  Assessment and Application of the Biotin Switch Technique for Examining Protein S-Nitrosylation under Conditions of Pharmacologically Induced Oxidative Stress* , 2007, Journal of Biological Chemistry.

[65]  Akira Sano,et al.  Titania as a Chemo-affinity Support for the Column-switching HPLC Analysis of Phosphopeptides: Application to the Characterization of Phosphorylation Sites in Proteins by Combination with Protease Digestion and Electrospray Ionization Mass Spectrometry , 2004, Analytical sciences : the international journal of the Japan Society for Analytical Chemistry.

[66]  G. Davies,et al.  A potent mechanism-inspired O-GlcNAcase inhibitor that blocks phosphorylation of tau in vivo. , 2008, Nature chemical biology.

[67]  Junfeng Ma,et al.  O-GlcNAc profiling: from proteins to proteomes , 2014, Clinical Proteomics.

[68]  A. Mencalha,et al.  Post-translational modifications disclose a dual role for redox stress in cardiovascular pathophysiology. , 2015, Life sciences.

[69]  H. Zou,et al.  Enrichment and separation techniques for large-scale proteomics analysis of the protein post-translational modifications. , 2014, Journal of chromatography. A.

[70]  B. Chait,et al.  Monitoring Reactions of Nitric Oxide with Peptides and Proteins by Electrospray Ionization-Mass Spectrometry (*) , 1995, The Journal of Biological Chemistry.

[71]  Tomas Bergman,et al.  Determination of site-specificity of S-glutathionylated cellular proteins. , 2005, Biochemical and biophysical research communications.

[72]  Albert Sickmann,et al.  The good, the bad, the ugly: Validating the mass spectrometric analysis of modified peptides , 2011, Proteomics.

[73]  J. Sadoshima,et al.  Redox modification of cell signaling in the cardiovascular system. , 2012, Journal of molecular and cellular cardiology.

[74]  Johan Auwerx,et al.  Sirt5 Is a NAD-Dependent Protein Lysine Demalonylase and Desuccinylase , 2011, Science.

[75]  K. Khoo,et al.  Cysteine S-Nitrosylation Protects Protein-tyrosine Phosphatase 1B against Oxidation-induced Permanent Inactivation* , 2008, Journal of Biological Chemistry.

[76]  A. Holmgren,et al.  Identification of S-glutathionylated cellular proteins during oxidative stress and constitutive metabolism by affinity purification and proteomic analysis. , 2002, Archives of biochemistry and biophysics.

[77]  P. Eaton,et al.  Redox regulation of cGMP-dependent protein kinase Iα in the cardiovascular system , 2015, Front. Pharmacol..

[78]  Pál Pacher,et al.  Interplay of oxidative, nitrosative/nitrative stress, inflammation, cell death and autophagy in diabetic cardiomyopathy. , 2015, Biochimica et biophysica acta.

[79]  D. Giustarini,et al.  Is ascorbate able to reduce disulfide bridges? A cautionary note. , 2008, Nitric oxide : biology and chemistry.

[80]  M. Mann,et al.  Global, In Vivo, and Site-Specific Phosphorylation Dynamics in Signaling Networks , 2006, Cell.

[81]  G. Hart,et al.  O-Linked GlcNAc Modification of Cardiac Myofilament Proteins: A Novel Regulator of Myocardial Contractile Function , 2008, Circulation research.

[82]  A. Saurin,et al.  Widespread sulfenic acid formation in tissues in response to hydrogen peroxide , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[83]  Steven P Jones,et al.  Cardioprotection by N-Acetylglucosamine Linkage to Cellular Proteins , 2008, Circulation.

[84]  R. Erbel,et al.  Oxidative modification of tropomyosin and myocardial dysfunction following coronary microembolization. , 2006, European heart journal.

[85]  Guanghui Wang,et al.  Simultaneous Measurement of Protein Oxidation and S-Nitrosylation During Preconditioning and Ischemia/Reperfusion Injury With Resin-Assisted Capture , 2011, Circulation research.

[86]  B. Kuster,et al.  Proteome Wide Purification and Identification of O‑glcnac-modified Proteins Using Click Chemistry and Mass Spectrometry , 2022 .

[87]  Guanghui Wang,et al.  Characterization of potential S-nitrosylation sites in the myocardium. , 2011, American journal of physiology. Heart and circulatory physiology.

[88]  E. Niki,et al.  Effective detection of peptides containing cysteine sulfonic acid using matrix-assisted laser desorption/ionization and laser desorption/ionization on porous silicon mass spectrometry. , 2006, Journal of mass spectrometry : JMS.

[89]  M. Gladwin,et al.  Role of the anion nitrite in ischemia-reperfusion cytoprotection and therapeutics. , 2007, Cardiovascular research.

[90]  A. Hershko,et al.  Role of the alpha-amino group of protein in ubiquitin-mediated protein breakdown. , 1984, Proceedings of the National Academy of Sciences of the United States of America.

[91]  Robert J Chalkley,et al.  Identification of protein O-GlcNAcylation sites using electron transfer dissociation mass spectrometry on native peptides , 2009, Proceedings of the National Academy of Sciences.

[92]  M. Horsch,et al.  N‐Acetylgucosaminono‐1,5‐lactone oxime and the corresponding (phenylcarbamoyl)oxime , 1991 .

[93]  R. Cohen,et al.  Isotope-coded Affinity Tag Approach to Identify and Quantify Oxidant-sensitive Protein Thiols* , 2004, Molecular & Cellular Proteomics.

[94]  Nick V Grishin,et al.  Lysine Acetylation Is a Highly Abundant and Evolutionarily Conserved Modification in Escherichia Coli*S , 2009, Molecular & Cellular Proteomics.

[95]  B. Buchanan,et al.  Redox regulation: a broadening horizon. , 2005, Annual review of plant biology.

[96]  T. Finkel,et al.  Signal transduction by reactive oxygen species , 2011, The Journal of cell biology.

[97]  Mark A. Wilson,et al.  The Parkinson's disease protein DJ-1 is neuroprotective due to cysteine-sulfinic acid-driven mitochondrial localization , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[98]  J. Shabanowitz,et al.  O-Linked β-N-Acetylglucosamine (O-GlcNAc) Regulates Emerin Binding to Barrier to Autointegration Factor (BAF) in a Chromatin- and Lamin B-enriched “Niche”* , 2013, The Journal of Biological Chemistry.

[99]  K. Khoo,et al.  Mass spectrometry-based quantitative proteomics for dissecting multiplexed redox cysteine modifications in nitric oxide-protected cardiomyocyte under hypoxia. , 2014, Antioxidants & redox signaling.

[100]  Perry Evans,et al.  Site-Specific Proteomic Mapping Identifies Selectively Modified Regulatory Cysteine Residues in Functionally Distinct Protein Networks. , 2015, Chemistry & biology.

[101]  J. Porath,et al.  Isolation of phosphoproteins by immobilized metal (Fe3+) affinity chromatography. , 1986, Analytical biochemistry.

[102]  Lance Wells,et al.  Multiple Tissue-specific Roles for the O-GlcNAc Post-translational Modification in the Induction of and Complications Arising from Type II Diabetes* , 2014, The Journal of Biological Chemistry.

[103]  Carolyn R Bertozzi,et al.  A chemical approach for identifying O-GlcNAc-modified proteins in cells , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[104]  Melanie Y. White,et al.  Functional decorations: post-translational modifications and heart disease delineated by targeted proteomics , 2013, Genome Medicine.

[105]  J. Shabanowitz,et al.  Peptide and protein sequence analysis by electron transfer dissociation mass spectrometry. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[106]  E. Carafoli,et al.  Nature and site of phospholamban regulation of the Ca2+ pump of sarcoplasmic reticulum , 1989, Nature.

[107]  Chunaram Choudhary,et al.  Proteome-Wide Mapping of the Drosophila Acetylome Demonstrates a High Degree of Conservation of Lysine Acetylation , 2011, Science Signaling.

[108]  Vidya Venkatraman,et al.  Cysteine oxidative posttranslational modifications: emerging regulation in the cardiovascular system. , 2013, Circulation research.

[109]  Yi Tang,et al.  Lysine Propionylation and Butyrylation Are Novel Post-translational Modifications in Histones*S , 2007, Molecular & Cellular Proteomics.

[110]  F. Di Lisa,et al.  Oxidation of myofibrillar proteins in human heart failure. , 2011, Journal of the American College of Cardiology.

[111]  Elizabeth Murphy,et al.  Estrogen Receptor-&bgr; Activation Results in S-Nitrosylation of Proteins Involved in Cardioprotection , 2009, Circulation.

[112]  Melanie Y. White,et al.  The role of post‐translational modifications in acute and chronic cardiovascular disease , 2014, Proteomics. Clinical applications.

[113]  D. Phillips The presence of acetyl groups of histones. , 1963, The Biochemical journal.

[114]  Sheng‐yu Huang,et al.  Mass spectrometry-based strategies for protein disulfide bond identification , 2013 .

[115]  J. Hanover,et al.  O-GlcNAc cycling mutants modulate proteotoxicity in Caenorhabditis elegans models of human neurodegenerative diseases , 2012, Proceedings of the National Academy of Sciences.

[116]  L. Jones,et al.  Phospholamban phosphorylation in intact ventricles. Phosphorylation of serine 16 and threonine 17 in response to beta-adrenergic stimulation. , 1989, The Journal of biological chemistry.

[117]  Kate S Carroll,et al.  Mining the thiol proteome for sulfenic acid modifications reveals new targets for oxidation in cells. , 2009, ACS chemical biology.

[118]  Leonardo Nogueira,et al.  Proteomic analysis of S-nitrosylation and denitrosylation by resin-assisted capture , 2009, Nature Biotechnology.

[119]  Mark J. Kohr,et al.  Nitric oxide signaling and the regulation of myocardial function. , 2008, Journal of molecular and cellular cardiology.

[120]  J. R. Brown,et al.  Location of disulphide bridges by diagonal paper electrophoresis. The disulphide bridges of bovine chymotrypsinogen A. , 1966, The Biochemical journal.

[121]  E. Olson,et al.  Control of cardiac growth by histone acetylation/deacetylation. , 2005, Circulation research.

[122]  Suresh Mathivanan,et al.  Global proteomic profiling of phosphopeptides using electron transfer dissociation tandem mass spectrometry , 2007, Proceedings of the National Academy of Sciences.

[123]  J. Zweier,et al.  The role of oxidants and free radicals in reperfusion injury. , 2006, Cardiovascular research.

[124]  R. Marchase,et al.  Glucosamine protects neonatal cardiomyocytes from ischemia-reperfusion injury via increased protein-associated O-GlcNAc. , 2007, American journal of physiology. Cell physiology.

[125]  Zhihong Zhang,et al.  Identification of lysine succinylation as a new post-translational modification. , 2011, Nature chemical biology.

[126]  Rong-Fong Shen,et al.  Preconditioning Results in S-Nitrosylation of Proteins Involved in Regulation of Mitochondrial Energetics and Calcium Transport , 2007, Circulation research.

[127]  P. Eaton,et al.  Gel-based methods in redox proteomics. , 2014, Biochimica et biophysica acta.

[128]  S. Snyder,et al.  H2S Signals Through Protein S-Sulfhydration , 2009, Science Signaling.

[129]  V. Allfrey,et al.  Chemical Studies of Histone Acetylation THE OCCURRENCE OF ε-N-ACETYLLYSINE IN THE f2a1 HISTONE , 1968 .

[130]  A. Weidinger,et al.  Biological Activities of Reactive Oxygen and Nitrogen Species: Oxidative Stress versus Signal Transduction , 2015, Biomolecules.

[131]  S. Guan,et al.  Sulfopeptide fragmentation in electron-capture and electron-transfer dissociation , 2007, Journal of the American Society for Mass Spectrometry.

[132]  H. Zou,et al.  Perspectives of comprehensive phosphoproteome analysis using shotgun strategy. , 2011, Analytical chemistry.

[133]  Jennifer E Van Eyk,et al.  Chasing Cysteine Oxidative Modifications: Proteomic Tools for Characterizing Cysteine Redox Status , 2012, Circulation. Cardiovascular genetics.

[134]  J. Thomson,et al.  Human embryonic stem cell phosphoproteome revealed by electron transfer dissociation tandem mass spectrometry , 2009, Proceedings of the National Academy of Sciences.

[135]  Steven P Jones,et al.  O-GlcNAc Signaling in the Cardiovascular System , 2010, Circulation research.

[136]  D. Vocadlo,et al.  Mapping O-GlcNAc modification sites on tau and generation of a site-specific O-GlcNAc tau antibody , 2011, Amino Acids.

[137]  M. Mortuaire,et al.  Identification of new O-GlcNAc modified proteins using a click-chemistry-based tagging , 2008, Analytical and bioanalytical chemistry.

[138]  J. Hanover,et al.  O-GlcNAc cycling: implications for neurodegenerative disorders. , 2009, The international journal of biochemistry & cell biology.

[139]  J. Michalski,et al.  Mapping of O‐linked β‐N‐acetylglucosamine modification sites in key contractile proteins of rat skeletal muscle , 2009, Proteomics.

[140]  M. Larsen,et al.  Purification and identification of O-GlcNAc-modified peptides using phosphate-based alkyne CLICK chemistry in combination with titanium dioxide chromatography and mass spectrometry. , 2011, Journal of proteome research.

[141]  Roland L Dunbrack,et al.  Structural profiling of endogenous S-nitrosocysteine residues reveals unique features that accommodate diverse mechanisms for protein S-nitrosylation , 2010, Proceedings of the National Academy of Sciences.

[142]  C. Carlson,et al.  Cardiac O-GlcNAc signaling is increased in hypertrophy and heart failure. , 2012, Physiological genomics.

[143]  A. Burlingame,et al.  Polycomb repressive complex 2 is necessary for the normal site-specific O-GlcNAc distribution in mouse embryonic stem cells , 2011, Proceedings of the National Academy of Sciences.

[144]  M. Buse,et al.  O-Linked N-Acetylglucosamine Modification of Insulin Receptor Substrate-1 Occurs in Close Proximity to Multiple SH2 Domain Binding Motifs* , 2009, Molecular & Cellular Proteomics.

[145]  Yi Zhang,et al.  The First Identification of Lysine Malonylation Substrates and Its Regulatory Enzyme* , 2011, Molecular & Cellular Proteomics.

[146]  Marjan Gucek,et al.  Measurement of S-Nitrosylation Occupancy in the Myocardium With Cysteine-Reactive Tandem Mass Tags: Short Communication , 2012, Circulation research.

[147]  Steven P Gygi,et al.  Large-scale characterization of HeLa cell nuclear phosphoproteins. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[148]  James T. Webber,et al.  Online Nanoflow Multidimensional Fractionation for High Efficiency Phosphopeptide Analysis* , 2011, Molecular & Cellular Proteomics.

[149]  Samie R Jaffrey,et al.  Nitrosothiol reactivity profiling identifies S-nitrosylated proteins with unexpected stability. , 2008, Chemistry & biology.

[150]  Steven P Gygi,et al.  The effects of mass accuracy, data acquisition speed, and search algorithm choice on peptide identification rates in phosphoproteomics , 2007, Analytical and bioanalytical chemistry.

[151]  Fabien Campagne,et al.  SNOSID, a proteomic method for identification of cysteine S-nitrosylation sites in complex protein mixtures. , 2006 .

[152]  Steven A Carr,et al.  Integrated proteomic analysis of post-translational modifications by serial enrichment , 2013, Nature Methods.

[153]  Melanie Y. White,et al.  Global Analysis of Myocardial Peptides Containing Cysteines With Irreversible Sulfinic and Sulfonic Acid Post-Translational Modifications* , 2015, Molecular & Cellular Proteomics.

[154]  Kate S Carroll,et al.  Peroxide-dependent sulfenylation of the EGFR catalytic site enhances kinase activity. , 2011, Nature chemical biology.

[155]  A. Salomon,et al.  Isolation of Phosphopeptides by Immobilized Metal Ion Affinity Chromatography , 2007, Current protocols in molecular biology.