Illuminating the dark phosphoproteome

Identifying the targets of “dark” kinases will provide new biological and disease insights. Gloss Protein phosphorylation is a posttranslational modification that regulates protein function. Many biological processes require phosphorylation, and its dysregulation is a hallmark of several complex diseases. Major developments in mass spectrometry now enable the measurement of thousands of changes in phosphorylation and mapping them to exact sites on specific proteins. More than 100,000 phosphorylation sites have been reported, but the kinases regulating these events are currently known only for a small fraction of these sites, and even fewer sites are linked to specific functions. A small subset of kinases dominates the annotated phosphosites, whereas many kinases have no known target proteins. Functional experiments linking human disease genes and mouse knockouts with kinases suggest that these lesser-studied kinases may also be important in health. This Review, with 4 figures, 4 videos, 3 data files, and 205 references, discusses how identifying regulatory kinases and functions of phosphorylated proteins will reveal mechanistic insights into biological function in healthy and disease contexts, point to new therapeutic targets, and enhance our understanding of drug action. Protein phosphorylation is a major regulator of protein function and biological outcomes. This was first recognized through functional biochemical experiments, and in the past decade, major technological advances in mass spectrometry have enabled the study of protein phosphorylation on a global scale. This rapidly growing field of phosphoproteomics has revealed that more than 100,000 distinct phosphorylation events occur in human cells, which likely affect the function of every protein. Phosphoproteomics has improved the understanding of the function of even the most well-characterized protein kinases by revealing new downstream substrates and biology. However, current biochemical and bioinformatic approaches have only identified kinases for less than 5% of the phosphoproteome, and functional assignments of phosphosites are almost negligible. Notably, our understanding of the relationship between kinases and their substrates follows a power law distribution, with almost 90% of phosphorylation sites currently assigned to the top 20% of kinases. In addition, more than 150 kinases do not have a single known substrate. Despite a small group of kinases dominating biomedical research, the number of substrates assigned to a kinase does not correlate with disease relevance as determined by pathogenic human mutation prevalence and mouse model phenotypes. Improving our understanding of the substrates targeted by all kinases and functionally annotating the phosphoproteome will be broadly beneficial. Advances in phosphoproteomics technologies, combined with functional screening approaches, should make it feasible to illuminate the connectivity and functionality of the entire phosphoproteome, providing enormous opportunities for discovering new biology, therapeutic targets, and possibly diagnostics.

[1]  Laura A. Sullivan,et al.  Global Survey of Phosphotyrosine Signaling Identifies Oncogenic Kinases in Lung Cancer , 2007, Cell.

[2]  N. Kelleher,et al.  Top Down proteomics: facts and perspectives. , 2014, Biochemical and biophysical research communications.

[3]  S. Gygi,et al.  Mass spectrometry based method to increase throughput for kinome analyses using ATP probes. , 2013, Analytical chemistry.

[4]  K. Nakayama,et al.  Reconstruction of insulin signal flow from phosphoproteome and metabolome data. , 2014, Cell reports.

[5]  C. Kahn,et al.  Hepatic phosphotyrosine phosphatase activity and its alterations in diabetic rats. , 1989, The Journal of clinical investigation.

[6]  Levi A Garraway,et al.  A Convergence-Based Framework for Cancer Drug Resistance. , 2018, Cancer cell.

[7]  D. Sabatini,et al.  The mTOR-Regulated Phosphoproteome Reveals a Mechanism of mTORC1-Mediated Inhibition of Growth Factor Signaling , 2011, Science.

[8]  J. Chin,et al.  Light-Activated Kinases Enable Temporal Dissection of Signaling Networks in Living Cells , 2011, Journal of the American Chemical Society.

[9]  Matthias Mann,et al.  BoxCar acquisition method enables single-shot proteomics at a depth of 10,000 proteins in 100 minutes , 2018, Nature Methods.

[10]  R. Aebersold A mass spectrometric journey into protein and proteome research , 2003, Journal of the American Society for Mass Spectrometry.

[11]  Gaudenz Danuser,et al.  LOVTRAP, An Optogenetic System for Photo-induced Protein Dissociation , 2016, Nature Methods.

[12]  B. Druker,et al.  Abl protein-tyrosine kinase inhibitor STI571 inhibits in vitro signal transduction mediated by c-kit and platelet-derived growth factor receptors. , 2000, The Journal of pharmacology and experimental therapeutics.

[13]  M. Birnbaum,et al.  Akt1/PKBα Is Required for Normal Growth but Dispensable for Maintenance of Glucose Homeostasis in Mice* , 2001, The Journal of Biological Chemistry.

[14]  Paul J. Wang,et al.  The CREB leucine zipper regulates CREB phosphorylation, cardiomyopathy, and lethality in a transgenic model of heart failure. , 2007, American journal of physiology. Heart and circulatory physiology.

[15]  Karl Mechtler,et al.  Unambiguous Phosphosite Localization using Electron-Transfer/Higher-Energy Collision Dissociation (EThcD) , 2013, Journal of proteome research.

[16]  Sean J. Humphrey,et al.  Protein Phosphorylation: A Major Switch Mechanism for Metabolic Regulation , 2015, Trends in Endocrinology & Metabolism.

[17]  Holger Conzelmann,et al.  Rapid phospho-turnover by receptor tyrosine kinases impacts downstream signaling and drug binding. , 2011, Molecular cell.

[18]  S. Mohammed,et al.  Signal Transduction Reaction Monitoring Deciphers Site-Specific PI3K-mTOR/MAPK Pathway Dynamics in Oncogene-Induced Senescence. , 2015, Journal of proteome research.

[19]  M. Mann,et al.  Kinase-selective enrichment enables quantitative phosphoproteomics of the kinome across the cell cycle. , 2008, Molecular cell.

[20]  M. Mann,et al.  Feasibility of large-scale phosphoproteomics with higher energy collisional dissociation fragmentation. , 2010, Journal of proteome research.

[21]  M. Clausen,et al.  Small-molecule kinase inhibitors: an analysis of FDA-approved drugs. , 2016, Drug discovery today.

[22]  P. Bork,et al.  Linear Motif Atlas for Phosphorylation-Dependent Signaling , 2008, Science Signaling.

[23]  Heiko Horn,et al.  In Vivo Phosphoproteomics Analysis Reveals the Cardiac Targets of β-Adrenergic Receptor Signaling , 2013, Science Signaling.

[24]  K. Shokat,et al.  Engineering Src family protein kinases with unnatural nucleotide specificity. , 1998, Chemistry & biology.

[25]  Christian Stolte,et al.  SnapShot: Insulin/IGF1 Signaling , 2015, Cell.

[26]  M. Mann,et al.  Higher-energy C-trap dissociation for peptide modification analysis , 2007, Nature Methods.

[27]  Bin Fang,et al.  Polypharmacology-based ceritinib repurposing using integrated functional proteomics. , 2017, Nature chemical biology.

[28]  Michael L. Gatza,et al.  Proteogenomics connects somatic mutations to signaling in breast cancer , 2016, Nature.

[29]  P. Beltrão,et al.  Identifications of Putative PKA Substrates with Quantitative Phosphoproteomics and Primary-Sequence-Based Scoring. , 2017, Journal of proteome research.

[30]  M. Mann,et al.  System-wide Perturbation Analysis with Nearly Complete Coverage of the Yeast Proteome by Single-shot Ultra HPLC Runs on a Bench Top Orbitrap* , 2011, Molecular & Cellular Proteomics.

[31]  Sean J Humphrey,et al.  High-throughput phosphoproteomics reveals in vivo insulin signaling dynamics , 2015, Nature Biotechnology.

[32]  James E. Ferrell,et al.  Mechanisms of specificity in protein phosphorylation , 2007, Nature Reviews Molecular Cell Biology.

[33]  A. Burlingame,et al.  Mass Spectrometry-Based Detection and Assignment of Protein Posttranslational Modifications , 2014, ACS chemical biology.

[34]  A. Heck,et al.  The quantitative proteomes of human-induced pluripotent stem cells and embryonic stem cells , 2011, Molecular systems biology.

[35]  A. Heck,et al.  Implementation of Ultraviolet Photodissociation on a Benchtop Q Exactive Mass Spectrometer and Its Application to Phosphoproteomics. , 2016, Analytical chemistry.

[36]  Alma L Burlingame,et al.  A semisynthetic epitope for kinase substrates , 2007, Nature Methods.

[37]  J. Ferrell,et al.  Ultrasensitivity part II: multisite phosphorylation, stoichiometric inhibitors, and positive feedback. , 2014, Trends in biochemical sciences.

[38]  S. Kajimura,et al.  Phosphoproteomics Identifies CK2 as a Negative Regulator of Beige Adipocyte Thermogenesis and Energy Expenditure. , 2015, Cell metabolism.

[39]  P. Beltrão,et al.  Uncovering Phosphorylation-Based Specificities through Functional Interaction Networks* , 2015, Molecular & Cellular Proteomics.

[40]  David E James,et al.  A Positive Feedback Loop between Akt and mTORC2 via SIN1 Phosphorylation. , 2015, Cell reports.

[41]  Matthias Mann,et al.  EASI-tag enables accurate multiplexed and interference-free MS2-based proteome quantification , 2017, bioRxiv.

[42]  Patricia L. Harris,et al.  Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. , 2004, The New England journal of medicine.

[43]  J. Brodbelt,et al.  193 nm Ultraviolet Photodissociation Mass Spectrometry for Phosphopeptide Characterization in the Positive and Negative Ion Modes. , 2016, Journal of proteome research.

[44]  B. Kholodenko,et al.  Signaling switches and bistability arising from multisite phosphorylation in protein kinase cascades , 2004, The Journal of cell biology.

[45]  Bin Fang,et al.  Dissection of TBK1 signaling via phosphoproteomics in lung cancer cells , 2013, Proceedings of the National Academy of Sciences.

[46]  Andrew R. Jones,et al.  ProteomeXchange provides globally co-ordinated proteomics data submission and dissemination , 2014, Nature Biotechnology.

[47]  Ruedi Aebersold,et al.  Dual Specificity Kinase DYRK3 Couples Stress Granule Condensation/Dissolution to mTORC1 Signaling , 2013, Cell.

[48]  P. Strålfors,et al.  Attenuation of Insulin-stimulated Insulin Receptor Substrate-1 Serine 307 Phosphorylation in Insulin Resistance of Type 2 Diabetes* , 2005, Journal of Biological Chemistry.

[49]  Matthias Mann,et al.  Phosphoproteomics reveals that Parkinson's disease kinase LRRK2 regulates a subset of Rab GTPases , 2016, eLife.

[50]  Tomohiro Suzuki,et al.  Forward-genetics analysis of sleep in randomly mutagenized mice , 2016, Nature.

[51]  Stefan Knapp,et al.  Recently targeted kinases and their inhibitors-the path to clinical trials. , 2014, Current opinion in pharmacology.

[52]  Martin Kircher,et al.  Deep proteome and transcriptome mapping of a human cancer cell line , 2011, Molecular systems biology.

[53]  B. Kuster,et al.  Phosphotyrosine mediated protein interactions of the discoidin domain receptor 1. , 2012, Journal of proteomics.

[54]  D E Koshland,et al.  Energy expenditure in the control of biochemical systems by covalent modification. , 1987, The Journal of biological chemistry.

[55]  Bin Zhang,et al.  PhosphoSitePlus, 2014: mutations, PTMs and recalibrations , 2014, Nucleic Acids Res..

[56]  David E James,et al.  Functional screening in human cardiac organoids reveals a metabolic mechanism for cardiomyocyte cell cycle arrest , 2017, Proceedings of the National Academy of Sciences.

[57]  I. Lax,et al.  The Selectivity of Receptor Tyrosine Kinase Signaling Is Controlled by a Secondary SH2 Domain Binding Site , 2009, Cell.

[58]  C. Landry,et al.  Weak functional constraints on phosphoproteomes. , 2009, Trends in genetics : TIG.

[59]  Albert J R Heck,et al.  PhosphoPath: Visualization of Phosphosite-centric Dynamics in Temporal Molecular Networks. , 2015, Journal of proteome research.

[60]  Chin-Rang Yang,et al.  Systems-level identification of PKA-dependent signaling in epithelial cells , 2017, Proceedings of the National Academy of Sciences.

[61]  David E. James,et al.  Dynamic Adipocyte Phosphoproteome Reveals that Akt Directly Regulates mTORC2 , 2013, Cell metabolism.

[62]  Matthew E Berginski,et al.  Coral: Clear and Customizable Visualization of Human Kinome Data. , 2018, Cell systems.

[63]  J. Fitzgibbon,et al.  Proteomic and genomic integration identifies kinase and differentiation determinants of kinase inhibitor sensitivity in leukemia cells. , 2017, Leukemia.

[64]  Tony Pawson,et al.  NetworKIN: a resource for exploring cellular phosphorylation networks , 2007, Nucleic Acids Res..

[65]  Steven P Gygi,et al.  Phosphoproteomic Analysis of the Developing Mouse Brain*S , 2004, Molecular & Cellular Proteomics.

[66]  Sandhya Rani,et al.  Human Protein Reference Database—2009 update , 2008, Nucleic Acids Res..

[67]  Dhruv Chauhan,et al.  A Genome-wide CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) Screen Identifies NEK7 as an Essential Component of NLRP3 Inflammasome Activation* , 2015, The Journal of Biological Chemistry.

[68]  Gary D. Bader,et al.  An automated method for finding molecular complexes in large protein interaction networks , 2003, BMC Bioinformatics.

[69]  Ronald J. Moore,et al.  Integrated Proteogenomic Characterization of Human High-Grade Serous Ovarian Cancer , 2016, Cell.

[70]  S. Gygi,et al.  Global Analysis of Cdk1 Substrate Phosphorylation Sites Provides Insights into Evolution , 2009, Science.

[71]  Man Kit Cheung,et al.  Human proteins with target sites of multiple post-translational modification types are more prone to be involved in disease. , 2014, Journal of proteome research.

[72]  B. Searle,et al.  “Plug-and-play” investigation of the human phosphoproteome by targeted high-resolution mass spectrometry , 2016, Nature Methods.

[73]  Chunaram Choudhary,et al.  Acetylation dynamics and stoichiometry in Saccharomyces cerevisiae , 2014, Molecular systems biology.

[74]  M. Tomita,et al.  Muscle-specific Knock-out of NUAK Family SNF1-like Kinase 1 (NUAK1) Prevents High Fat Diet-induced Glucose Intolerance* , 2012, The Journal of Biological Chemistry.

[75]  G. Hart,et al.  Cross-talk between GlcNAcylation and phosphorylation: Site-specific phosphorylation dynamics in response to globally elevated O-GlcNAc , 2008, Proceedings of the National Academy of Sciences.

[76]  Matthias Mann,et al.  In vivo brain GPCR signaling elucidated by phosphoproteomics , 2018, Science.

[77]  Sean J. Humphrey,et al.  Glucose-regulated and drug-perturbed phosphoproteome reveals molecular mechanisms controlling insulin secretion , 2016, Nature Communications.

[78]  Chi-Ying F. Huang,et al.  Ultrasensitivity in the mitogen-activated protein kinase cascade. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[79]  M. Mann,et al.  Temporal analysis of phosphotyrosine-dependent signaling networks by quantitative proteomics , 2004, Nature Biotechnology.

[80]  J. Cox,et al.  Proteomics strategy for quantitative protein interaction profiling in cell extracts , 2009, Nature Methods.

[81]  Jean Yee Hwa Yang,et al.  Positive-unlabeled ensemble learning for kinase substrate prediction from dynamic phosphoproteomics data , 2015, Bioinform..

[82]  J. Gribben,et al.  Empirical inference of circuitry and plasticity in a kinase signaling network , 2015, Proceedings of the National Academy of Sciences.

[83]  Tony Hunter,et al.  pHisphorylation: the emergence of histidine phosphorylation as a reversible regulatory modification. , 2017, Current opinion in cell biology.

[84]  P. Cohen,et al.  The selectivity of protein kinase inhibitors: a further update. , 2007, The Biochemical journal.

[85]  M. Mann,et al.  Ultradeep human phosphoproteome reveals a distinct regulatory nature of Tyr and Ser/Thr-based signaling. , 2014, Cell reports.

[86]  Cornelia Schönbauer,et al.  Systematic genetic analysis of muscle morphogenesis and function in Drosophila , 2010, Nature.

[87]  Jan C. Refsgaard,et al.  From Phosphosites to Kinases. , 2016, Methods in molecular biology.

[88]  S. Tooze,et al.  siRNA Screening of the Kinome Identifies ULK1 as a Multidomain Modulator of Autophagy* , 2007, Journal of Biological Chemistry.

[89]  Emily J. Girard,et al.  Genome-wide CRISPR-Cas9 Screens Reveal Loss of Redundancy between PKMYT1 and WEE1 in Glioblastoma Stem-like Cells. , 2015, Cell reports.

[90]  Dario R Alessi,et al.  Kinase drug discovery--what's next in the field? , 2013, ACS chemical biology.

[91]  J. Shabanowitz,et al.  Phosphoproteome analysis by mass spectrometry and its application to Saccharomyces cerevisiae , 2002, Nature Biotechnology.

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

[93]  Waltraud X. Schulze,et al.  A Novel Proteomic Screen for Peptide-Protein Interactions* , 2004, Journal of Biological Chemistry.

[94]  Hui Li,et al.  Control of muscle formation by the fusogenic micropeptide myomixer , 2017, Science.

[95]  P. Cohen,et al.  The origins of protein phosphorylation , 2002, Nature Cell Biology.

[96]  P. Alam ‘L’ , 2021, Composites Engineering: An A–Z Guide.

[97]  Trey Ideker,et al.  Cytoscape 2.8: new features for data integration and network visualization , 2010, Bioinform..

[98]  S. Ross,et al.  New Perspectives, Opportunities, and Challenges in Exploring the Human Protein Kinome. , 2018, Cancer research.

[99]  Toru Okamoto,et al.  The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. , 2013, Immunity.

[100]  E. Birney,et al.  Pfam: the protein families database , 2013, Nucleic Acids Res..

[101]  P. Alam ‘S’ , 2021, Composites Engineering: An A–Z Guide.

[102]  Mike Tyers,et al.  Machine Learning of Global Phosphoproteomic Profiles Enables Discrimination of Direct versus Indirect Kinase Substrates* , 2017, Molecular & Cellular Proteomics.

[103]  Michael J. MacCoss,et al.  A Library of Phosphoproteomic and Chromatin Signatures for Characterizing Cellular Responses to Drug Perturbations , 2017, bioRxiv.

[104]  A. Biankin,et al.  Resolution of Novel Pancreatic Ductal Adenocarcinoma Subtypes by Global Phosphotyrosine Profiling* , 2016, Molecular & Cellular Proteomics.

[105]  Nolan J Hoffman,et al.  Global Phosphoproteomic Analysis of Human Skeletal Muscle Reveals a Network of Exercise-Regulated Kinases and AMPK Substrates. , 2015, Cell metabolism.

[106]  David E James,et al.  High-throughput and high-sensitivity phosphoproteomics with the EasyPhos platform , 2018, Nature Protocols.

[107]  M. Yanagisawa,et al.  Quantitative phosphoproteomic analysis of the molecular substrates of sleep need , 2018, Nature.

[108]  Deanna M. Church,et al.  ClinVar: public archive of relationships among sequence variation and human phenotype , 2013, Nucleic Acids Res..

[109]  G. Drewes,et al.  Tracking cancer drugs in living cells by thermal profiling of the proteome , 2014, Science.

[110]  T. Hunter The age of crosstalk: phosphorylation, ubiquitination, and beyond. , 2007, Molecular cell.

[111]  K. Kaestner,et al.  Insulin Resistance and a Diabetes Mellitus-Like Syndrome in Mice Lacking the Protein Kinase Akt2 (PKBβ) , 2001 .

[112]  Tony Pawson,et al.  Temporal regulation of EGF signaling networks by the scaffold protein Shc1 , 2013, Nature.

[113]  J. Ellenberg,et al.  The quantitative proteome of a human cell line , 2011, Molecular systems biology.

[114]  Krister Wennerberg,et al.  Quantitative scoring of differential drug sensitivity for individually optimized anticancer therapies , 2014, Scientific Reports.

[115]  Bin Zhang,et al.  PhosphoSitePlus: a comprehensive resource for investigating the structure and function of experimentally determined post-translational modifications in man and mouse , 2011, Nucleic Acids Res..

[116]  J. Ferrell,et al.  Tuning the Activation Threshold of a Kinase Network by Nested Feedback Loops , 2009, Science.

[117]  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.

[118]  J. Coon,et al.  Phosphoproteomics in the Age of Rapid and Deep Proteome Profiling , 2015, Analytical chemistry.

[119]  Gautier Koscielny,et al.  The International Mouse Phenotyping Consortium Web Portal, a unified point of access for knockout mice and related phenotyping data , 2013, Nucleic Acids Res..

[120]  Sean J. Humphrey,et al.  Phosphorylation Is a Central Mechanism for Circadian Control of Metabolism and Physiology. , 2017, Cell metabolism.

[121]  Sean J. Humphrey,et al.  Targeted phosphoproteomics of insulin signaling using data-independent acquisition mass spectrometry , 2015, Science Signaling.

[122]  L. Wodicka,et al.  A small molecule–kinase interaction map for clinical kinase inhibitors , 2005, Nature Biotechnology.

[123]  Le Cong,et al.  Multiplex Genome Engineering Using CRISPR/Cas Systems , 2013, Science.

[124]  J. Fitzgibbon,et al.  Proteomic and genomic integration identifies kinase and differentiation determinants of kinase inhibitor sensitivity in leukemia cells , 2017, Leukemia.

[125]  Blagoy Blagoev,et al.  Functional proteomics defines the molecular switch underlying FGF receptor trafficking and cellular outputs. , 2013, Molecular cell.

[126]  J. Doudna,et al.  The new frontier of genome engineering with CRISPR-Cas9 , 2014, Science.

[127]  E. Nishida,et al.  A conserved docking motif in MAP kinases common to substrates, activators and regulators , 2000, Nature Cell Biology.

[128]  Heiner Koch,et al.  The target landscape of clinical kinase drugs , 2017, Science.

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

[130]  M. Mann,et al.  A large synthetic peptide and phosphopeptide reference library for mass spectrometry–based proteomics , 2013, Nature Biotechnology.

[131]  John P. Overington,et al.  The promise and peril of chemical probes. , 2015, Nature chemical biology.

[132]  Ronald W. Davis,et al.  Functional profiling of the Saccharomyces cerevisiae genome , 2002, Nature.

[133]  Bernhard Mlecnik,et al.  CluePedia Cytoscape plugin: pathway insights using integrated experimental and in silico data , 2013, Bioinform..

[134]  Chad J. Miller,et al.  Homing in: Mechanisms of Substrate Targeting by Protein Kinases. , 2018, Trends in biochemical sciences.

[135]  Chern Han Yong,et al.  Thermal proximity coaggregation for system-wide profiling of protein complex dynamics in cells , 2018, Science.

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

[137]  Christian von Mering,et al.  Cell-wide analysis of protein thermal unfolding reveals determinants of thermostability , 2017, Science.

[138]  Danny Reinberg,et al.  Methylation-Acetylation Interplay Activates p53 in Response to DNA Damage , 2007, Molecular and Cellular Biology.

[139]  M. Mann,et al.  Organellar Proteomics and Phospho-Proteomics Reveal Subcellular Reorganization in Diet-Induced Hepatic Steatosis. , 2018, Developmental cell.

[140]  Masaru Tomita,et al.  Microscale phosphoproteome analysis of 10,000 cells from human cancer cell lines. , 2011, Analytical chemistry.

[141]  P. Nordlund,et al.  Monitoring Drug Target Engagement in Cells and Tissues Using the Cellular Thermal Shift Assay , 2013, Science.

[142]  S. Knapp,et al.  A systematic interaction map of validated kinase inhibitors with Ser/Thr kinases , 2007, Proceedings of the National Academy of Sciences.

[143]  Toshifumi Takao,et al.  Transmembrane phosphoprotein Cbp regulates the activities of Src-family tyrosine kinases , 2000, Nature.

[144]  S. Mohammed,et al.  Improved peptide identification by targeted fragmentation using CID, HCD and ETD on an LTQ-Orbitrap Velos. , 2011, Journal of proteome research.

[145]  Gary D Bader,et al.  The human genome and drug discovery after a decade. Roads (still) not taken , 2011, 1102.0448.

[146]  A. Heck,et al.  Crosstalk between phosphorylation and O‐GlcNAcylation: friend or foe , 2018, The FEBS journal.

[147]  S. Brunak,et al.  Phosphoproteomics of Primary Cells Reveals Druggable Kinase Signatures in Ovarian Cancer , 2017, Cell reports.

[148]  Edward L. Huttlin,et al.  A large-scale method to measure absolute protein phosphorylation stoichiometries , 2011, Nature Methods.

[149]  Lars Malmström,et al.  Inference and quantification of peptidoforms in large sample cohorts by SWATH-MS , 2017, Nature Biotechnology.

[150]  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.

[151]  S. Brunak,et al.  Quantitative Phosphoproteomics Reveals Widespread Full Phosphorylation Site Occupancy During Mitosis , 2010, Science Signaling.

[152]  M. Mann,et al.  In vivo SILAC-based proteomics reveals phosphoproteome changes during mouse skin carcinogenesis. , 2013, Cell reports.

[153]  M. Baccarani,et al.  Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. , 2002, The New England journal of medicine.

[154]  L. Castagnoli,et al.  The human phosphatase interactome: An intricate family portrait , 2012, FEBS letters.

[155]  M. Mann,et al.  The coming age of complete, accurate, and ubiquitous proteomes. , 2013, Molecular cell.

[156]  A. Hauschild,et al.  Improved survival with vemurafenib in melanoma with BRAF V600E mutation. , 2011, The New England journal of medicine.

[157]  Matthias Mann,et al.  Revisiting biomarker discovery by plasma proteomics , 2017, Molecular systems biology.

[158]  D. Goodlett,et al.  Multiplexed and data-independent tandem mass spectrometry for global proteome profiling. , 2014, Mass spectrometry reviews.

[159]  Bryan P. Early,et al.  A Protease for Middle Down Proteomics , 2012, Nature Methods.

[160]  J. Schapansky,et al.  The complex relationships between microglia, alpha-synuclein, and LRRK2 in Parkinson’s disease , 2015, Neuroscience.

[161]  Patrick G. A. Pedrioli,et al.  Phosphoproteomic Analysis Reveals Interconnected System-Wide Responses to Perturbations of Kinases and Phosphatases in Yeast , 2010, Science Signaling.

[162]  Bernhard Kuster,et al.  Comparing immobilized kinase inhibitors and covalent ATP probes for proteomic profiling of kinase expression and drug selectivity. , 2013, Journal of proteome research.

[163]  Caleb J Bashor,et al.  The Ste5 Scaffold Allosterically Modulates Signaling Output of the Yeast Mating Pathway , 2006, Science.

[164]  T. Hunter,et al.  Evidence for in vivo phosphorylation of the Grb2 SH2-domain binding site on focal adhesion kinase by Src-family protein-tyrosine kinases , 1996, Molecular and cellular biology.

[165]  G. Lienhard,et al.  Non-functional phosphorylations? , 2008, Trends in biochemical sciences.

[166]  H. Wiśniewski,et al.  Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. , 1986, Proceedings of the National Academy of Sciences of the United States of America.

[167]  S. Gygi,et al.  Chemical genetic screen for AMPKα2 substrates uncovers a network of proteins involved in mitosis. , 2011, Molecular cell.

[168]  T. McGraw,et al.  Development of a new model system to dissect isoform specific Akt signalling in adipocytes , 2015, The Biochemical journal.

[169]  K. Shokat,et al.  Escape from HER family tyrosine kinase inhibitor therapy by the kinase inactive HER3 , 2007, Nature.

[170]  Albert J R Heck,et al.  Widespread bacterial protein histidine phosphorylation revealed by mass spectrometry-based proteomics , 2018, Nature Methods.

[171]  M. Mann,et al.  Quantitative Phosphoproteomics Applied to the Yeast Pheromone Signaling Pathway*S , 2005, Molecular & Cellular Proteomics.

[172]  Heiner Koch,et al.  Phosphoproteome Profiling Reveals Molecular Mechanisms of Growth-Factor-Mediated Kinase Inhibitor Resistance in EGFR-Overexpressing Cancer Cells. , 2016, Journal of proteome research.

[173]  Shabaz Mohammed,et al.  Comparative Phosphoproteomic Analysis of Checkpoint Recovery Identifies New Regulators of the DNA Damage Response , 2013, Science Signaling.

[174]  B. Garcia,et al.  Differential quantification of isobaric phosphopeptides using data-independent acquisition mass spectrometry. , 2016, Molecular bioSystems.

[175]  S. Fields,et al.  Global analysis of phosphorylation and ubiquitylation cross-talk in protein degradation , 2013, Nature Methods.

[176]  J. Ferrell,et al.  Mechanisms of specificity in protein phosphorylation , 2007, Nature Reviews Molecular Cell Biology.

[177]  James E. DiCarlo,et al.  RNA-Guided Human Genome Engineering via Cas9 , 2013, Science.

[178]  D. James,et al.  Kinome Screen Identifies PFKFB3 and Glucose Metabolism as Important Regulators of the Insulin/Insulin-like Growth Factor (IGF)-1 Signaling Pathway* , 2015, The Journal of Biological Chemistry.

[179]  B. Taylor,et al.  Tumours with class 3 BRAF mutants are sensitive to the inhibition of activated RAS , 2017, Nature.

[180]  T. Hunter,et al.  Protein kinases and phosphatases: The Yin and Yang of protein phosphorylation and signaling , 1995, Cell.

[181]  Ian Collins,et al.  Probing the Probes: Fitness Factors For Small Molecule Tools , 2010, Chemistry & biology.

[182]  Florian Gnad,et al.  Evolutionary Constraints of Phosphorylation in Eukaryotes, Prokaryotes, and Mitochondria* , 2010, Molecular & Cellular Proteomics.

[183]  J. Graff,et al.  Drosophila glucome screening identifies Ck1alpha as a regulator of mammalian glucose metabolism , 2015, Nature Communications.

[184]  Yingming Zhao,et al.  Metabolic Regulation by Lysine Malonylation, Succinylation, and Glutarylation* , 2015, Molecular & Cellular Proteomics.

[185]  Jürg Zimmermann,et al.  Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr–Abl positive cells , 1996, Nature Medicine.

[186]  T. Hunter,et al.  Oncogenic kinase signalling , 2001, Nature.

[187]  M. Pool,et al.  Phosphorylation-mimicking glutamate clusters in the proline-rich region are sufficient to simulate the functional deficiencies of hyperphosphorylated tau protein. , 2001, The Biochemical journal.

[188]  K. Shokat,et al.  Identification of AMPK Phosphorylation Sites Reveals a Network of Proteins Involved in Cell Invasion and Facilitates Large-Scale Substrate Prediction. , 2015, Cell metabolism.

[189]  P. Alam ‘E’ , 2021, Composites Engineering: An A–Z Guide.

[190]  S. Gabriel,et al.  EGFR Mutations in Lung Cancer: Correlation with Clinical Response to Gefitinib Therapy , 2004, Science.

[191]  Derek J. Bailey,et al.  The One Hour Yeast Proteome* , 2013, Molecular & Cellular Proteomics.

[192]  Edwin Smith,et al.  The Language of Histone Crosstalk , 2010, Cell.

[193]  L. Iakoucheva,et al.  The importance of intrinsic disorder for protein phosphorylation. , 2004, Nucleic acids research.

[194]  Raymond J. Deshaies,et al.  Multisite Phosphorylation and the Countdown to S Phase , 2001, Cell.

[195]  J. Coon,et al.  Proteome sequencing goes deep. , 2015, Current opinion in chemical biology.

[196]  Tony Pawson,et al.  Comparative Analysis Reveals Conserved Protein Phosphorylation Networks Implicated in Multiple Diseases , 2009, Science Signaling.

[197]  G. Drewes,et al.  Thermal proteome profiling for unbiased identification of direct and indirect drug targets using multiplexed quantitative mass spectrometry , 2015, Nature Protocols.

[198]  Jianmin Wu,et al.  Phosphoproteomic Profiling Identifies Focal Adhesion Kinase as a Mediator of Docetaxel Resistance in Castrate-Resistant Prostate Cancer , 2013, Molecular Cancer Therapeutics.

[199]  K. Shokat,et al.  Targeted polypharmacology: Discovery of dual inhibitors of tyrosine and phosphoinositide kinases , 2008, Nature chemical biology.

[200]  Karina D. Sørensen,et al.  An Optimized Shotgun Strategy for the Rapid Generation of Comprehensive Human Proteomes , 2017, Cell systems.

[201]  M. Goumans,et al.  Defined Engineered Human Myocardium With Advanced Maturation for Applications in Heart Failure Modeling and Repair , 2017, Circulation.

[202]  D. Iliopoulos,et al.  Phosphoproteomics screen reveals akt isoform-specific signals linking RNA processing to lung cancer. , 2014, Molecular cell.

[203]  B. Kemp,et al.  Protein kinase recognition sequence motifs. , 1990, Trends in biochemical sciences.

[204]  K. Shokat,et al.  A molecular gate which controls unnatural ATP analogue recognition by the tyrosine kinase v-Src. , 1998, Bioorganic & medicinal chemistry.

[205]  G. Hart,et al.  Cross talk between O-GlcNAcylation and phosphorylation: roles in signaling, transcription, and chronic disease. , 2011, Annual review of biochemistry.

[206]  Matthias Mann,et al.  Systematic proteomic analysis of LRRK2-mediated Rab GTPase phosphorylation establishes a connection to ciliogenesis , 2017, eLife.

[207]  Nolan J Hoffman,et al.  Structural Basis for Phosphorylation and Lysine Acetylation Cross-talk in a Kinase Motif Associated with Myocardial Ischemia and Cardioprotection* , 2014, The Journal of Biological Chemistry.