Evaluation of Chemically-Cleavable Linkers for Quantitative Mapping of Small Molecule-Cysteinome Reactivity.

Numerous reagents have been developed to enable chemical proteomic analysis of small molecule-protein interactomes. However, the performance of these reagents has not been systematically evaluated and compared. Herein, we report our efforts to conduct a parallel assessment of two widely-used chemically-cleavable linkers equipped with dialkoxydiphenylsilane (DADPS linker) and azobenzene (AZO linker) moieties. Profiling a cellular cysteinome using iodoacetamide alkyne probe demonstrated a significant discrepancy between the experimental results obtained through the application of each of the reagents. To better understand the source of observed discrepancy, we evaluated the key sample preparation steps. We also performed mass tolerant database search strategy using MSFragger software. This resulted in identifying a previously unreported artifactual modification on the residual mass of the azobenzene linker. Furthermore, we conducted a comparative analysis of enrichment modes using both cleavable linkers. This effort determined that enrichment of proteolytic digests yielded a far greater number of identified cysteine residues than the enrichment conducted prior to protein digest. Inspired by recent studies where multiplexed quantitative labeling strategies were applied to cleavable biotin linkers, we combined this further optimized protocol using the DADPS cleavable linker with tandem mass tag (TMT) labeling to profile the FDA-approved covalent EGFR kinase inhibitor dacomitinib against the cysteinome of an epidermoid cancer cell line. Our analysis resulted in the detection and quantification of over 10,000 unique cysteine residues, a nearly 3-fold increase over previous studies that used cleavable biotin linkers for enrichment. Critically, cysteine residues corresponding to proteins directly as well as indirectly modulated by dacomitinib treatment were identified. Overall, our study suggests that the dialkoxydiphenylsilane linker could be broadly applied wherever chemically cleavable linkers are required for chemical proteomic characterization of cellular proteomes.

[1]  S. Knapp,et al.  The Cysteinome of Protein Kinases as a Target in Drug Development. , 2018, Angewandte Chemie.

[2]  J. Casanova,et al.  Dimethyl Fumarate Disrupts Human Innate Immune Signaling by Targeting the IRAK4–MyD88 Complex , 2019, The Journal of Immunology.

[3]  David Baker,et al.  Quantitative reactivity profiling predicts functional cysteines in proteomes , 2010, Nature.

[4]  Luis Mendoza,et al.  PTMProphet: Fast and Accurate Mass Modification Localization for the Trans-Proteomic Pipeline. , 2019, Journal of proteome research.

[5]  Chun Xing Li,et al.  Isotopically-Labeled Iodoacetamide-Alkyne Probes for Quantitative Cysteine-Reactivity Profiling. , 2017, Molecular pharmaceutics.

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

[7]  N. Gray,et al.  Leveraging Gas-Phase Fragmentation Pathways for Improved Identification and Selective Detection of Targets Modified by Covalent Probes. , 2016, Analytical chemistry.

[8]  Benjamin F. Cravatt,et al.  Chemical Proteomics Identifies Druggable Vulnerabilities in a Genetically Defined Cancer , 2017, Cell.

[9]  H. Zou,et al.  Selective Enrichment of Cysteine-Containing Phosphopeptides for Subphosphoproteome Analysis. , 2015, Journal of proteome research.

[10]  A. Mfuh,et al.  Small Molecule Interactome Mapping by Photoaffinity Labeling Reveals Binding Site Hotspots for the NSAIDs. , 2018, Journal of the American Chemical Society.

[11]  C. Bertozzi,et al.  Isotope-targeted glycoproteomics (IsoTaG): a mass-independent platform for intact N- and O-glycopeptide discovery and analysis , 2015, Nature Methods.

[12]  Stacy T. Knutson,et al.  Functional site profiling and electrostatic analysis of cysteines modifiable to cysteine sulfenic acid , 2008, Protein science : a publication of the Protein Society.

[13]  Fan Yang,et al.  A Dimethyl-Labeling-Based Strategy for Site-Specifically Quantitative Chemical Proteomics. , 2018, Analytical chemistry.

[14]  Christina M. Woo,et al.  Mapping the Small Molecule Interactome by Mass Spectrometry. , 2018, Biochemistry.

[15]  B. Cravatt,et al.  A tandem orthogonal proteolysis strategy for high-content chemical proteomics. , 2005, Journal of the American Chemical Society.

[16]  A. Olson,et al.  Proteome-wide covalent ligand discovery in native biological systems , 2016, Nature.

[17]  Steven P Gygi,et al.  Streamlined Tandem Mass Tag (SL-TMT) Protocol: An Efficient Strategy for Quantitative (Phospho)proteome Profiling Using Tandem Mass Tag-Synchronous Precursor Selection-MS3. , 2018, Journal of proteome research.

[18]  J. Vilo,et al.  g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update) , 2019, Nucleic Acids Res..

[19]  A. Doucette,et al.  Preventing N‐ and O‐formylation of proteins when incubated in concentrated formic acid , 2016, Proteomics.

[20]  Jing Yang,et al.  Global, in situ, site-specific analysis of protein S-sulfenylation , 2015, Nature Protocols.

[21]  Jigang Wang,et al.  Mapping sites of aspirin-induced acetylations in live cells by quantitative acid-cleavable activity-based protein profiling (QA-ABPP) , 2015, Scientific Reports.

[22]  Sarah Lin,et al.  TagGraph reveals vast protein modification landscapes from large tandem mass spectrometry data sets , 2019, Nature Biotechnology.

[23]  E. Weerapana,et al.  A Quantitative Chemoproteomic Platform to Monitor Selenocysteine Reactivity within a Complex Proteome. , 2018, Cell chemical biology.

[24]  B. Cravatt,et al.  Tandem orthogonal proteolysis-activity-based protein profiling (TOP-ABPP)—a general method for mapping sites of probe modification in proteomes , 2007, Nature Protocols.

[25]  L. Fu,et al.  Multiplexed Thiol Reactivity Profiling for Target Discovery of Electrophilic Natural Products. , 2017, Cell chemical biology.

[26]  Thawfeek M. Varusai,et al.  The Reactome Pathway Knowledgebase , 2017, Nucleic acids research.

[27]  Stefano Forli,et al.  Global profiling of lysine reactivity and ligandability in the human proteome. , 2017, Nature chemistry.

[28]  G. Charron,et al.  Comparative analysis of cleavable azobenzene-based affinity tags for bioorthogonal chemical proteomics. , 2010, Chemistry & biology.

[29]  Xing Chen,et al.  Quantitative Profiling of Protein O-GlcNAcylation Sites by an Isotope-Tagged Cleavable Linker. , 2018, ACS chemical biology.

[30]  J. Yates,et al.  Chemoproteomic profiling and discovery of protein electrophiles in human cells , 2016, Nature chemistry.

[31]  Alexey I Nesvizhskii,et al.  MSFragger: ultrafast and comprehensive peptide identification in shotgun proteomics , 2017, Nature Methods.

[32]  John R. Yates,et al.  Direct Detection of Biotinylated Proteins by Mass Spectrometry , 2014, Journal of proteome research.

[33]  B. Cravatt,et al.  Enzyme inhibitor discovery by activity-based protein profiling. , 2014, Annual review of biochemistry.

[34]  F. Khuri,et al.  Tyrosine phosphorylation of mitochondrial pyruvate dehydrogenase kinase 1 is important for cancer metabolism. , 2011, Molecular cell.

[35]  M. Nikolic,et al.  Epidermal growth factor receptor expression by human squamous cell carcinomas of the head and neck, cell lines and xenografts. , 1994, British Journal of Cancer.

[36]  Qian Wang,et al.  Bioconjugation by copper(I)-catalyzed azide-alkyne [3 + 2] cycloaddition. , 2003, Journal of the American Chemical Society.

[37]  R. Ewing,et al.  ELF3 is an antagonist of oncogenic-signalling-induced expression of EMT-TF ZEB1 , 2018, Cancer biology & therapy.

[38]  P. Sorger,et al.  Leveraging Compound Promiscuity to Identify Targetable Cysteines within the Kinome. , 2019, Cell chemical biology.

[39]  J. Woodgett,et al.  Modulation of the glycogen synthase kinase‐3 family by tyrosine phosphorylation. , 1993, The EMBO journal.

[40]  T. Harris,et al.  Structural Basis of Perturbed pKa Values of Catalytic Groups in Enzyme Active Sites , 2002, IUBMB life.

[41]  E. Weerapana,et al.  An Isotopically Tagged Azobenzene‐Based Cleavable Linker for Quantitative Proteomics , 2013, Chembiochem : a European journal of chemical biology.

[42]  B. Remington,et al.  Cysteine pKa depression by a protonated glutamic acid in human DJ-1. , 2008, Biochemistry.

[43]  K. Geoghegan,et al.  Mapping the Binding Site of BMS-708163 on γ-Secretase with Cleavable Photoprobes. , 2017, Cell chemical biology.

[44]  Youli Xiao,et al.  Characterization of the Artemisinin Binding Site for Translationally Controlled Tumor Protein (TCTP) by Bioorthogonal Click Chemistry. , 2016, Bioconjugate chemistry.

[45]  Stephen R. Johnson,et al.  Ligand and Target Discovery by Fragment-Based Screening in Human Cells , 2017, Cell.

[46]  M. Bogyo,et al.  A mild chemically cleavable linker system for functional proteomic applications. , 2007, Angewandte Chemie.

[47]  Daniela C Dieterich,et al.  Cleavable biotin probes for labeling of biomolecules via azide-alkyne cycloaddition. , 2010, Journal of the American Chemical Society.

[48]  J. P. Danehy,et al.  Iodometric method for the determination of dithionite, bisulfite, and thiosulfate in the presence of each other and its use in following the decomposition of aqueous solutions of sodium dithionite. , 1974, Analytical chemistry.

[49]  M. Bogyo,et al.  Activity‐based probes for the ubiquitin conjugation–deconjugation machinery: new chemistries, new tools, and new insights , 2017, The FEBS journal.

[50]  M. Bogyo,et al.  Reactive-site-centric chemoproteomics identifies a distinct class of deubiquitinase enzymes , 2018, Nature Communications.

[51]  Yinliang Yang,et al.  Cleavable Linkers in Chemical Proteomics Applications. , 2017, Methods in molecular biology.

[52]  E. Weerapana,et al.  Identifying Functional Cysteine Residues in the Mitochondria. , 2017, ACS chemical biology.

[53]  Edward L. Huttlin,et al.  An ultra-tolerant database search reveals that a myriad of modified peptides contributes to unassigned spectra in shotgun proteomics , 2015, Nature Biotechnology.

[54]  T. Hunter,et al.  The eukaryotic protein kinase superfamily: kinase (catalytic) domain structure and classification 1 , 1995, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[55]  C. D. de Koster,et al.  Selective enrichment of azide-containing peptides from complex mixtures. , 2009, Journal of proteome research.

[56]  L. Degroot,et al.  A human early response gene homologous to murine nur77 and rat NGFI-B, and related to the nuclear receptor superfamily. , 1990, Molecular endocrinology.

[57]  Martin Eisenacher,et al.  The PRIDE database and related tools and resources in 2019: improving support for quantification data , 2018, Nucleic Acids Res..

[58]  Salvatore Oliviero,et al.  Histone Crosstalk between H3S10ph and H4K16ac Generates a Histone Code that Mediates Transcription Elongation , 2009, Cell.

[59]  G. Drewes,et al.  Chemoproteomics and Chemical Probes for Target Discovery. , 2018, Trends in biotechnology.

[60]  Benjamin F. Cravatt,et al.  A roadmap to evaluate the proteome-wide selectivity of covalent kinase inhibitors , 2014, Nature chemical biology.

[61]  K. Gajiwala,et al.  Proteome-wide Map of Targets of T790M-EGFR-Directed Covalent Inhibitors. , 2017, Cell chemical biology.