Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Global targeting of functional tyrosines using sulfur-triazole exchange chemistry

Abstract

Covalent probes serve as valuable tools for global investigation of protein function and ligand binding capacity. Despite efforts to expand coverage of residues available for chemical proteomics (e.g., cysteine and lysine), a large fraction of the proteome remains inaccessible with current activity-based probes. Here, we introduce sulfur-triazole exchange (SuTEx) chemistry as a tunable platform for developing covalent probes with broad applications for chemical proteomics. We show modifications to the triazole leaving group can furnish sulfonyl probes with ~5-fold enhanced chemoselectivity for tyrosines over other nucleophilic amino acids to investigate more than 10,000 tyrosine sites in lysates and live cells. We discover that tyrosines with enhanced nucleophilicity are enriched in enzymatic, protein–protein interaction and nucleotide recognition domains. We apply SuTEx as a chemical phosphoproteomics strategy to monitor activation of phosphotyrosine sites. Collectively, we describe SuTEx as a biocompatible chemistry for chemical biology investigations of the human proteome.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Development of sulfur-triazole exchange (SuTEx) chemistry for chemical proteomics.
Fig. 2: Functional tyrosine profiling in proteomes and live cells.
Fig. 3: SuTEx-enabled discovery of intrinsically nucleophilic tyrosines in human cell proteomes.
Fig. 4: Tuning SuTEx probes for tyrosine chemoselectivity in cell proteomes.
Fig. 5: Triazole LG enhances phenol reactivity of sulfonyl probes in solution.
Fig. 6: Chemical phosphotyrosine-proteomics by SuTEx.

Similar content being viewed by others

Data availability

All data produced or analyzed for this study are included in the published article (and its Supplementary Information files) or are available from the corresponding author on reasonable request. Crystallographic data for small molecules has been deposited in the Cambridge Crystallographic Data Centre and have been assigned the following deposition numbers HHS-465 (CCDC 1954297), HHS-475 (CCDC 1954298) and HHS-483 (CCDC 1954299). These data can be obtained free of charge from The Cambridge Crystallographic Data Centre via www.ccdc.cam.ac.uk/structures.

Code availability

All code is available upon reasonable request from the corresponding author.

References

  1. Cravatt, B. F., Wright, A. T. & Kozarich, J. W. Activity-based protein profiling: from enzyme chemistry to proteomic chemistry. Annu. Rev. Biochem. 77, 383–414 (2008).

    Article  CAS  PubMed  Google Scholar 

  2. Sadaghiani, A. M., Verhelst, S. H. & Bogyo, M. Tagging and detection strategies for activity-based proteomics. Curr. Opin. Chem. Biol. 11, 20–28 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Niphakis, M. J. & Cravatt, B. F. Enzyme inhibitor discovery by activity-based protein profiling. Annu. Rev. Biochem. 83, 341–377 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. Bachovchin, D. A. & Cravatt, B. F. The pharmacological landscape and therapeutic potential of serine hydrolases. Nat. Rev. Drug Disco. 11, 52–68 (2012).

    Article  CAS  Google Scholar 

  5. Deu, E., Verdoes, M. & Bogyo, M. New approaches for dissecting protease functions to improve probe development and drug discovery. Nat. Struct. Mol. Biol. 19, 9–16 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Patricelli, M. P. et al. Functional interrogation of the kinome using nucleotide acyl phosphates. Biochemistry 46, 350–358 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Kumar, S. et al. Activity-based probes for protein tyrosine phosphatases. Proc. Natl Acad. Sci. USA 101, 7943–7948 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Vocadlo, D. J. & Bertozzi, C. R. A strategy for functional proteomic analysis of glycosidase activity from cell lysates. Angew. Chem. Int. Ed. Engl. 43, 5338–5342 (2004).

    Article  CAS  PubMed  Google Scholar 

  9. Liu, Y., Patricelli, M. P. & Cravatt, B. F. Activity-based protein profiling: the serine hydrolases. Proc. Natl Acad. Sci. USA 96, 14694–14699 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Weerapana, E. et al. Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 468, 790–795 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hacker, S. M. et al. Global profiling of lysine reactivity and ligandability in the human proteome. Nat. Chem. 9, 1181–1190 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lin, S. et al. Redox-based reagents for chemoselective methionine bioconjugation. Science 355, 597–602 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Matthews, M. L. et al. Chemoproteomic profiling and discovery of protein electrophiles in human cells. Nat. Chem. 9, 234–243 (2017).

    Article  CAS  PubMed  Google Scholar 

  14. Parker, C. G. et al. Ligand and target discovery by fragment-based screening in human cells. Cell 168, 527–541 e529 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Narayanan, A. & Jones, L. H. Sulfonyl fluorides as privileged warheads in chemical biology. Chem. Sci. 6, 2650–2659 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gao, B. et al. Bifluoride-catalysed sulfur(VI) fluoride exchange reaction for the synthesis of polysulfates and polysulfonates. Nat. Chem. 9, 1083–1088 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Dong, J., Sharpless, K. B., Kwisnek, L., Oakdale, J. S. & Fokin, V. V. SuFEx-based synthesis of polysulfates. Angew. Chem. Int. Ed. Engl. 53, 9466–9470 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Fahrney, D. E. & Gold, A. M. Sulfonyl fluorides as inhibitors of esterases. I. rates of reaction with acetylcholinesterase, α-chymotrypsin, and trypsin. J. Am. Chem. Soc. 85, 997–1000 (1963).

    Article  CAS  Google Scholar 

  19. Shannon, D. A. et al. Sulfonyl fluoride analogues as activity-based probes for serine proteases. Chem. Bio. Chem. 13, 2327–2330 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Gu, C. et al. Chemical proteomics with sulfonyl fluoride probes reveals selective labeling of functional tyrosines in glutathione transferases. Chem. Biol. 20, 541–548 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Zhao, Q. et al. Broad-spectrum kinase profiling in live cells with lysine-targeted sulfonyl fluoride probes. J. Am. Chem. Soc. 139, 680–685 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yang, B. et al. Proximity-enhanced SuFEx chemical cross-linker for specific and multitargeting cross-linking mass spectrometry. Proc. Natl Acad. Sci. USA 115, 11162–11167 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Yang, X. et al. An affinity-based probe for the human adenosine A2A receptor. J. Med. Chem. 61, 7892–7901 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Dong, J., Krasnova, L., Finn, M. G. & Sharpless, K. B. Sulfur(VI) fluoride exchange (SuFEx): another good reaction for click chemistry. Angew. Chem. Int. Ed. Engl. 53, 9430–9448 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Chen, W. et al. Arylfluorosulfates inactivate intracellular lipid binding protein(s) through chemoselective SuFEx reaction with a binding site tyr residue. J. Am. Chem. Soc. 138, 7353–7364 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Mortenson, D. E. et al. ‘Inverse Drug Discovery’ strategy to identify proteins that are targeted by latent electrophiles as exemplified by Aryl fluorosulfates. J. Am. Chem. Soc. 140, 200–210 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Fadeyi, O. O. et al. Covalent enzyme inhibition through fluorosulfate modification of a noncatalytic serine residue. ACS Chem. Biol. 12, 2015–2020 (2017).

    Article  CAS  PubMed  Google Scholar 

  28. Liu, Z. et al. SuFEx click chemistry enabled late-stage drug functionalization. J. Am. Chem. Soc. 140, 2919–2925 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Adibekian, A. et al. Click-generated triazole ureas as ultrapotent in vivo-active serine hydrolase inhibitors. Nat. Chem. Biol. 7, 469–478 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ahn, K. et al. Discovery of a selective covalent inhibitor of lysophospholipase-like 1 (LYPLAL1) as a tool to evaluate the role of this serine hydrolase in metabolism. ACS Chem. Biol. 11, 2529–2540 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Bern, M., Kil, Y. J. & Becker, C. Byonic: advanced peptide and protein identification software. Curr. Protoc. Bioinforma. 13, Unit13 20 (2012).

    Google Scholar 

  32. Hornbeck, P. V. et al. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res. 43, D512–D520 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Wishart, D. S. et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 46, D1074–D1082 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. Hentze, M. W., Castello, A., Schwarzl, T. & Preiss, T. A brave new world of RNA-binding proteins. Nat. Rev. Mol. Cell Biol. 19, 327–341 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Yaffe, M. B. Phosphotyrosine-binding domains in signal transduction. Nat. Rev. Mol. Cell Biol. 3, 177–186 (2002).

    Article  CAS  PubMed  Google Scholar 

  36. Shin, M., Franks, C. E. & Hsu, K. L. Isoform-selective activity-based profiling of ERK signaling. Chem. Sci. 9, 2419–2431 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Choi, E. J., Jung, D., Kim, J. S., Lee, Y. & Kim, B. M. Chemoselective tyrosine bioconjugation through sulfate click reaction. Chemistry 24, 10948–10952 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Shannon, D. A. et al. Investigating the proteome reactivity and selectivity of aryl halides. J. Am. Chem. Soc. 136, 3330–3333 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Humphrey, S. J. et al. Dynamic adipocyte phosphoproteome reveals that Akt directly regulates mTORC2. Cell Metab. 17, 1009–1020 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Lundby, A. et al. Quantitative maps of protein phosphorylation sites across 14 different rat organs and tissues. Nat. Commun. 3, 876 (2012).

    Article  PubMed  CAS  Google Scholar 

  41. Hilger, M., Bonaldi, T., Gnad, F. & Mann, M. Systems-wide analysis of a phosphatase knock-down by quantitative proteomics and phosphoproteomics. Mol. Cell Proteom. 8, 1908–1920 (2009).

    Article  CAS  Google Scholar 

  42. Song, G. et al. Proteome-wide tyrosine phosphorylation analysis reveals dysregulated signaling pathways in ovarian tumors. Mol. Cell Proteom. 18, 448–460 (2019).

    Article  CAS  Google Scholar 

  43. Song, L., Turkson, J., Karras, J. G., Jove, R. & Haura, E. B. Activation of Stat3 by receptor tyrosine kinases and cytokines regulates survival in human non-small cell carcinoma cells. Oncogene 22, 4150–4165 (2003).

    Article  CAS  PubMed  Google Scholar 

  44. Hong, J. Y., Oh, I. H. & McCrea, P. D. Phosphorylation and isoform use in p120-catenin during development and tumorigenesis. Biochim Biophys. Acta 1863, 102–114 (2016).

    Article  CAS  PubMed  Google Scholar 

  45. Hitosugi, T. et al. Tyrosine phosphorylation inhibits PKM2 to promote the Warburg effect and tumor growth. Sci. Signal 2, ra73 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Weerapana, E., Simon, G. M. & Cravatt, B. F. Disparate proteome reactivity profiles of carbon electrophiles. Nat. Chem. Biol. 4, 405–407 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Manley, J. L. & Krainer, A. R. A rational nomenclature for serine/arginine-rich protein splicing factors (SR proteins). Genes Dev. 24, 1073–1074 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Hargous, Y. et al. Molecular basis of RNA recognition and TAP binding by the SR proteins SRp20 and 9G8. EMBO J. 25, 5126–5137 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Harris, T. K. & Turner, G. J. Structural basis of perturbed pKa values of catalytic groups in enzyme active sites. IUBMB Life 53, 85–98 (2002).

    Article  CAS  PubMed  Google Scholar 

  50. Decker, C. J. & Parker, R. P-bodies and stress granules: possible roles in the control of translation and mRNA degradation. Cold Spring Harb. Perspect. Biol. 4, a012286 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Franks, C. E., Campbell, S. T., Purow, B. W., Harris, T. E. & Hsu, K. L. The ligand binding landscape of diacylglycerol kinases. Cell Chem. Biol. 24, 870–880 e875 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Chen, H. & Boutros, P. C. VennDiagram: a package for the generation of highly-customizable Venn and Euler diagrams in R. BMC Bioinforma. 12, 35 (2011).

    Article  Google Scholar 

  53. Sigrist, C. J. et al. New and continuing developments at PROSITE. Nucleic Acids Res. 41, D344–D347 (2013).

    Article  CAS  PubMed  Google Scholar 

  54. Mi, H., Muruganujan, A., Casagrande, J. T. & Thomas, P. D. Large-scale gene function analysis with the PANTHER classification system. Nat. Protoc. 8, 1551–1566 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Bereman, M. S. et al. An automated pipeline to monitor system performance in liquid chromatography-tandem mass spectrometry proteomic experiments. J. Proteome Res. 15, 4763–4769 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Wickham, H. ggplot2: Elegant Graphics for Data Analysis 2nd edn (Springer International Publishing, 2016).

Download references

Acknowledgements

We thank all members of the Hsu Laboratory and colleagues at the University of Virginia for helpful discussions and review of the manuscript. We thank M. Ross for his assistance with mass spectrometry experiments and data analysis. We thank S. Campbell for assistance with molecular biology experiments. We thank D. Dickie for assistance with small molecule crystallography studies. We thank R. Rumana for assistance with cell culture studies. This work was supported by the University of Virginia (start-up funds to K.-L.H.), University of Virginia Cancer Center (A.H.L. and K.-L.H), National Institutes of Health grant nos. GM801868 (T.B.W.), GM007055 (J.W.B.), CA009109 (A.L.B.) and DA043571 (K.-L.H.), the Schiff Foundation (K.-L.H.), the Wagner Fellowship (A.L.B), the National Science Foundation Graduate Research Fellowship (grant no. 2018255830 to R.L.M.) and the U.S. Department of Defense (no. W81XWH-17-1-0487 to K.-L.H.).

Author information

Authors and Affiliations

Authors

Contributions

H.S.H., E.K.T., A.L.B., J.W.B. and K.-L.H. conceived of the project, designed experiments and analyzed data. H.S.H. and E.K.T. performed mass spectrometry experiments and data analysis. A.L.B. wrote software and performed bioinformatics analysis. H.S.H., J.W.B. and K.Y. synthesized compounds. J.W.B. expressed proteins, conducted inhibition studies and performed biochemical assays. E.K.T. and J.W.B. conducted cellular studies. A.H.L. assisted with compound synthesis and characterization. T.B.W., A.M.C. and R.L.M. performed site-directed mutagenesis and assisted with cloning and expression of proteins. H.S.H., E.K.T., A.L.B., J.W.B. and K.-L.H. wrote the manuscript.

Corresponding author

Correspondence to Ku-Lung Hsu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Figs. 1–31

Reporting Summary

Supplementary Dataset 1

Supplementary Note

Synthetic Procedures

X-ray crystal structures of compounds 2, 3, and 6

Individual CIF files for HHS-465, HHS-475 and HHS-483 crystal structures combined into a single CIF file.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hahm, H.S., Toroitich, E.K., Borne, A.L. et al. Global targeting of functional tyrosines using sulfur-triazole exchange chemistry. Nat Chem Biol 16, 150–159 (2020). https://doi.org/10.1038/s41589-019-0404-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-019-0404-5

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research