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:

Direct mapping of ligandable tyrosines and lysines in cells with chiral sulfonyl fluoride probes

Abstract

Advances in chemoproteomic technology have revealed covalent interactions between small molecules and protein nucleophiles, primarily cysteine, on a proteome-wide scale. Most chemoproteomic screening approaches are indirect, relying on competition between electrophilic fragments and a minimalist electrophilic probe with inherently limited proteome coverage. Here we develop a chemoproteomic platform for direct electrophile-site identification based on enantiomeric pairs of clickable arylsulfonyl fluoride probes. Using stereoselective site modification as a proxy for ligandability in intact cells, we identify 634 tyrosines and lysines within functionally diverse protein sites, liganded by structurally diverse probes. Among multiple validated sites, we discover a chiral probe that modifies Y228 in the MYC binding site of the epigenetic regulator WDR5, as revealed by a high-resolution crystal structure. A distinct chiral probe stimulates tumour cell phagocytosis by covalently modifying Y387 in the recently discovered immuno-oncology target APMAP. Our work provides a deep resource of ligandable tyrosines and lysines for the development of covalent chemical probes.

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

Access options

Buy this article

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

Fig. 1: Design of ten enantiomeric pairs of chiral sulfonyl fluoride probes to identify ligandable tyrosines and lysines in living cells.
Fig. 2: Global analysis of tyrosines and lysines modified by chiral sulfonyl fluoride probes.
Fig. 3: Chiral SF probes target functionally diverse protein sites.
Fig. 4: (R)-9-SF modifies PARP1 Y889 in cells.
Fig. 5: (R)-2-SF blocks WDR5 binding to MYC.
Fig. 6: Covalent modification of APMAP Y387 by (R)-1-SF induces cancer-cell phagocytosis.

Similar content being viewed by others

Data availability

Uncropped gel images are provided as Source Data files accompanying this paper. The reported crystal structure has been deposited to the Protein Data Bank (PDB) under accession no. 8F93. All proteomic raw data have been deposited to MassIVE (http://massive.ucsd.edu) with accession no. MSV000090778, as well as in ProteomeXchange (http://www.proteomexchange.org) with accession no. PXD042307. Source data are provided with this paper.

Code availability

The script used for identifying modified sites and proximal ligands in the PDB has been deposited to GitHub (https://github.com/aacuesta/Ying2023NatChem).

References

  1. Cross, D. A. et al. AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov. 4, 1046–1061 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Byrd, J. C. et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N. Engl. J. Med. 369, 32–42 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hong, D. S. et al. KRAS(G12C) inhibition with sotorasib in advanced solid tumors. N. Engl. J. Med. 383, 1207–1217 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Cuesta, A., Wan, X., Burlingame, A. L. & Taunton, J. Ligand conformational bias drives enantioselective modification of a surface-exposed lysine on Hsp90. J. Am. Chem. Soc. 142, 3392–3400 (2020).

    Article  CAS  PubMed  Google Scholar 

  5. Wan, X. et al. Discovery of lysine-targeted eIF4E inhibitors through covalent docking. J. Am. Chem. Soc. 142, 4960–4964 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Dalton, S. E. et al. Selectively targeting the kinome-conserved lysine of PI3Kδ as a general approach to covalent kinase inhibition. J. Am. Chem. Soc. 140, 932–939 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Baggio, C. et al. Design of potent pan-IAP and Lys-covalent XIAP selective inhibitors using a thermodynamics driven approach. J. Med. Chem. 61, 6350–6363 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Bum-Erdene, K. et al. Small-molecule covalent bond formation at tyrosine creates a binding site and inhibits activation of Ral GTPases. Proc. Natl Acad. Sci. USA 117, 7131–7139 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Teng, M. et al. Rationally designed covalent BCL6 inhibitor that targets a tyrosine residue in the homodimer interface. ACS Med. Chem. Lett. 11, 1269–1273 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hatcher, J. M. et al. SRPKIN-1: a covalent SRPK1/2 inhibitor that potently converts VEGF from pro-angiogenic to anti-angiogenic isoform. Cell Chem. Biol. 25, 460–470 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Mukherjee, H. et al. Discovery and optimization of covalent Bcl-xL antagonists. Bioorg. Med. Chem. Lett. 29, 126682 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Glukhova, A. et al. Structure of the adenosine A(1) receptor reveals the basis for subtype selectivity. Cell 168, 867–877 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Backus, K. M. et al. Proteome-wide covalent ligand discovery in native biological systems. Nature 534, 570–574 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Brulet, J. W., Borne, A. L., Yuan, K., Libby, A. H. & Hsu, K.-L. Liganding functional tyrosine sites on proteins using sulfur-triazole exchange chemistry. J. Am. Chem. Soc. 142, 8270–8280 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Abbasov, M. E. et al. A proteome-wide atlas of lysine-reactive chemistry. Nat. Chem. 13, 1081–1092 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. 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 

  17. Kuljanin, M. et al. Reimagining high-throughput profiling of reactive cysteines for cell-based screening of large electrophile libraries. Nat. Biotechnol. 39, 630–641 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. McCloud, R. L. et al. Direct target site identification of a sulfonyl-triazole covalent kinase probe by LC-MS chemical proteomics. Anal. Chem. 93, 11946–11955 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wang, Y. et al. Expedited mapping of the ligandable proteome using fully functionalized enantiomeric probe pairs. Nat. Chem. 11, 1113–1123 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Vinogradova, E. V. et al. An activity-guided map of electrophile-cysteine interactions in primary human T cells. Cell 182, 1009–1026 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tao, Y. et al. Targeted protein degradation by electrophilic PROTACs that stereoselectively and site-specifically engage DCAF1. J. Am. Chem. Soc. 144, 18688–18699 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Szychowski, J. et al. Cleavable biotin probes for labeling of biomolecules via azide-alkyne cycloaddition. J. Am. Chem. Soc. 132, 18351–18360 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Rabalski, A. J., Bogdan, A. R. & Baranczak, A. Evaluation of chemically-cleavable linkers for quantitative mapping of small molecule-cysteinome reactivity. ACS Chem. Biol. 14, 1940–1950 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Li, Z., Liu, K., Xu, P. & Yang, J. Benchmarking cleavable biotin tags for peptide-centric chemoproteomics. J Proteome Res. 21, 1349–1358 (2022).

    Article  CAS  PubMed  Google Scholar 

  25. Erickson, B. K. et al. Active instrument engagement combined with a real-time database search for improved performance of sample multiplexing workflows. J. Proteome Res. 18, 1299–1306 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Schweppe, D. K. et al. Full-featured, real-time database searching platform enables fast and accurate multiplexed quantitative proteomics. J. Proteome Res. 19, 2026–2034 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Cruite, J. T. et al. Cereblon covalent modulation through structure-based design of histidine targeting chemical probes. RSC Chem. Biol. 3, 1105–1110 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Tam, J. et al. An activity-based probe for high-throughput measurements of triacylglycerol lipases. Anal. Biochem. 414, 254–260 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Gambini, L. et al. Covalent inhibitors of protein-protein interactions targeting lysine, tyrosine or histidine residues. J. Med. Chem. 62, 5616–5627 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. 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 

  31. Zanon, P. R. A. et al. Profiling the proteome-wide selectivity of diverse electrophiles. Preprint at https://chemrxiv.org/engage/chemrxiv/article-details/60c755f2bb8c1a7d393dc505 (2021).

  32. 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 

  33. Mukherjee, H. et al. A study of the reactivity of S((VI))-F containing warheads with nucleophilic amino-acid side chains under physiological conditions. Org. Biomol. Chem. 15, 9685–9695 (2017).

    Article  CAS  PubMed  Google Scholar 

  34. Brighty, G. J. et al. Using sulfuramidimidoyl fluorides that undergo sulfur(VI) fluoride exchange for inverse drug discovery. Nat. Chem. 12, 906–913 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Stanevich, V. et al. The structural basis for tight control of PP2A methylation and function by LCMT-1. Mol. Cell 41, 331–342 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hwang, J., Lee, J. A. & Pallas, D. C. Leucine carboxyl methyltransferase 1 (LCMT-1) methylates protein phosphatase 4 (PP4) and protein phosphatase 6 (PP6) and differentially regulates the stable formation of different PP4 holoenzymes. J. Biol. Chem. 291, 21008–21019 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. De Baere, I. et al. Purification of porcine brain protein phosphatase 2A leucine carboxyl methyltransferase and cloning of the human homologue. Biochemistry 38, 16539–16547 (1999).

    Article  PubMed  Google Scholar 

  38. Reckel, S. et al. Structural and functional dissection of the DH and PH domains of oncogenic Bcr-Abl tyrosine kinase. Nat. Commun. 8, 2101 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Lee, J. H. & Skalnik, D. G. Wdr82 is a C-terminal domain-binding protein that recruits the Setd1A Histone H3-Lys4 methyltransferase complex to transcription start sites of transcribed human genes. Mol. Cell. Biol. 28, 609–618 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Wu, M. et al. Molecular regulation of H3K4 trimethylation by Wdr82, a component of human Set1/COMPASS. Mol. Cell. Biol. 28, 7337–7344 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Thomas, L. R. et al. Interaction with WDR5 promotes target gene recognition and tumorigenesis by MYC. Mol. Cell 58, 440–452 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chacón Simon, S. et al. Discovery of WD repeat-containing protein 5 (WDR5)–MYC inhibitors using fragment-based methods and structure-based design. J. Med. Chem. 63, 4315–4333 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Tian, J. et al. Discovery and structure-based optimization of potent and selective WD repeat domain 5 (WDR5) inhibitors containing a dihydroisoquinolinone bicyclic core. J. Med. Chem. 63, 656–675 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ding, J. et al. Discovery of potent small-molecule inhibitors of WDR5-MYC interaction. ACS Chem. Biol. 18, 34–40 (2023).

    Article  CAS  PubMed  Google Scholar 

  45. Kamber, R. A. et al. Inter-cellular CRISPR screens reveal regulators of cancer cell phagocytosis. Nature 597, 549–554 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hahm, H. S. et al. Global targeting of functional tyrosines using sulfur-triazole exchange chemistry. Nat. Chem. Biol. 16, 150–159 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. 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 

  48. 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 

  49. Wang, N. et al. Genetically encoding fluorosulfate-l-tyrosine to react with lysine, histidine and tyrosine via SuFEx in proteins in vivo. J. Am. Chem. Soc. 140, 4995–4999 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Hett, E. C. et al. Rational targeting of active-site tyrosine residues using sulfonyl fluoride probes. ACS Chem. Biol. 10, 1094–1098 (2015).

    Article  CAS  PubMed  Google Scholar 

  51. Cuesta, A. & Taunton, J. Lysine-targeted inhibitors and chemoproteomic probes. Annu. Rev. Biochem. 88, 365–381 (2019).

    Article  CAS  PubMed  Google Scholar 

  52. Yang, T. et al. Reversible lysine-targeted probes reveal residence time-based kinase selectivity. Nat. Chem. Biol. 18, 934–941 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Metcalf, B. et al. Discovery of GBT440, an orally bioavailable R-state stabilizer of sickle cell hemoglobin. ACS Med. Chem. Lett. 8, 321–326 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Funding for this study was provided by the Ono Pharma Foundation (J.T.), Pfizer and the Tobacco-Related Disease Research Program Postdoctoral Fellowship Award (T32FT4880 to G.B.C.). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript. We thank B. Küster for guidance on MS data acquisition and analysis.

Author information

Authors and Affiliations

Authors

Contributions

Y.C. and J.T. conceived the project, designed the experiments and analysed the data. S.Z. and Y.S.M. synthesized the SFs. Y.C. performed the chemoproteomic and validation experiments. G.B.C. solved the WDR5 X-ray structure and performed biochemical experiments with recombinant APMAP. A.C. wrote the script for identifying modified sites in the PDB. R.A.K. performed the phagocytosis assays, with input from M.C.B. Y.C. and J.T. wrote the manuscript, with input from all of the authors.

Corresponding author

Correspondence to Jack Taunton.

Ethics declarations

Competing interests

J.T. is a cofounder of Kezar Life Sciences and Terremoto Biosciences, and a scientific advisor to Entos. M.C.B. and R.A.K. have outside interests in DEM Biopharma. S.Z. and Y.S.M. are employees of Enamine Ltd and Chemspace LLC, respectively. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Chemistry thanks Benjamin Cravatt and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Volcano plots showing modified sites (MS2 and RTS-MS3 data) for probes 110.

a-j, Log2ratio((R)/(S)) is on the x-axis and –log10(P value) is on the y-axis. Modified sites are indicated as (R)-selective (blue), (S)-selective (orange), and nonselective (grey). P values were determined by Student’s t-test (two-tailed, two-sample equal variance).

Extended Data Fig. 2 Analysis of tyrosines and lysines modified by chiral sulfonyl fluoride probes.

a-f, Number of liganded and total Tyr and Lys identified in this study compared with the indicated previous studies14,15. g, The number of enantiomeric probe pairs (based on fragments 1-10) per site (x- axis) was plotted against the total number of sites modified by the indicated number of enantiomeric probe pairs (y-axis).

Extended Data Fig. 3 Functionally diverse protein sites targeted by chiral sulfonyl fluoride probes.

a-f, Structures of FUS, YBX1, KIF11, KDM1A, PRMT5, and SIRT2, highlighting the probe-modified Tyr proximal to bound ligands.

Extended Data Fig. 4 Probe modification of LCMT1.

a, HEK293T cells were transfected with Flag-LCMT1 and treated with SF probes (10 μM, 1 h). After cell lysis, anti-Flag enrichment, 3xFlag peptide elution and click reaction with TAMRA-azide, samples were analyzed by in-gel fluorescence and western blotting. NC indicates nontransfected cells. Data are representative of two independent experiments. b, HEK293T cells were transfected with WT or Y315F Flag- LCMT1 and treated with 4-SF probes (10 μM, 1 h). The cells were lysed and processed as described above. Data are representative of two independent experiments.

Source data

Extended Data Fig. 5 Probe modification of ABR.

a, HEK293T cells were transfected with Flag-ABR and treated with SF probes (10 μM, 1 h). After cell lysis, anti-Flag enrichment, 3xFlag peptide elution and click reaction with TAMRA-azide, samples were analyzed by in-gel fluorescence and western blotting. NC indicates nontransfected cells. Data are representative of two independent experiments. b, HEK293T cells were transfected with WT or Y344F Flag-ABR and treated with 7-SF probes (10 μM, 1 h). The cells were lysed and processed as described above. Data are representative of two independent experiments.

Source data

Extended Data Fig. 6 Probe modification of WDR82.

a, HEK293T cells were transfected with WT or Y102F Flag-WDR82 and treated with 7-SF probes (50 μM, 1 h). After cell lysis, anti- Flag enrichment, 3xFlag peptide elution and click reaction with TAMRA-azide, samples were analyzed by in-gel fluorescence and western blotting. Data are representative of two independent experiments. NC indicates nontransfected cells. b, Recombinant WT and Y102F WDR82 (0.44 μM) were incubated with 7-SF probes (40 μM, 37 °C, overnight) and analyzed by intact-protein mass spectrometry. Data are representative of two independent experiments. c, HEK293T cells were transiently transfected with Flag-WDR82 and treated with the indicated SF probes (50 μM, 1 h). The cells were lysed and processed as described above. Data are representative of two independent experiments. d, Overlay of the AlphaFold structure of WDR82 with the crystal structure of WDR5 bound to KANSL1 peptide (PDB: 4CY2).

Source data

Extended Data Fig. 7 Electron density map of (R)-2-SF.

Electron density (2Fo-Fc) map of (R)-2-SF is shown at a contour level of 1σ.

Extended Data Fig. 8 Data collection and refinement statistics.

Data collection and refinement statistics (molecular replacement) for WDR5-(R)-2-SF complex.

Extended Data Fig. 9 Chemoproteomic analysis of 1-SF probes in dose-response mode.

a, HEK293T cells that stably overexpress Flag-APMAP were treated separately with 0, 3.75, 7.5, 15, 30 and 60 µM of (R)-1-SF and (S)-1-SF for 1.5 h at 37 °C. Cell lysates were conjugated with biotin–DADPS-azide via click chemistry. After enrichment and trypsin digestion, probe-modified peptides from each sample were separately labeled with TMT 11-plex reagents. Probe-modified peptides from each sample were eluted with 2% formic acid, combined, and analyzed by LC-MS/MS. b-d, TMT reporter ion intensities were plotted as a function of probe concentration for peptides quantified by MS2 and RTS: APMAP Y387 (b), APMAP K347 (c), and MTHFD1 Y52 (d). e, Crystal structure (PDB: 1DIA) depicting MTHFD1 Y52 proximal to the inhibitor LY249543.

Extended Data Fig. 10 Antibody-dependent phagocytosis assay.

a, APMAPKO Ramos- Cas9 cells (rescued with Flag-APMAP WT, E103A and Y387F) were resuspended in SDS loading buffer containing 100 mM DTT, sonicated and heated (70 °C, 10 min). Samples were analyzed by western blotting. Data are representative of two independent experiments. b, Scheme of phagocytosis assay.

Source data

Supplementary information

Supplementary Information

Supplementary dataset legends, tables, biological methods, synthetic methods, NMR spectra, references.

Reporting Summary

Supplementary Data 1

MS data from TMT6 experiments. (Table 1) Modified sites and enantiomer modification ratios for 10 chiral sulfonyl fluoride probe pairs. (Tables 2–21) Each table displays the MS data, including TMT6 intensity values (Maxquant output), for sites modified by the indicated chiral sulfonyl fluoride probe pair (MS2 and RTS methods). P-values were determined by Student’s t-test (two-tailed, two-sample equal variance).

Supplementary Data 2

MS data from the 1-SF dose-response experiment (Maxquant output), related to Extended Data Fig. 9. (Table 1) MS2 method. (Table 2) RTS method.

Supplementary Data 3

Sequences of oligonucleotides used in this work.

Source data

Source Data Fig. 4

Unprocessed western blots and/or gels.

Source Data Fig. 5

Unprocessed western blots and/or gels.

Source Data Fig. 6

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 4

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 5

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 6

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 10

Unprocessed western blots and/or gels.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Y., Craven, G.B., Kamber, R.A. et al. Direct mapping of ligandable tyrosines and lysines in cells with chiral sulfonyl fluoride probes. Nat. Chem. 15, 1616–1625 (2023). https://doi.org/10.1038/s41557-023-01281-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41557-023-01281-3

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