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:

A chemoproteomic portrait of the oncometabolite fumarate

Abstract

Hereditary cancer disorders often provide an important window into novel mechanisms supporting tumor growth. Understanding these mechanisms thus represents a vital goal. Toward this goal, here we report a chemoproteomic map of fumarate, a covalent oncometabolite whose accumulation marks the genetic cancer syndrome hereditary leiomyomatosis and renal cell carcinoma (HLRCC). We applied a fumarate-competitive chemoproteomic probe in concert with LC–MS/MS to discover new cysteines sensitive to fumarate hydratase (FH) mutation in HLRCC cell models. Analysis of this dataset revealed an unexpected influence of local environment and pH on fumarate reactivity, and enabled the characterization of a novel FH-regulated cysteine residue that lies at a key protein–protein interface in the SWI-SNF tumor-suppressor complex. Our studies provide a powerful resource for understanding the covalent imprint of fumarate on the proteome and lay the foundation for future efforts to exploit this distinct aspect of oncometabolism for cancer diagnosis and therapy.

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: Fumarate is a covalent oncometabolite.
Fig. 2: Global chemoproteomic profiling of FH-regulated cysteine residues
Fig. 3: Analyzing the reactivity and abundance of FH-regulated cysteines.
Fig. 4: Establishing the molecular determinants of fumarate–protein interactions.
Fig. 5: Functional analyses of FH-regulated Cys residues.
Fig. 6: Chemoproteomic discovery of ligandable cysteines upregulated by FH mutation.

Similar content being viewed by others

Data availability

The authors declare that all data supporting the findings of this study are available within the paper and its supplementary information files. The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the identifiers PXD009378 (Supplementary Datasets 14) and PXD009202 (Supplementary Dataset 5). All of the data are accessible in the supplemental data sets (Supplementary Datasets 17) and can further be explored using our web-based resource (https://ccr2.cancer.gov/resources/Cbl/proteomics/fumarate).

References

  1. Launonen, V. et al. Inherited susceptibility to uterine leiomyomas and renal cell cancer. Proc. Natl Acad. Sci. USA 98, 3387–3392 (2001).

    Article  CAS  Google Scholar 

  2. Tomlinson, I. P. et al. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat. Genet. 30, 406–410 (2002).

    Article  CAS  Google Scholar 

  3. Meier, J. L. Metabolic mechanisms of epigenetic regulation. ACS. Chem. Biol. 8, 2607–2621 (2013).

    Article  CAS  Google Scholar 

  4. Isaacs, J. S. et al. HIF overexpression correlates with biallelic loss of fumarate hydratase in renal cancer: novel role of fumarate in regulation of HIF stability. Cancer Cell 8, 143–153 (2005).

    Article  CAS  Google Scholar 

  5. Pollard, P. J. et al. Accumulation of Krebs cycle intermediates and over-expression of HIF1alpha in tumours which result from germline FH and SDH mutations. Hum. Mol. Genet. 14, 2231–2239 (2005).

    Article  CAS  Google Scholar 

  6. MacKenzie, E. D. et al. Cell-permeating α-ketoglutarate derivatives alleviate pseudohypoxia in succinate dehydrogenase-deficient cells. Mol. Cell. Biol. 27, 3282–3289 (2007).

    Article  CAS  Google Scholar 

  7. Sciacovelli, M. et al. Fumarate is an epigenetic modifier that elicits epithelial-to-mesenchymal transition. Nature 537, 544–547 (2016).

    Article  CAS  Google Scholar 

  8. Alderson, N. L. et al. S-(2-Succinyl)cysteine: a novel chemical modification of tissue proteins by a Krebs cycle intermediate. Arch. Biochem. Biophys. 450, 1–8 (2006).

    Article  CAS  Google Scholar 

  9. Sullivan, L. B. et al. The proto-oncometabolite fumarate binds glutathione to amplify ROS-dependent signaling. Mol. Cell 51, 236–248 (2013).

    Article  CAS  Google Scholar 

  10. Sudarshan, S. et al. Fumarate hydratase deficiency in renal cancer induces glycolytic addiction and hypoxia-inducible transcription factor 1α stabilization by glucose-dependent generation of reactive oxygen species. Mol. Cell. Biol. 29, 4080–4090 (2009).

    Article  CAS  Google Scholar 

  11. Kinch, L., Grishin, N. V. & Brugarolas, J. Succination of Keap1 and activation of Nrf2-dependent antioxidant pathways in FH-deficient papillary renal cell carcinoma type 2. Cancer Cell 20, 418–420 (2011).

    Article  CAS  Google Scholar 

  12. Bardella, C. et al. Aberrant succination of proteins in fumarate hydratase-deficient mice and HLRCC patients is a robust biomarker of mutation status. J. Pathol. 225, 4–11 (2011).

    Article  CAS  Google Scholar 

  13. Piroli, G. G. et al. Succination is increased on select proteins in the brainstem of the NADH dehydrogenase (ubiquinone) Fe-S protein 4 (Ndufs4) knockout mouse, a model of leigh syndrome. Mol. Cell. Proteomics 15, 445–461 (2016).

    Article  CAS  Google Scholar 

  14. Adam, J. et al. Fumarate hydratase deletion in pancreatic β cells leads to progressive diabetes. Cell Rep. 20, 3135–3148 (2017).

    Article  CAS  Google Scholar 

  15. Ternette, N. et al. Inhibition of mitochondrial aconitase by succination in fumarate hydratase deficiency. Cell Rep. 3, 689–700 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  18. Schmidt, T. J., Ak, M. & Mrowietz, U. Reactivity of dimethyl fumarate and methylhydrogen fumarate towards glutathione and N-acetyl-l-cysteine–preparation of S-substituted thiosuccinic acid esters. Bioorg. Med. Chem. 15, 333–342 (2007).

    Article  CAS  Google Scholar 

  19. Blewett, M. M. et al. Chemical proteomic map of dimethyl fumarate-sensitive cysteines in primary human T cells. Sci. Signal. 9, rs10 (2016).

    Article  Google Scholar 

  20. Zhou, Y. et al. Chemoproteomic strategy to quantitatively monitor transnitrosation uncovers functionally relevant S-nitrosation sites on cathepsin D and HADH2. CellChem. Biol. 23, 727–737 (2016).

    CAS  Google Scholar 

  21. Yang, Y. et al. UOK 262 cell line, fumarate hydratase deficient (FH /FH ) hereditary leiomyomatosis renal cell carcinoma: in vitro and in vivo model of an aberrant energy metabolic pathway in human cancer. Cancer Genet. Cytogenet. 196, 45–55 (2010).

    Article  CAS  Google Scholar 

  22. Zheng, L. et al. Fumarate induces redox-dependent senescence by modifying glutathione metabolism. Nat. Commun. 6, 6001 (2015).

    Article  CAS  Google Scholar 

  23. Bar-Peled, L. et al. Chemical proteomics identifies druggable vulnerabilities in a genetically defined cancer. Cell 171, 696–709.e623 (2017).

    Article  CAS  Google Scholar 

  24. Poole, L. B. The basics of thiols and cysteines in redox biology and chemistry. Free Radic. Biol. Med. 80, 148–157 (2015).

    Article  CAS  Google Scholar 

  25. Wang, C., Weerapana, E., Blewett, M. M. & Cravatt, B. F. A chemoproteomic platform to quantitatively map targets of lipid-derived electrophiles. Nat. Methods 11, 79–85 (2014).

    Article  Google Scholar 

  26. Zengeya, T. T. et al. Co-opting a bioorthogonal reaction for oncometabolite Detection. J. Am. Chem. Soc. 138, 15813–15816 (2016).

    Article  CAS  Google Scholar 

  27. Reisz, J. A., Bechtold, E., King, S. B., Poole, L. B. & Furdui, C. M. Thiol-blocking electrophiles interfere with labeling and detection of protein sulfenic acids. FEBS. J. 280, 6150–6161 (2013).

    Article  CAS  Google Scholar 

  28. Doorn, J. A. & Petersen, D. R. Covalent modification of amino acid nucleophiles by the lipid peroxidation products 4-hydroxy-2-nonenal and 4-oxo-2-nonenal. Chem. Res. Toxicol. 15, 1445–1450 (2002).

    Article  CAS  Google Scholar 

  29. Winterbourn, C. C. & Hampton, M. B. Thiol chemistry and specificity in redox signaling. Free Radic. Biol. Med. 45, 549–561 (2008).

    Article  CAS  Google Scholar 

  30. Reva, B., Antipin, Y. & Sander, C. Predicting the functional impact of protein mutations: application to cancer genomics. Nucleic Acids Res. 39, e118 (2011).

    Article  CAS  Google Scholar 

  31. DelBove, J. et al. Identification of a core member of the SWI/SNF complex, BAF155/SMARCC1, as a human tumor suppressor gene. Epigenetics. 6, 1444–1453 (2011).

    Article  CAS  Google Scholar 

  32. Brugarolas, J. PBRM1 and BAP1 as novel targets for renal cell carcinoma. Cancer J. 19, 324–332 (2013).

    Article  CAS  Google Scholar 

  33. Sohn, D. H. et al. SRG3 interacts directly with the major components of the SWI/SNF chromatin remodeling complex and protects them from proteasomal degradation. J. Biol. Chem. 282, 10614–10624 (2007).

    Article  CAS  Google Scholar 

  34. Yan, L., Xie, S., Du, Y. & Qian, C. Structural insights into BAF47 and BAF155 complex formation. J. Mol. Biol. 429, 1650–1660 (2017).

    Article  CAS  Google Scholar 

  35. Keppler, B. R. & Archer, T. K. Ubiquitin-dependent and ubiquitin-independent control of subunit stoichiometry in the SWI/SNF complex. J. Biol. Chem. 285, 35665–35674 (2010).

    Article  CAS  Google Scholar 

  36. Knutson, S. K. et al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc. Natl Acad. Sci. USA 110, 7922–7927 (2013).

    Article  CAS  Google Scholar 

  37. Harmel, R. & Fiedler, D. Features and regulation of non-enzymatic post-translational modifications. Nat. Chem. Biol. 14, 244–252 (2018).

    Article  CAS  Google Scholar 

  38. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  Google Scholar 

  39. Erez, A. & DeBerardinis, R. J. Metabolic dysregulation in monogenic disorders and cancer - finding method in madness. Nat. Rev. Cancer 15, 440–448 (2015).

    Article  CAS  Google Scholar 

  40. Yang, J., Gupta, V., Carroll, K. S. & Liebler, D. C. Site-specific mapping and quantification of protein S-sulphenylation in cells. Nat. Commun. 5, 4776 (2014).

    Article  CAS  Google Scholar 

  41. Strelko, C. L. et al. Itaconic acid is a mammalian metabolite induced during macrophage activation. J. Am. Chem. Soc. 133, 16386–16389 (2011).

    Article  CAS  Google Scholar 

  42. Kulkarni, R. A. et al. Discovering targets of non-enzymatic acylation by thioester reactivity profiling. Cell Chem. Biol. 24, 231–242 (2017).

    Article  CAS  Google Scholar 

  43. Wagner, G. R. et al. A class of reactive acyl-CoA species reveals the non-enzymatic origins of protein acylation. Cell. Metab. 25, 823–837.e828 (2017).

    Article  CAS  Google Scholar 

  44. Frizzell, N., Thomas, S. A., Carson, J. A. & Baynes, J. W. Mitochondrial stress causes increased succination of proteins in adipocytes in response to glucotoxicity. Biochem. J. 445, 247–254 (2012).

    Article  CAS  Google Scholar 

  45. Agaimy, A. et al. SWI/SNF protein expression status in fumarate hydratase-deficient renal cell carcinoma: immunohistochemical analysis of 32 tumors from 28 patients. Hum. Pathol. 77, 139–146 (2018).

    Article  CAS  Google Scholar 

  46. Reddie, K. G. & Carroll, K. S. Expanding the functional diversity of proteins through cysteine oxidation. Curr. Opin. Chem. Biol. 12, 746–754 (2008).

    Article  CAS  Google Scholar 

  47. Li, R. & Kast, J. Biotin switch assays for quantitation of reversible cysteine Oxidation. Methods Enzymol. 585, 269–284 (2017).

    Article  CAS  Google Scholar 

  48. Jost, M. & Weissman, J. S. CRISPR approaches to small molecule target identification. ACS. Chem. Biol. 13, 366–375 (2018).

    Article  CAS  Google Scholar 

  49. Candiano, G. et al. Blue silver: a very sensitive colloidal Coomassie G-250 staining for proteome analysis. Electrophoresis. 25, 1327–1333 (2004).

    Article  CAS  Google Scholar 

  50. Eng, J. K., McCormack, A. L. & Yates, J. R. An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J. Am. Soc. Mass. Spectrom. 5, 976–989 (1994).

    Article  CAS  Google Scholar 

  51. Tabb, D. L., McDonald, W. H. & Yates, J. R. III. DTASelect and Contrast: tools for assembling and comparing protein identifications from shotgun proteomics. J. Proteome. Res. 1, 21–26 (2002).

    Article  CAS  Google Scholar 

  52. Bak, D. W., Pizzagalli, M. D. & Weerapana, E. Identifying functional cysteine residues in the mitochondria. ACS. Chem. Biol. 12, 947–957 (2017).

    Article  CAS  Google Scholar 

  53. Lee, B. & Richards, F. M. The interpretation of protein structures: estimation of static accessibility. J. Mol. Biol. 55, 379–400 (1971).

    Article  CAS  Google Scholar 

  54. Florens, L. & Washburn, M. P. Proteomic analysis by multidimensional protein identification technology. Methods Mol. Biol. 328, 159–175 (2006).

    CAS  PubMed  Google Scholar 

  55. Zhang, Y., Wen, Z., Washburn, M. P. & Florens, L. Effect of dynamic exclusion duration on spectral count based quantitative proteomics. Anal. Chem. 81, 6317–6326 (2009).

    Article  CAS  Google Scholar 

  56. Zhang, Y., Wen, Z., Washburn, M. P. & Florens, L. Improving proteomics mass accuracy by dynamic offline lock mass. Anal. Chem. 83, 9344–9351 (2011).

    Article  CAS  Google Scholar 

  57. Xu, T. et al. ProLuCID: An improved SEQUEST-like algorithm with enhanced sensitivity and specificity. J. Proteomics. 129, 16–24 (2015).

    Article  CAS  Google Scholar 

  58. Zhang, Y., Wen, Z., Washburn, M. P. & Florens, L. Refinements to label free proteome quantitation: how to deal with peptides shared by multiple proteins. Anal. Chem. 82, 2272–2281 (2010).

    Article  CAS  Google Scholar 

  59. Anders, S., Pyl, P. T. & Huber, W. HTSeq--a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  Google Scholar 

  60. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  Google Scholar 

  61. Alexander Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2012).

    Article  Google Scholar 

  62. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  Google Scholar 

Download references

Acknowledgements

The authors thank C. Grose (Protein Expression Laboratory) for cloning and preparation of plasmid DNA, T. Archer (NIEHS) for the gift of the SMARCC1 and SNF5 plasmids, B. Weinberg (MIT) for the gift of the pLKO.1 puro plasmid (Addgene plasmid # 8453), A. Roberts, J. Garlick, and T. Zengeya (NCI) for assisting with preliminary studies. This work was supported by the Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research (ZIA BC011488-05, ZIA BC011038-10) and the CCR FLEX Program. Support for E.W. was provided by the NIH (1R01GM117004 and 1R01GM118431-01A1).

Author information

Authors and Affiliations

Authors

Contributions

R.A.K., D.W.B., E.W. and J.L.M. designed experiments. R.A.K. and D.W.B. performed all chemical proteomic labeling and enrichment experiments. D.W.B. and E.W. performed all LC–MS/MS studies and relative stoichiometry analyses of IA-alkyne enrichment experiments. R.A.K. and A.E.B. synthesized compounds. R.A.K., S.E.B. and J.H.S. performed all cell-based analyses and co-immunoprecipitation experiments. C.A.B. assisted with cell-based analyses and performed S-succination reversibility studies. J.L.M., D.W.B., A.L.T. and R.A.K. performed bioinformatics analyses. D.W. and W.M.L. performed HLRCC spheroid growth inhibition studies and assisted with SWI–SNF analyses. A.A. and E.C.D. performed glycerol gradient fractionation analysis of the SWI–SNF complex and SNF5-dependent gene expression in HLRCC cell lines. N.F. provided the S-succination antibody and literature analysis. W.M.L. and D.R.C. provided HLRCC cell lines and advised experimental design. M.P.W., L.F. and M.L. performed whole-proteome MudPIT S-succination analyses of HLRCC cells. R.A.K. and J.L.M. analyzed data and wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Jordan L. Meier.

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 Text and Figures

Supplementary Figures 1–10

Reporting Summary

Supplementary Note 1

Synthetic Procedures

Supplementary Dataset 1

FH-regulated cysteines identified by comparative profiling of FH−/− HLRCC cell line (UOK262) and a FH+/+ rescue HLRCC cell line (UOK262WT); n = 3 independent experiments.

Supplementary Dataset 2

Compiled list of S-succinated cysteine residues previously characterized in the literature, and annotation with chemoproteomic data (if available).

Supplementary Dataset 3

Sequences used for motif analysis, as well as results for analyses of conservation based functional impact (FI), gene ontology (GO), and genomic lesions found in covalent fumarate targets in kidney cancer.

Supplementary Dataset 4

Fumarate-sensitive cysteines identified by competitive profiling of HEK-293 cells treated and untreated with fumarate (1 mM); n = 4 independent experiments.

Supplementary Dataset 5

Peptides identified as targets of S-succination in MudPIT LC–MS/MS analyses of HLRCC cell (UOK262 and UOK268) proteomes.

Supplementary Dataset 6

Analysis of transcripts co-regulated by FH and SNF5 in publicly accessible RNA-seq datasets.

Supplementary Dataset 7

Solvent-exposed surface area analysis of FH-regulated (Supplementary Dataset 1), exogenous fumarate sensitive (Supplementary Dataset 4), and hyperreactive cysteines.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kulkarni, R.A., Bak, D.W., Wei, D. et al. A chemoproteomic portrait of the oncometabolite fumarate. Nat Chem Biol 15, 391–400 (2019). https://doi.org/10.1038/s41589-018-0217-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-018-0217-y

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer