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:

S-glycosylation-based cysteine profiling reveals regulation of glycolysis by itaconate

Abstract

Itaconate has been recently recognized as an anti-inflammatory metabolite involved in the pathogen–macrophage interface. Due to its weak electrophilicity, itaconate could modify cysteines of the protein KEAP1 and glutathione, which contribute to its anti-inflammatory effect. However, the substrates of itaconate modification in macrophages have not been systematically profiled, which largely impedes the understanding of its roles in immune responses. Here, we developed a specific thiol-reactive probe, 1-OH-Az, for quantitative chemoproteomic profiling of cysteine modifications by itaconate, and provided a global portrait of its proteome reactivity. We found that itaconate covalently modifies key glycolytic enzymes and impairs glycolytic flux mainly through inhibition of fructose-bisphosphate aldolase A (ALDOA). Moreover, itaconate attenuates the inflammatory response in stimulated macrophages by impairing the glycolysis. Our study provides a valuable resource of protein targets of itaconate in macrophages and establishes a negative-feedback link between glycolysis and itaconate, elucidating new functional insights for this anti-inflammatory metabolite.

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: Development of 1-OH-Az as an efficient and unique probe for cysteinome profiling based on its S-glycosylation reactivity.
Fig. 2: Chemoproteomic profiling of itaconate-modified cysteines by competitive isoTOP-ABPP using 1-OH-Az.
Fig. 3: Itaconate modifies and inhibits ALDOA.
Fig. 4: Itaconate impairs glycolysis through modifying ALDOA.
Fig. 5: Itaconate’s anti-inflammatory effect is mediated by impairing glycolysis through ALDOA inhibition.

Similar content being viewed by others

Data availability

The proteomics data (ID: 206029, 206030, 206031) has been deposited at https://chorusproject.org/pages/index.html. The data that support the findings of this study are available from the corresponding authors upon request.

References

  1. Kelly, B. & O’Neill, L. A. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 25, 771–784 (2015).

    Article  Google Scholar 

  2. Vats, D. et al. Oxidative metabolism and PGC-1beta attenuate macrophage-mediated inflammation. Cell Metab. 4, 13–24 (2006).

    Article  CAS  Google Scholar 

  3. Palsson-McDermott, E. M. et al. Pyruvate kinase M2 regulates Hif-1α activity and IL-1β induction and is a critical determinant of the Warburg effect in LPS-activated macrophages. Cell Metab. 21, 65–80 (2015).

    Article  CAS  Google Scholar 

  4. Van den Bossche, J., O’Neill, L. A. & Menon, D. Macrophage immunometabolism: where are we (going)? Trends Immun. 38, 395–406 (2017).

    Article  Google Scholar 

  5. Kornberg, M. D. et al. Dimethyl fumarate targets GAPDH and aerobic glycolysis to modulate immunity. Science 360, 449–453 (2018).

    Article  CAS  Google Scholar 

  6. Tannahill, G. M. et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature 496, 238–242 (2013).

    Article  CAS  Google Scholar 

  7. Murphy, M. P. & O’Neill, L. A. J. Krebs cycle reimagined: the emerging roles of succinate and itaconate as signal transducers. Cell 174, 780–784 (2018).

    Article  CAS  Google Scholar 

  8. Lampropoulou, V. et al. Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metab. 24, 158–166 (2016).

    Article  CAS  Google Scholar 

  9. Dominguez-Andres, J. et al. The itaconate pathway is a central regulatory node linking innate immune tolerance and trained immunity. Cell Metab. 29, 211–220 (2019).

    Article  CAS  Google Scholar 

  10. Michelucci, A. et al. Immune-responsive gene 1 protein links metabolism to immunity by catalyzing itaconic acid production. Proc. Natl Acad. Sci. USA 110, 7820–7825 (2013).

    Article  CAS  Google Scholar 

  11. 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 

  12. Sasikaran, J., Ziemski, M., Zadora, P. K., Fleig, A. & Berg, I. A. Bacterial itaconate degradation promotes pathogenicity. Nat. Chem. Biol. 10, 371–377 (2014).

    Article  CAS  Google Scholar 

  13. Shen, H. et al. The human knockout gene CLYBL connects itaconate to vitamin B12. Cell 171, 771–782 (2017).

    Article  CAS  Google Scholar 

  14. O’Neill, L. A. J. & Artyomov, M. N. Itaconate: the poster child of metabolic reprogramming in macrophage function. Nat. Rev. Immunol. 19, 273–281 (2019).

    Article  Google Scholar 

  15. Mills, E. L. et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature 556, 113–117 (2018).

    Article  CAS  Google Scholar 

  16. Bambouskova, M. et al. Electrophilic properties of itaconate and derivatives regulate the IκBζ-ATF3 inflammatory axis. Nature 556, 501–504 (2018).

    Article  CAS  Google Scholar 

  17. Nomura, D. K., Dix, M. M. & Cravatt, B. F. Activity-based protein profiling for biochemical pathway discovery in cancer. Nat. Rev. Cancer 10, 630–638 (2010).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  19. Weerapana, E. et al. Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 468, 790–U779 (2010).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  21. Abegg, D. et al. Proteome-Wide profiling of targets of cysteine reactive small molecules by using ethynyl benziodoxolone reagents. Angew. Chem. Int. Edit. 54, 10852–10857 (2015).

    Article  CAS  Google Scholar 

  22. Akter, S. et al. Chemical proteomics reveals new targets of cysteine sulfinic acid reductase. Nat. Chem. Biol. 14, 995–1004 (2018).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  24. 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 

  25. Gupta, V., Yang, J., Liebler, D. C. & Carroll, K. S. Diverse redoxome reactivity profiles of carbon nucleophiles. J. Am. Chem. Soc. 139, 5588–5595 (2017).

    Article  CAS  Google Scholar 

  26. Lentz, C. S. et al. Identification of a S. aureus virulence factor by activity-based protein profiling (ABPP). Nat. Chem. Biol. 14, 609–617 (2018).

    Article  CAS  Google Scholar 

  27. 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 

  28. Kulkarni, R. A. et al. A chemoproteomic portrait of the oncometabolite fumarate. Nat. Chem. Biol. 15, 391–400 (2019).

    Article  CAS  Google Scholar 

  29. Counihan, J. L., Wiggenhorn, A. L., Anderson, K. E. & Nomura, D. K. Chemoproteomics-enabled covalent ligand screening reveals ALDH3A1 as a lung cancer therapy target. ACS Chem. Biol. 13, 1970–1977 (2018).

    Article  CAS  Google Scholar 

  30. Tian, C. et al. Multiplexed thiol reactivity profiling for target discovery of electrophilic natural products. Cell Chem. Biol. 24, 1416–1427 (2017).

    Article  CAS  Google Scholar 

  31. Blewett, M. M. et al. Chemical proteomic map of dimethyl fumarate-sensitive cysteines in primary human T cells. Sci. Signal 9, 445 (2016).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  33. Qin, W. et al. Artificial cysteine S-glycosylation induced by per-O-acetylated unnatural monosaccharides during metabolic glycan labeling. Angew. Chem. Int. Edn 57, 1817–1820 (2018).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  35. Kolb, H. C., Finn, M. G. & Sharpless, K. B. Click chemistry: diverse chemical function from a few good reactions. Angew. Chem. Int. Edn 40, 2004–2021 (2001).

    Article  CAS  Google Scholar 

  36. Qin, K. et al. Quantitative profiling of protein O-GlcNAcylation sites by an isotope-tagged cleavable linker. ACS Chem. Bio. 13, 1983–1989 (2018).

    Article  CAS  Google Scholar 

  37. O’Shea, J. P. et al. pLogo: a probabilistic approach to visualizing sequence motifs. Nat. Methods 10, 1211–1212 (2013).

    Article  Google Scholar 

  38. Yao, D. C. et al. Hemolytic anemia and severe rhabdomyolysis caused by compound heterozygous mutations of the gene for erythrocyte/muscle isozyme of aldolase, ALDOA((Arg303X/Cys338Tyr)). Blood 103, 2401–2403 (2004).

    Article  CAS  Google Scholar 

  39. Jang, C., Chen, L. & Rabinowitz, J. D. Metabolomics and isotope tracing. Cell 173, 822–837 (2018).

    Article  CAS  Google Scholar 

  40. Fu, J. et al. Hyperactivity of the transcription factor Nrf2 causes metabolic reprogramming in mouse esophagus. J. Bio. Chem. 294, 327–340 (2018).

    Article  Google Scholar 

  41. Singh, A. et al. Small molecule inhibitor of NRF2 selectively intervenes therapeutic resistance in KEAP1-Deficient NSCLC tumors. ACS Chem. Biol. 11, 3214–3225 (2016).

    Article  CAS  Google Scholar 

  42. Puchalska, P. et al. Isotope tracing untargeted metabolomics reveals macrophage polarization-state-specific metabolic coordination across intracellular compartments. iScience 9, 298–313 (2018).

    Article  CAS  Google Scholar 

  43. Qin, W. et al. Quantitative time-resolved chemoproteomics reveals that stable O-GlcNAc regulates box C/D snoRNP biogenesis. Proc. Natl Acad. Sci. USA 114, E6749–E6758 (2017).

    Article  CAS  Google Scholar 

  44. Tarbet, H. J., Toleman, C. A. & Boyce, M. A sweet embrace: control of protein-protein interactions by O-Linked beta-N-Acetylglucosamine. Biochemistry 57, 13–21 (2018).

    Article  CAS  Google Scholar 

  45. Chen, Y. et al. Quantitative profiling of protein carbonylations in ferroptosis by an aniline-derived probe. J. Am. Chem. Soc. 140, 4712–4720 (2018).

    Article  CAS  Google Scholar 

  46. Yang, J., Tallman, K. A., Porter, N. A. & Liebler, D. C. Quantitative chemoproteomics for site-specific analysis of protein alkylation by 4-hydroxy-2-nonenal in cells. Anal. Chem. 87, 2535–2541 (2015).

    Article  CAS  Google Scholar 

  47. Isobe, Y. et al. Identification of protein targets of 12/15-Lipoxygenase-derived lipid electrophiles in mouse peritoneal macrophages using omega-alkynyl fatty acid. ACS Chem. Biol. 13, 887–893 (2018).

    Article  CAS  Google Scholar 

  48. Moellering, R. E. & Cravatt, B. F. Functional lysine modification by an intrinsically reactive primary glycolytic metabolite. Science 341, 549–553 (2013).

    Article  CAS  Google Scholar 

  49. Bollong, M. J. et al. A metabolite-derived protein modification integrates glycolysis with KEAP1-NRF2 signalling. Nature 562, 600–604 (2018).

    Article  CAS  Google Scholar 

  50. Huang, H. et al. EP300-mediated lysine 2-hydroxyisobutyrylation regulates glycolysis. Mol. Cell 70, 663–678 (2018).

    Article  CAS  Google Scholar 

  51. Yang, F., Gao, J., Che, J., Jia, G. & Wang, C. A dimethyl-labeling-based strategy for site-specifically quantitative chemical proteomics. Anal. Chem. 90, 9576–9582 (2018).

    Article  CAS  Google Scholar 

  52. Wisniewski, J. R., Zougman, A., Nagaraj, N. & Mann, M. Universal sample preparation method for proteome analysis. Nat. Methods 6, 359–362 (2009).

    Article  CAS  Google Scholar 

  53. Kulevich, S. E., Frey, B. L., Kreitinger, G. & Smith, L. M. Alkylating tryptic peptides to enhance electrospray ionization mass spectrometry analysis. Anal. Chem. 82, 10135–10142 (2010).

    Article  CAS  Google Scholar 

  54. Saxon, E. & Bertozzi, C. R. Cell surface engineering by a modified Staudinger reaction. Science 287, 2007–2010 (2000).

    Article  CAS  Google Scholar 

  55. Wang, H. et al. Selective in vivo metabolic cell-labeling-mediated cancer targeting. Nat. Chem. Biol. 13, 415–424 (2017).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the Computing Platform of the Center for Life Science for supporting the proteomics data analysis and Metabolomics Facility Center in National Protein Science Technology Center of Tsinghua University for C13 isotope tracing experiments. This work is supported by the National Key Research and Development Projects (grant no. 2016YFA0501500) to C.W and X.C., and (no. 2018YFA0507600) to X.C., and the National Natural Science Foundation of China (nos. 21521003, 81490740 and 21778004) to C.W. and (nos. 21425204, 21521003 and 21672013) to X.C., and a ‘1,000 Talents Plan’ Young Investigator Award to C.W.

Author information

Authors and Affiliations

Authors

Contributions

W.Q., X.C. and C.W. conceived the project. W.Q. conducted most of the experiments unless specified otherwise. K.Q. carried out the LC–MS/MS analysis. Y.Z. carried out the gene cloning. W.J. cultured BMDMs under the guidance of Y-L.W. Y.C. performed IA-alkyne-based rdTOP-ABPP. B.C. synthesized 1-OH-Az and its derivatives. L.P. helped evaluate the 1-OH-Az labeling. N.C. synthesized UK-alkyne probe. Y.L. analyzed ALDOA structure and W.Z. performed ETD-based LC–MS/MS analysis. W.Q., X.C. and C.W. analyzed the data and wrote the manuscript with input from all the authors.

Corresponding authors

Correspondence to Xing Chen or Chu Wang.

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. 1–14

Reporting Summary

Dataset 1

Identification of the cysteines modified by 1-OH-Az and Ac4ManNAz in MCF-7 cells.

Dataset 2

Identification of the proteins modified by 1-OH-Az and Ac4ManNAz in MCF-7 cells.

Dataset 3

Competitive isoTOP-ABPP profiling of itaconate-modified cysteines using the 1-OH-Az probe in Raw264.7 cells with 250 μM of itaconate treatment.

Dataset 4

Competitive isoTOP-ABPP profiling of itaconate-modified cysteines using the 1-OH-Az probe in Raw264.7 cells with 1 mM of itaconate treatment.

Dataset 5

Competitive rdTOP-ABPP profiling of itaconate-modified cysteines using the IA-alkyne probe in Raw264.7 cells.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qin, W., Qin, K., Zhang, Y. et al. S-glycosylation-based cysteine profiling reveals regulation of glycolysis by itaconate. Nat Chem Biol 15, 983–991 (2019). https://doi.org/10.1038/s41589-019-0323-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-019-0323-5

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