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.

  • Review Article
  • Published:

Coupling Krebs cycle metabolites to signalling in immunity and cancer

Abstract

Metabolic reprogramming has become a key focus for both immunologists and cancer biologists, with exciting advances providing new insights into the mechanisms underlying disease. There is now extensive evidence that intermediates and derivatives of the mitochondrial Krebs cycle—metabolites traditionally associated with bioenergetics or biosynthesis—also possess ‘non-metabolic’ signalling functions. In this review, we summarize the non-metabolic signalling mechanisms of succinate, fumarate, itaconate, 2-hydroxyglutarate isomers (d-2-hydroxyglutarate and l-2-hydroxyglutarate) and acetyl-CoA, with a specific focus on how such signalling pathways alter immune cell and transformed cell function. We believe that the insights gained from immune and cancer cells that are summarized here will also be useful for understanding and treating a range of other diseases.

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: The Krebs cycle.
Fig. 2: The diverse signalling roles of succinate.
Fig. 3: Itaconate is a thiol-reactive anti-inflammatory metabolite.
Fig. 4: Fumarate is an oncometabolite and epigenetic modifier.
Fig. 5: 2-HG is an oncometabolite and epigenetic modifier.
Fig. 6: Acetyl-CoA as an epigenetic modifier in immunity and cancer.
Fig. 7: Therapeutic opportunities targeting metabolic signalling pathways.

Similar content being viewed by others

References

  1. Jha, A. K. et al. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity 42, 419–430 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. He, W. et al. Citric acid cycle intermediates as ligands for orphan G-protein-coupled receptors. Nature 429, 188–193 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Chouchani, E. T. et al. A unifying mechanism for mitochondrial superoxide production during ischemia–reperfusion injury. Cell. Metab. 23, 254–263 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Rubic, T. et al. Triggering the succinate receptor GPR91 on dendritic cells enhances immunity. Nat. Immunol. 9, 1261–1269 (2008).

    Article  CAS  PubMed  Google Scholar 

  6. Lei, W. et al. Activation of intestinal tuft cell-expressed Sucnr1 triggers type 2 immunity in the mouse small intestine. Proc. Natl Acad. Sci. USA. 115, 5552–5557 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Nadjsombati, M. S. et al. Detection of succinate by intestinal tuft cells triggers a type 2 innate immune circuit. Immunity 49, 33–41.e7 (2018).

  8. Schneider, C. et al. A metabolite-triggered tuft cell–ILC2 circuit drives small intestinal remodeling. Cell 174, 271–284.e14 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Littlewood-Evans, A. et al. GPR91 senses extracellular succinate released from inflammatory macrophages and exacerbates rheumatoid arthritis. J. Exp. Med. 213, 1655–1662 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kim, S. et al. Global metabolite profiling of synovial fluid for the specific diagnosis of rheumatoid arthritis from other inflammatory arthritis. PLoS One 9, e97501 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Hollander, A. P., Corke, K. P., Freemont, A. J. & Lewis, C. E. Expression of hypoxia-inducible factor 1α by macrophages in the rheumatoid synovium: implications for targeting of therapeutic genes to the inflamed joint. Arthritis Rheum. 44, 1540–1544 (2001).

    Article  CAS  PubMed  Google Scholar 

  12. Sadagopan, N. et al. Circulating succinate is elevated in rodent models of hypertension and metabolic disease. Am. J. Hypertens. 20, 1209–1215 (2007).

    CAS  PubMed  Google Scholar 

  13. van Diepen, J. A. et al. SUCNR1-mediated chemotaxis of macrophages aggravates obesity-induced inflammation and diabetes. Diabetologia 60, 1304–1313 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Mills, E. L. et al. Accumulation of succinate controls activation of adipose tissue thermogenesis. Nature 560, 102–106 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lewis, G. D. et al. Metabolic signatures of exercise in human plasma. Sci. Transl. Med. 2, 33ra37 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Peruzzotti-Jametti, L. et al. Macrophage-derived extracellular succinate licenses neural stem cells to suppress chronic neuroinflammation. Cell Stem Cell 22, 355–368.e13 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bhuniya, D. et al. Discovery of a potent and selective small molecule hGPR91 antagonist. Bioorg. Med. Chem. Lett. 21, 3596–3602 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Geubelle, P. et al. Identification and pharmacological characterization of succinate receptor agonists. Br. J. Pharmacol. 174, 796–808 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Tannahill, G. M. & O’Neill, L. A. The emerging role of metabolic regulation in the functioning of Toll-like receptors and the NOD-like receptor Nlrp3. FEBS Lett. 585, 1568–1572 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Semenza, G. L. & Wang, G. L. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol. Cell. Biol. 12, 5447–5454 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Semenza, G. L. Regulation of oxygen homeostasis by hypoxia-inducible factor 1. Physiology (Bethesda) 24, 97–106 (2009).

    CAS  Google Scholar 

  22. Epstein, A. C. R. et al. C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 107, 43–54 (2001).

    Article  CAS  PubMed  Google Scholar 

  23. Maxwell, P. H. et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399, 271–275 (1999).

    Article  CAS  PubMed  Google Scholar 

  24. Pearce, E. L. & Pearce, E. J. Metabolic pathways in immune cell activation and quiescence. Immunity 38, 633–643 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Frede, S., Stockmann, C., Freitag, P. & Fandrey, J. Bacterial lipopolysaccharide induces HIF-1 activation in human monocytes via p44/42 MAPK and NF-κB. Biochem. J. 396, 517–527 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wu, W. & Zhao, S. Metabolic changes in cancer: beyond the Warburg effect. Acta Biochim. Biophys. Sin. (Shanghai) 45, 18–26 (2013).

    Article  CAS  Google Scholar 

  27. Selak, M. A. et al. Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-α prolyl hydroxylase. Cancer Cell. 7, 77–85 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Rasola, A., Neckers, L. & Picard, D. Mitochondrial oxidative phosphorylation TRAP(1)ped in tumor cells. Trends Cell Biol. 24, 455–463 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Mu, X. et al. Oncometabolite succinate promotes angiogenesis by upregulating VEGF expression through GPR91-mediated STAT3 and ERK activation. Oncotarget 8, 13174–13185 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Guzy, R. D., Sharma, B., Bell, E., Chandel, N. S. & Schumacker, P. T. Loss of the SdhB, but not the SdhA, subunit of complex II triggers reactive oxygen species-dependent hypoxia-inducible factor activation and tumorigenesis. Mol. Cell. Biol. 28, 718–731 (2008).

    Article  CAS  PubMed  Google Scholar 

  31. Hagen, T. Oxygen versus reactive oxygen in the regulation of HIF-1α: the balance tips. Biochem. Res. Int. 2012, 436981 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Folbergrová, J., Ljunggren, B., Norberg, K. & Siesjö, B. K. Influence of complete ischemia on glycolytic metabolites, citric acid cycle intermediates, and associated amino acids in the rat cerebral cortex. Brain Res. 80, 265–279 (1974).

    Article  PubMed  Google Scholar 

  33. Chouchani, E. T. et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature 515, 431–435 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhang, W. et al. Evidence that hypoxia-inducible factor-1 (HIF-1) mediates transcriptional activation of interleukin-1β (IL-1β) in astrocyte cultures. J. Neuroimmunol. 174, 63–73 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Peyssonnaux, C. et al. Cutting edge: essential role of hypoxia inducible factor-1α in development of lipopolysaccharide-induced sepsis. J. Immunol. 178, 7516–7519 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Mills, E. L. et al. Succinate dehydrogenase supports metabolic repurposing of mitochondria to drive inflammatory macrophages. Cell 167, 457–470.e13 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. El-Khoury, R. et al. Engineering the alternative oxidase gene to better understand and counteract mitochondrial defects: state of the art and perspectives. Br. J. Pharmacol. 171, 2243–2249 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Jin, Z., Wei, W., Yang, M., Du, Y. & Wan, Y. Mitochondrial complex I activity suppresses inflammation and enhances bone resorption by shifting macrophage-osteoclast polarization. Cell. Metab. 20, 483–498 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Murphy, M. P. How mitochondria produce reactive oxygen species. Biochem. J. 417, 1–13 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Bénit, P. et al. Unsuspected task for an old team: succinate, fumarate and other Krebs cycle acids in metabolic remodeling. Biochim. Biophys. Acta 1837, 1330–1337 (2014).

    Article  PubMed  CAS  Google Scholar 

  41. Brigati, C. et al. Inflammation, HIF-1, and the epigenetics that follows. Mediators Inflamm. 2010, 263914 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Benn, C. S., Netea, M. G., Selin, L. K. & Aaby, P. A small jab—a big effect: nonspecific immunomodulation by vaccines. Trends Immunol. 34, 431–439 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Saeed, S. et al. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science 345, 1251086 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Cheng, S. C. et al. mTOR- and HIF-1α-mediated aerobic glycolysis as metabolic basis for trained immunity. Science 345, 1250684 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Park, J. et al. SIRT5-mediated lysine desuccinylation impacts diverse metabolic pathways. Mol. Cell 50, 919–930 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zhang, Z. et al. Identification of lysine succinylation as a new post-translational modification. Nat. Chem. Biol. 7, 58–63 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Xie, L. et al. First succinyl-proteome profiling of extensively drug-resistant Mycobacterium tuberculosis revealed involvement of succinylation in cellular physiology. J. Proteome Res. 14, 107–119 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Feng, S. et al. Succinyl-proteome profiling of Dendrobium officinale, an important traditional Chinese orchid herb, revealed involvement of succinylation in the glycolysis pathway. BMC Genomics 18, 598 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. He, D. et al. Global proteome analyses of lysine acetylation and succinylation reveal the widespread involvement of both modification in metabolism in the embryo of germinating rice seed. J. Proteome Res. 15, 879–890 (2016).

    Article  CAS  PubMed  Google Scholar 

  50. Xie, Z. et al. Lysine succinylation and lysine malonylation in histones. Mol. Cell. Proteomics 11, 100–107 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Du, J. et al. Sirt5 is a NAD-dependent protein lysine demalonylase and desuccinylase. Science 334, 806–809 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wang, Y. et al. KAT2A coupled with the α-KGDH complex acts as a histone H3 succinyltransferase. Nature 552, 273–277 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gibson, G. E. et al. α-ketoglutarate dehydrogenase complex–dependent succinylation of proteins in neurons and neuronal cell lines. J. Neurochem. 134, 86–96 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wang, F. et al. SIRT5 desuccinylates and activates pyruvate kinase M2 to block macrophage IL-1β production and to prevent dss-induced colitis in mice. Cell Rep. 19, 2331–2344 (2017).

    Article  CAS  PubMed  Google Scholar 

  55. Cordes, T. et al. Immunoresponsive gene 1 and itaconate inhibit succinate dehydrogenase to modulate intracellular succinate levels. J. Biol. Chem. 291, 14274–14284 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. 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  PubMed  PubMed Central  Google Scholar 

  57. Meiser, J. et al. Pro-inflammatory macrophages sustain pyruvate oxidation through pyruvate dehydrogenase for the synthesis of itaconate and to enable cytokine expression. J. Biol. Chem. 291, 3932–3946 (2016).

    Article  CAS  PubMed  Google Scholar 

  58. Németh, B. et al. Abolition of mitochondrial substrate-level phosphorylation by itaconic acid produced by LPS-induced Irg1 expression in cells of murine macrophage lineage. FASEB J. 30, 286–300 (2016).

    Article  PubMed  CAS  Google Scholar 

  59. Okabe, M., Lies, D., Kanamasa, S. & Park, E. Y. Biotechnological production of itaconic acid and its biosynthesis in Aspergillus terreus. Appl. Microbiol. Biotechnol. 84, 597–606 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Lee, C. G., Jenkins, N. A., Gilbert, D. J., Copeland, N. G. & O’Brien, W. E. Cloning and analysis of gene regulation of a novel LPS-inducible cDNA. Immunogenetics 41, 263–270 (1995).

    Article  CAS  PubMed  Google Scholar 

  61. Shin, J. H. et al. 1H NMR-based metabolomic profiling in mice infected with Mycobacterium tuberculosis. J. Proteome Res. 10, 2238–2247 (2011).

    Article  CAS  PubMed  Google Scholar 

  62. Sugimoto, M. S. H. Y. et al. Non-targeted metabolite profiling in activated macrophage secretion. Metabolomics 8, 624–633 (2011).

    Article  CAS  Google Scholar 

  63. 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  PubMed  PubMed Central  Google Scholar 

  64. McFadden, B. A. & Purohit, S. Itaconate, an isocitrate lyase-directed inhibitor in Pseudomonas indigofera. J. Bacteriol. 131, 136–144 (1977).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Naujoks, J. et al. IFNs modify the proteome of Legionella-containing vacuoles and restrict infection via IRG1-derived itaconic acid. PLoS Pathog. 12, e1005408 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Williams, J. O., Roche, T. E. & McFadden, B. A. Mechanism of action of isocitrate lyase from Pseudomonas indigofera. Biochemistry 10, 1384–1390 (1971).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. 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  PubMed  PubMed Central  Google Scholar 

  69. Nair, S. et al. Irg1 expression in myeloid cells prevents immunopathology during M. tuberculosis infection. J. Exp. Med. 215, 1035–1045 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ackermann, W. W. & Potter, V. R. Enzyme inhibition in relation to chemotherapy. Proc. Soc. Exp. Biol. Med. 72, 1–9 (1949).

    Article  CAS  PubMed  Google Scholar 

  71. El Azzouny, M. et al. Dimethyl itaconate is not metabolized into itaconate intracellularly. J. Biol. Chem. 292, 4766–4769 (2017).

    Article  CAS  Google Scholar 

  72. Kobayashi, E. H. et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nat. Commun. 7, 11624 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Brennan, M. S. et al. Dimethyl fumarate and monoethyl fumarate exhibit differential effects on KEAP1, NRF2 activation, and glutathione depletion in vitro. PLoS One 10, e0120254 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Saito, R. et al. Characterizations of three major cysteine sensors of Keap1 in stress response. Mol. Cell Biol. 36, 271–284 (2015).

    Article  PubMed  CAS  Google Scholar 

  75. Li, Y. et al. Immune responsive gene 1 (IRG1) promotes endotoxin tolerance by increasing A20 expression in macrophages through reactive oxygen species. J. Biol. Chem. 288, 16225–16234 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Van Quickelberghe, E. et al. Identification of immune-responsive gene 1 (IRG1) as a target of A20. J. Proteome Res. 17, 2182–2191 (2018).

    Article  PubMed  CAS  Google Scholar 

  77. Jamal Uddin, M. et al. IRG1 induced by heme oxygenase-1/carbon monoxide inhibits LPS-mediated sepsis and pro-inflammatory cytokine production. Cell Mol. Immunol. 13, 170–179 (2016).

    Article  CAS  PubMed  Google Scholar 

  78. Cheon, Y. P., Xu, X., Bagchi, M. K. & Bagchi, I. C. Immune-responsive gene 1 is a novel target of progesterone receptor and plays a critical role during implantation in the mouse. Endocrinology 144, 5623–5630 (2003).

    Article  CAS  PubMed  Google Scholar 

  79. Luan, H. H. & Medzhitov, R. Food fight: role of itaconate and other metabolites in antimicrobial defense. Cell. Metab. 24, 379–387 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Sherwin, J. R. et al. Identification of genes regulated by leukemia-inhibitory factor in the mouse uterus at the time of implantation. Mol. Endocrinol. 18, 2185–2195 (2004).

    Article  CAS  PubMed  Google Scholar 

  81. Weiss, J. M. et al. Itaconic acid mediates crosstalk between macrophage metabolism and peritoneal tumors. J. Clin. Invest. 128, 3794–3805 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  82. Hall, C. J. et al. Immunoresponsive gene 1 augments bactericidal activity of macrophage-lineage cells by regulating β-oxidation-dependent mitochondrial ROS production. Cell. Metab. 18, 265–278 (2013).

    Article  CAS  PubMed  Google Scholar 

  83. Shi, H. Z., Wang, D., Sun, X. N. & Sheng, L. MicroRNA-378 acts as a prognosis marker and inhibits cell migration, invasion and epithelial-mesenchymal transition in human glioma by targeting IRG1. Eur. Rev. Med. Pharmacol. Sci. 22, 3837–3846 (2018).

    PubMed  Google Scholar 

  84. Pan, J. et al. Immune responsive gene 1, a novel oncogene, increases the growth and tumorigenicity of glioma. Oncol. Rep. 32, 1957–1966 (2014).

    Article  CAS  PubMed  Google Scholar 

  85. Frezza, C. Mitochondrial metabolites: undercover signalling molecules. Interface Focus 7, 20160100 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  86. Arts, R. J. et al. Glutaminolysis and fumarate accumulation integrate immunometabolic and epigenetic programs in trained immunity. Cell. Metab. 24, 807–819 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. 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  PubMed  Google Scholar 

  88. Xiao, M. et al. Inhibition of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev. 26, 1326–1338 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Delage, B. et al. Arginine deprivation and argininosuccinate synthetase expression in the treatment of cancer. Int. J. Cancer 126, 2762–2772 (2010).

    CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Angiari, S. & O’Neill, L. A. Dimethyl fumarate: targeting glycolysis to treat MS. Cell Res. 28, 613–615 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Brück, J., Dringen, R., Amasuno, A., Pau-Charles, I. & Ghoreschi, K. A review of the mechanisms of action of dimethylfumarate in the treatment of psoriasis. Exp. Dermatol. 27, 611–624 (2018).

    Article  PubMed  CAS  Google Scholar 

  94. Mills, E. A., Ogrodnik, M. A., Plave, A. & Mao-Draayer, Y. Emerging understanding of the mechanism of action for dimethyl fumarate in the treatment of multiple sclerosis. Front. Neurol. 9, 5 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  96. Sciacovelli, M. & Frezza, C. Oncometabolites: unconventional triggers of oncogenic signalling cascades. Free Radic. Biol. Med. 100, 175–181 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Rustin, P. et al. Inborn errors of the Krebs cycle: a group of unusual mitochondrial diseases in human. Biochim. Biophys. Acta 1361, 185–197 (1997).

    Article  CAS  PubMed  Google Scholar 

  98. Allegri, G. et al. Fumaric aciduria: an overview and the first Brazilian case report. J. Inherit. Metab. Dis. 33, 411–419 (2010).

    Article  PubMed  Google Scholar 

  99. Blatnik, M., Thorpe, S. R. & Baynes, J. W. Succination of proteins by fumarate: mechanism of inactivation of glyceraldehyde-3-phosphate dehydrogenase in diabetes. Ann. NY Acad. Sci. 1126, 272–275 (2008).

    Article  CAS  PubMed  Google Scholar 

  100. Adam, J. et al. Renal cyst formation in Fh1-deficient mice is independent of the Hif/Phd pathway: roles for fumarate in KEAP1 succination and Nrf2 signaling. Cancer Cell 20, 524–537 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Ooi, A. et al. An antioxidant response phenotype shared between hereditary and sporadic type 2 papillary renal cell carcinoma. Cancer Cell 20, 511–523 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Linehan, W. M. et al. Comprehensive molecular characterization of papillary renal-cell carcinoma. N. Engl. J. Med. 374, 135–145 (2016).

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Kerins, M. J. et al. Fumarate mediates a chronic proliferative signal in fumarate hydratase-inactivated cancer cells by increasing transcription and translation of ferritin genes. Mol. Cell Biol. 37, e00079-17 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  106. Tyrakis, P. A. et al. Fumarate hydratase loss causes combined respiratory chain defects. Cell Rep. 21, 1036–1047 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. 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  PubMed  Google Scholar 

  108. Laukka, T. et al. Fumarate and succinate regulate expression of hypoxia-inducible genes via TET enzymes. J. Biol. Chem. 291, 4256–4265 (2016).

    Article  CAS  PubMed  Google Scholar 

  109. Salminen, A., Kauppinen, A. & Kaarniranta, K. 2-Oxoglutarate-dependent dioxygenases are sensors of energy metabolism, oxygen availability, and iron homeostasis: potential role in the regulation of aging process. Cell. Mol. Life Sci. 72, 3897–3914 (2015).

    Article  CAS  PubMed  Google Scholar 

  110. 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  PubMed  Google Scholar 

  111. Yang, M., Soga, T., Pollard, P. J. & Adam, J. The emerging role of fumarate as an oncometabolite. Front. Oncol. 2, 85 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Brabletz, T., Kalluri, R., Nieto, M. A. & Weinberg, R. A. EMT in cancer. Nat. Rev. Cancer 18, 128–134 (2018).

    Article  CAS  PubMed  Google Scholar 

  114. Brat, D. J. et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N. Engl. J. Med. 372, 2481–2498 (2015).

    Article  CAS  PubMed  Google Scholar 

  115. Yogev, O. et al. Fumarase: a mitochondrial metabolic enzyme and a cytosolic/nuclear component of the DNA damage response. PLoS Biol. 8, e1000328 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Jiang, Y. et al. Local generation of fumarate promotes DNA repair through inhibition of histone H3 demethylation. Nat. Cell Biol. 17, 1158–1168 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Sulkowski, P. L. et al. Krebs-cycle-deficient hereditary cancer syndromes are defined by defects in homologous-recombination DNA repair. Nat. Genet. 50, 1086–1092 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Johnson, T. I., Costa, A. S. H., Ferguson, A. N. & Frezza, C. Fumarate hydratase loss promotes mitotic entry in the presence of DNA damage after ionising radiation. Cell Death Dis. 9, 913 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Ye, D., Guan, K. L. & Xiong, Y. Metabolism, activity, and targeting of d- and l-2-Hydroxyglutarates. Trends Cancer 4, 151–165 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Chalmers, R. A. et al. d-2-hydroxyglutaric aciduria: case report and biochemical studies. J. Inherit. Metab. Dis. 3, 11–15 (1980).

    Article  CAS  PubMed  Google Scholar 

  121. Duran, M., Kamerling, J. P., Bakker, H. D., van Gennip, A. H. & Wadman, S. K. l-2-Hydroxyglutaric aciduria: an inborn error of metabolism? J. Inherit. Metab. Dis. 3, 109–112 (1980).

    Article  CAS  PubMed  Google Scholar 

  122. Kranendijk, M., Struys, E. A., Salomons, G. S., Van der Knaap, M. S. & Jakobs, C. Progress in understanding 2-hydroxyglutaric acidurias. J. Inherit. Metab. Dis. 35, 571–587 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Rzem, R. et al. A gene encoding a putative FAD-dependent l-2-hydroxyglutarate dehydrogenase is mutated in l-2-hydroxyglutaric aciduria. Proc. Natl. Acad. Sci. USA 101, 16849–16854 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Struys, E. A. et al. Mutations in the d-2-hydroxyglutarate dehydrogenase gene cause D-2-hydroxyglutaric aciduria. Am. J. Hum. Genet. 76, 358–360 (2005).

    Article  CAS  PubMed  Google Scholar 

  125. Topçu, M. et al. l-2-Hydroxyglutaric aciduria: identification of a mutant gene C14orf160, localized on chromosome 14q22.1. Hum. Mol. Genet. 13, 2803–2811 (2004).

    Article  PubMed  Google Scholar 

  126. Kranendijk, M. et al. IDH2 mutations in patients with d-2-hydroxyglutaric aciduria. Science 330, 336 (2010).

    Article  CAS  PubMed  Google Scholar 

  127. Nota, B. et al. Deficiency in SLC25A1, encoding the mitochondrial citrate carrier, causes combined d-2- and l-2-hydroxyglutaric aciduria. Am. J. Hum. Genet. 92, 627–631 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Dang, L. & Su, S. M. Isocitrate dehydrogenase mutation and R-2-hydroxyglutarate: from basic discovery to therapeutics development. Annu. Rev. Biochem. 86, 305–331 (2017).

    Article  CAS  PubMed  Google Scholar 

  129. Mardis, E. R. et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N. Engl. J. Med. 361, 1058–1066 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Parsons, D. W. et al. An integrated genomic analysis of human glioblastoma multiforme. Science 321, 1807–1812 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. MacIver, N. J., Michalek, R. D. & Rathmell, J. C. Metabolic regulation of T lymphocytes. Annu. Rev. Immunol. 31, 259–283 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Dang, E. V. et al. Control of TH17/Treg balance by hypoxia-inducible factor 1. Cell 146, 772–784 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Xu, T. et al. Metabolic control of TH17 and induced Treg cell balance by an epigenetic mechanism. Nature 548, 228–233 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Figueroa, M. E. et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 18, 553–567 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Losman, J. A. et al. R-2-hydroxyglutarate is sufficient to promote leukemogenesis and its effects are reversible. Science 339, 1621–1625 (2013).

    Article  CAS  PubMed  Google Scholar 

  136. Tyrakis, P. A. et al. S-2-hydroxyglutarate regulates CD8+ T-lymphocyte fate. Nature 540, 236–241 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Doedens, A. L. et al. Hypoxia-inducible factors enhance the effector responses of CD8+ T cells to persistent antigen. Nat. Immunol. 14, 1173–1182 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Finlay, D. K. et al. PDK1 regulation of mTOR and hypoxia-inducible factor 1 integrate metabolism and migration of CD8+ T cells. J. Exp. Med. 209, 2441–2453 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Suzuki, H. et al. Mutational landscape and clonal architecture in grade II and III gliomas. Nat. Genet. 47, 458–468 (2015).

    Article  CAS  PubMed  Google Scholar 

  140. Watanabe, T., Nobusawa, S., Kleihues, P. & Ohgaki, H. IDH1 mutations are early events in the development of astrocytomas and oligodendrogliomas. Am. J. Pathol. 174, 1149–1153 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Dang, L. et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 462, 739–744 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Ward, P. S. et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting α-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 17, 225–234 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Yan, H. et al. IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 360, 765–773 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Chowdhury, R. et al. The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep. 12, 463 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Xu, W. et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell. 19, 17–30 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Noushmehr, H. et al. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell. 17, 510–522 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Turcan, S. et al. IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 483, 479–483 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Wang, P. et al. Mutations in isocitrate dehydrogenase 1 and 2 occur frequently in intrahepatic cholangiocarcinomas and share hypermethylation targets with glioblastomas. Oncogene 32, 3091–3100 (2013).

    Article  CAS  PubMed  Google Scholar 

  149. Flavahan, W. A. et al. Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature 529, 110–114 (2016).

    Article  CAS  PubMed  Google Scholar 

  150. Zhao, S. et al. Glioma-derived mutations in IDH1 dominantly inhibit IDH1 catalytic activity and induce HIF-1α. Science 324, 261–265 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Intlekofer, A. M. et al. Hypoxia induces production of l-2-hydroxyglutarate. Cell Metab. 22, 304–311 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Intlekofer, A. M. et al. l-2-Hydroxyglutarate production arises from noncanonical enzyme function at acidic pH. Nat. Chem. Biol. 13, 494–500 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Nadtochiy, S. M. et al. Acidic pH Is a metabolic switch for 2-hydroxyglutarate generation and signaling. J. Biol. Chem. 291, 20188–20197 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Sasaki, M. et al. d-2-hydroxyglutarate produced by mutant IDH1 perturbs collagen maturation and basement membrane function. Genes Dev. 26, 2038–2049 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Tarhonskaya, H. et al. Non-enzymatic chemistry enables 2-hydroxyglutarate-mediated activation of 2-oxoglutarate oxygenases. Nat. Commun. 5, 3423 (2014).

    Article  PubMed  CAS  Google Scholar 

  156. Wang, P. et al. Oncometabolite d-2-hydroxyglutarate inhibits ALKBH DNA repair enzymes and sensitizes IDH mutant cells to alkylating agents. Cell Rep. 13, 2353–2361 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Inoue, S. et al. Mutant IDH1 downregulates ATM and alters DNA repair and sensitivity to DNA damage independent of TET2. Cancer Cell 30, 337–348 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Carbonneau, M. et al. The oncometabolite 2-hydroxyglutarate activates the mTOR signalling pathway. Nat. Commun. 7, 12700 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Reitman, Z. J. et al. Profiling the effects of isocitrate dehydrogenase 1 and 2 mutations on the cellular metabolome. Proc. Natl. Acad. Sci. USA 108, 3270–3275 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Chesnelong, C. et al. Lactate dehydrogenase A silencing in IDH mutant gliomas. Neuro-Oncol. 16, 686–695 (2014).

    Article  CAS  PubMed  Google Scholar 

  161. Grassian, A. R. et al. IDH1 mutations alter citric acid cycle metabolism and increase dependence on oxidative mitochondrial metabolism. Cancer Res. 74, 3317–3331 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Izquierdo-Garcia, J. L. et al. IDH1 mutation induces reprogramming of pyruvate metabolism. Cancer Res. 75, 2999–3009 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Chen, J. Y. et al. The oncometabolite R-2-hydroxyglutarate activates NF-κB-dependent tumor-promoting stromal niche for acute myeloid leukemia cells. Sci. Rep. 6, 1 (2016).

    Article  CAS  Google Scholar 

  164. Li, F. et al. NADP+–IDH mutations promote hypersuccinylation that impairs mitochondria respiration and induces apoptosis resistance. Mol. Cell 60, 661–675 (2015).

    Article  CAS  PubMed  Google Scholar 

  165. Jiang, B. et al. IDH1 mutation promotes tumorigenesis by inhibiting JNK activation and apoptosis induced by serum starvation. Cell Rep. 19, 389–400 (2017).

    Article  CAS  PubMed  Google Scholar 

  166. Yang, Z. et al. 2-HG inhibits necroptosis by stimulating DNMT1-dependent hypermethylation of the RIP3 promoter. Cell Rep. 19, 1846–1857 (2017).

    Article  CAS  PubMed  Google Scholar 

  167. Kurdistani, S. K. & Grunstein, M. Histone acetylation and deacetylation in yeast. Nat. Rev. Mol. Cell Biol. 4, 276–284 (2003).

    Article  CAS  PubMed  Google Scholar 

  168. Wellen, K. E. et al. ATP-citrate lyase links cellular metabolism to histone acetylation. Science 324, 1076–1080 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Peng, M. et al. Aerobic glycolysis promotes T helper 1 cell differentiation through an epigenetic mechanism. Science 354, 481–484 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Chang, C. H. et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 153, 1239–1251 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Osinalde, N. et al. Nuclear phosphoproteomic screen uncovers ACLY as mediator of IL-2-induced proliferation of CD4+ T lymphocytes. Mol. Cell. Proteomics 15, 2076–2092 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Lee, J. V. et al. Akt-dependent metabolic reprogramming regulates tumor cell histone acetylation. Cell. Metab. 20, 306–319 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Zhang, L., Liu, Z., Ma, W. & Wang, B. The landscape of histone acetylation involved in epithelial-mesenchymal transition in lung cancer. J. Can. Res. Ther. 9, S86–S91 (2013).

    Article  Google Scholar 

  174. Mi, W. et al. YEATS2 links histone acetylation to tumorigenesis of non-small cell lung cancer. Nat. Commun. 8, 1088 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  175. Sivanand, S. et al. Nuclear acetyl-CoA production by ACLY promotes homologous recombination. Mol. Cell 67, 252–265.e56 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Valls-Lacalle, L. et al. Succinate dehydrogenase inhibition with malonate during reperfusion reduces infarct size by preventing mitochondrial permeability transition. Cardiovasc. Res. 109, 374–384 (2016).

    Article  CAS  PubMed  Google Scholar 

  177. Valls-Lacalle, L. et al. Selective inhibition of succinate dehydrogenase in reperfused myocardium with intracoronary malonate reduces infarct size. Sci. Rep. 8, 2442 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  178. Kohlhauer, M. et al. Metabolomic profiling in acute ST-segment-elevation myocardial infarction identifies succinate as an early marker of human ischemia-reperfusion injury. J. Am. Heart Assoc. 7, e007546 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  179. Zhang, J. et al. accumulation of succinate in cardiac ischemia primarily occurs via canonical Krebs cycle activity. Cell Rep. 23, 2617–2628 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Ariza, A. C., Deen, P. M. & Robben, J. H. The succinate receptor as a novel therapeutic target for oxidative and metabolic stress-related conditions. Front. Endocrinol. (Lausanne) 3, 22 (2012).

    Article  Google Scholar 

  181. Hamel, D. et al. G-protein-coupled receptor 91 and succinate are key contributors in neonatal postcerebral hypoxia-ischemia recovery. Arterioscler. Thromb. Vasc. Biol. 34, 285–293 (2014).

    Article  CAS  PubMed  Google Scholar 

  182. Jean, S. R., Ahmed, M., Lei, E. K., Wisnovsky, S. P. & Kelley, S. O. Peptide-mediated delivery of chemical probes and therapeutics to mitochondria. Acc. Chem. Res. 49, 1893–1902 (2016).

    Article  CAS  PubMed  Google Scholar 

  183. An, J., Rao, A. & Ko, M. TET family dioxygenases and DNA demethylation in stem cells and cancers. Exp. Mol. Med. 49, e323 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Vissers, M. C., Kuiper, C. & Dachs, G. U. Regulation of the 2-oxoglutarate-dependent dioxygenases and implications for cancer. Biochem. Soc. Trans. 42, 945–951 (2014).

    Article  CAS  PubMed  Google Scholar 

  185. Zhang, J. et al. Effect of TET inhibitor on bovine parthenogenetic embryo development. PLoS One 12, e0189542 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  186. Hatch, S. B. et al. Assessing histone demethylase inhibitors in cells: lessons learned. Epigenetics Chromatin 10, 9 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  187. Maes, T., Carceller, E., Salas, J., Ortega, A. & Buesa, C. Advances in the development of histone lysine demethylase inhibitors. Curr. Opin. Pharmacol. 23, 52–60 (2015).

    Article  CAS  PubMed  Google Scholar 

  188. Pergola, P. E., Spinowitz, B. S., Hartman, C. S., Maroni, B. J. & Haase, V. H. Vadadustat, a novel oral HIF stabilizer, provides effective anemia treatment in nondialysis-dependent chronic kidney disease. Kidney Int. 90, 1115–1122 (2016).

    Article  CAS  PubMed  Google Scholar 

  189. Yeh, T. L. et al. Molecular and cellular mechanisms of HIF prolyl hydroxylase inhibitors in clinical trials. Chem. Sci. 8, 7651–7668 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Yen, K. et al. AG-221, a first-in-class therapy targeting acute myeloid leukemia harboring oncogenic IDH2 mutations. Cancer Discov. 7, 478–493 (2017).

    Article  CAS  PubMed  Google Scholar 

  191. Smith, R. A., Hartley, R. C., Cochemé, H. M. & Murphy, M. P. Mitochondrial pharmacology. Trends Pharmacol. Sci. 33, 341–352 (2012).

    Article  CAS  PubMed  Google Scholar 

  192. Smith, R. A., Hartley, R. C. & Murphy, M. P. Mitochondria-targeted small molecule therapeutics and probes. Antioxid. Redox Signal. 15, 3021–3038 (2011).

    Article  CAS  PubMed  Google Scholar 

  193. Yousif, L. F., Stewart, K. M. & Kelley, S. O. Targeting mitochondria with organelle-specific compounds: strategies and applications. Chembiochem 10, 1939–1950 (2009).

    Article  CAS  PubMed  Google Scholar 

  194. Fan, T. W. et al. Altered regulation of metabolic pathways in human lung cancer discerned by 13C stable isotope-resolved metabolomics (SIRM). Mol. Cancer 8, 41 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  195. Sellers, K. et al. Pyruvate carboxylase is critical for non-small-cell lung cancer proliferation. J. Clin. Invest. 125, 687–698 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  196. Maher, E. A. et al. Metabolism of [U-13C]glucose in human brain tumors in vivo. NMR Biomed. 25, 1234–1244 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Hensley, C. T. et al. Metabolic heterogeneity in human lung tumors. Cell 164, 681–694 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Joshi, S. et al. The genomic landscape of renal oncocytoma identifies a metabolic barrier to tumorigenesis. Cell Rep. 13, 1895–1908 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by a grant to M.P.M. from the Medical Research Council UK (MC_U105663142) and a Wellcome Trust Investigator award to MPM (110159/Z/15/Z). The O’Neill laboratory acknowledges the following grant support: European Research Council (ECFP7-ERC-MICROINNATE), Science Foundation Ireland Investigator Award (SFI 12/IA/1531), GlaxoSmithKline Visiting Scientist Programme and The Wellcome Trust (oneill-wellcometrust-metabolic, grant number 205455).

Author information

Authors and Affiliations

Authors

Contributions

All authors wrote, edited and approved final manuscript.

Corresponding author

Correspondence to Luke A. O’Neill.

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.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ryan, D.G., Murphy, M.P., Frezza, C. et al. Coupling Krebs cycle metabolites to signalling in immunity and cancer. Nat Metab 1, 16–33 (2019). https://doi.org/10.1038/s42255-018-0014-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-018-0014-7

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