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:

H3K9me3 represses G6PD expression to suppress the pentose phosphate pathway and ROS production to promote human mesothelioma growth

Abstract

The role of glucose-6-phosphate dehydrogenase (G6PD) in human cancer is incompletely understood. In a metabolite screening, we observed that inhibition of H3K9 methylation suppressed aerobic glycolysis and enhances the PPP in human mesothelioma cells. Genome-wide screening identified G6PD as an H3K9me3 target gene whose expression is correlated with increased tumor cell apoptosis. Inhibition of aerobic glycolysis enzyme LDHA and G6PD had no significant effects on tumor cell survival. Ablation of G6PD had no significant effect on human mesothelioma and colon carcinoma xenograft growth in athymic mice. However, activation of G6PD with the G6PD-selective activator AG1 induced tumor cell death. AG1 increased tumor cell ROS production and the resultant extrinsic and intrinsic death pathways, mitochondrial processes, and unfolded protein response in tumor cells. Consistent with increased tumor cell death in vitro, AG1 suppressed human mesothelioma xenograft growth in a dose-dependent manner in vivo. Furthermore, AG1 treatment significantly increased tumor-bearing mouse survival in an intra-peritoneum xenograft athymic mouse model. Therefore, in human mesothelioma and colon carcinoma, G6PD is not essential for tumor growth. G6PD acts as a metabolic checkpoint to control metabolic flux towards the PPP to promote tumor cell apoptosis, and its expression is repressed by its promotor H3K9me3 deposition.

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: Epigenetic regulation of metabolism in human mesothelioma cells.
Fig. 2: G6PD is repressed by its promoter H3K9me3 deposition in human mesothelioma cells.
Fig. 3: Inhibition of histone methylation induces human mesothelioma cell apoptosis.
Fig. 4: Aerobic glycolysis and G6PD do not maintain human mesothelioma cell survival in vitro.
Fig. 5: G6PD increases ROS promotion to regulate ROS-regulated cell death pathway to promote human mesothelioma cell death in vitro.
Fig. 6: G6PD is not essential for human mesothelioma survival in vitro and in vivo.
Fig. 7: Pharmacological activation of G6PD enzymatic activity suppresses human mesothelioma growth in vivo.

Similar content being viewed by others

References

  1. Warburg O. On the origin of cancer cells. Science. 1956;123:309–14.

    Article  CAS  PubMed  Google Scholar 

  2. Pathria G, Scott DA, Feng Y, Sang Lee J, Fujita Y, Zhang G, et al. Targeting the Warburg effect via LDHA inhibition engages ATF4 signaling for cancer cell survival. EMBO J. 2018;37:e99735.

  3. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029–33.

    Article  CAS  Google Scholar 

  4. Ge T, Yang J, Zhou S, Wang Y, Li Y, Tong X. The role of the pentose phosphate pathway in diabetes and cancer. Front Endocrinol. 2020;11:365.

    Article  Google Scholar 

  5. Wu S, Wang H, Li Y, Xie Y, Huang C, Zhao H, et al. Transcription factor YY1 promotes cell proliferation by directly activating the pentose phosphate pathway. Cancer Res. 2018;78:4549–62.

    Article  CAS  PubMed  Google Scholar 

  6. Zhang X, Zhang X, Li Y, Shao Y, Xiao J, Zhu G, et al. PAK4 regulates G6PD activity by p53 degradation involving colon cancer cell growth. Cell Death Dis. 2017;8:e2820.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Nakamura M, Nagase K, Yoshimitsu M, Magara T, Nojiri Y, Kato H, et al. Glucose-6-phosphate dehydrogenase correlates with tumor immune activity and programmed death ligand-1 expression in Merkel cell carcinoma. J Immunother Cancer. 2020;8:e001679.

  8. Ding H, Chen Z, Wu K, Huang SM, Wu WL, LeBoeuf SE, et al. Activation of the NRF2 antioxidant program sensitizes tumors to G6PD inhibition. Sci Adv. 2021;7:eabk1023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kowalik MA, Columbano A, Perra A. Emerging role of the pentose phosphate pathway in hepatocellular carcinoma. Front Oncol. 2017;7:87.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Nagashio R, Oikawa S, Yanagita K, Hagiuda D, Kuchitsu Y, Igawa S, et al. Prognostic significance of G6PD expression and localization in lung adenocarcinoma. Biochim Biophys Acta Proteins Proteom. 2019;1867:38–46.

    Article  CAS  PubMed  Google Scholar 

  11. Ghergurovich JM, Esposito M, Chen Z, Wang JZ, Bhatt V, Lan T, et al. Glucose-6-phosphate dehydrogenase is not essential for K-RAS-driven tumor growth or metastasis. Cancer Res. 2020;80:3820–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Chen L, Zhang Z, Hoshino A, Zheng HD, Morley M, Arany Z, et al. NADPH production by the oxidative pentose-phosphate pathway supports folate metabolism. Nat Metab. 2019;1:404–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Liu B, Bai W, Ou G, Zhang J. Cdh1-mediated metabolic switch from pentose phosphate pathway to glycolysis contributes to sevoflurane-induced neuronal apoptosis in developing brain. ACS Chem Neurosci. 2019;10:2332–44.

    Article  CAS  PubMed  Google Scholar 

  14. Figueroa M, Graf TN, Ayers S, Adcock AF, Kroll DJ, Yang J, et al. Cytotoxic epipolythiodioxopiperazine alkaloids from filamentous fungi of the Bionectriaceae. J Antibiot. 2012;65:559–64.

    Article  CAS  Google Scholar 

  15. Amrine CSM, Raja HA, Darveaux BA, Pearce CJ, Oberlies NH. Media studies to enhance the production of verticillins facilitated by in situ chemical analysis. J Ind Microbiol Biotechnol. 2018;45:1053–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Raub AG, Hwang S, Horikoshi N, Cunningham AD, Rahighi S, Wakatsuki S, et al. Small-molecule activators of glucose-6-phosphate dehydrogenase (G6PD) bridging the dimer interface. ChemMedChem. 2019;14:1321–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hwang S, Mruk K, Rahighi S, Raub AG, Chen CH, Dorn LE, et al. Correcting glucose-6-phosphate dehydrogenase deficiency with a small-molecule activator. Nat Commun. 2018;9:4045.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Ghergurovich JM, Garcia-Canaveras JC, Wang J, Schmidt E, Zhang Z, TeSlaa T, et al. A small molecule G6PD inhibitor reveals immune dependence on pentose phosphate pathway. Nat Chem Biol. 2020;16:731–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lu C, Klement JD, Smith AD, Yang D, Waller JL, Browning DD, et al. p50 suppresses cytotoxic T lymphocyte effector function to regulate tumor immune escape and response to immunotherapy. J Immunother Cancer. 2020;8:e001365.

  20. Fiehn O, Wohlgemuth G, Scholz M, Kind T, Lee DY, Lu Y, et al. Quality control for plant metabolomics: reporting MSI-compliant studies. Plant J. 2008;53:691–704.

    Article  CAS  PubMed  Google Scholar 

  21. Ding J, Li T, Wang X, Zhao E, Choi JH, Yang L, et al. The histone H3 methyltransferase G9A epigenetically activates the serine-glycine synthesis pathway to sustain cancer cell survival and proliferation. Cell Metab. 2013;18:896–907.

    Article  CAS  PubMed  Google Scholar 

  22. Lu C, Liu Z, Klement JD, Yang D, Merting AD, Poschel D, et al. WDR5-H3K4me3 epigenetic axis regulates OPN expression to compensate PD-L1 function to promote pancreatic cancer immune escape. J Immunother Cancer. 2021;9:e002624.

  23. Paschall AV, Yang D, Lu C, Choi JH, Li X, Liu F, et al. H3K9 trimethylation silences fas expression to confer colon carcinoma immune escape and 5-fluorouracil chemoresistance. J Immunol. 2015;195:1868–82.

    Article  CAS  PubMed  Google Scholar 

  24. Lu C, Paschall AV, Shi H, Savage N, Waller JL, Sabbatini ME, et al. The MLL1-H3K4me3 axis-mediated PD-L1 expression and pancreatic cancer immune evasion. J Natl Cancer Inst. 2017;109:djw283.

    Article  PubMed Central  CAS  Google Scholar 

  25. Ravindran Menon D, Hammerlindl H, Torrano J, Schaider H, Fujita M. Epigenetics and metabolism at the crossroads of stress-induced plasticity, stemness and therapeutic resistance in cancer. Theranostics. 2020;10:6261–77.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. DeBerardinis RJ, Mancuso A, Daikhin E, Nissim I, Yudkoff M, Wehrli S, et al. Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc Natl Acad Sci USA. 2007;104:19345–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lachner M, O’Carroll D, Rea S, Mechtler K, Jenuwein T. Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature. 2001;410:116–20.

    Article  CAS  PubMed  Google Scholar 

  28. Lu C, Yang D, Klement JD, Colson YL, Oberlies NH, Pearce CJ, et al. G6PD functions as a metabolic checkpoint to regulate granzyme B expression in tumor-specific cytotoxic T lymphocytes. J Immunother Cancer. 2022;10:e003543.

  29. Fritsch L, Robin P, Mathieu JR, Souidi M, Hinaux H, Rougeulle C, et al. A subset of the histone H3 lysine 9 methyltransferases Suv39h1, G9a, GLP, and SETDB1 participate in a multimeric complex. Mol Cell. 2010;37:46–56.

    Article  CAS  PubMed  Google Scholar 

  30. Rice JC, Briggs SD, Ueberheide B, Barber CM, Shabanowitz J, Hunt DF, et al. Histone methyltransferases direct different degrees of methylation to define distinct chromatin domains. Mol Cell. 2003;12:1591–8.

    Article  CAS  PubMed  Google Scholar 

  31. Muller MM, Fierz B, Bittova L, Liszczak G, Muir TW. A two-state activation mechanism controls the histone methyltransferase Suv39h1. Nat Chem Biol. 2016;12:188–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Abbosh PH, Montgomery JS, Starkey JA, Novotny M, Zuhowski EG, Egorin MJ, et al. Dominant-negative histone H3 lysine 27 mutant derepresses silenced tumor suppressor genes and reverses the drug-resistant phenotype in cancer cells. Cancer Res. 2006;66:5582–91.

    Article  CAS  PubMed  Google Scholar 

  33. Matei D, Nephew KP. Epigenetic attire in ovarian cancer: the emperor’s new clothes. Cancer Res. 2020;80:3775–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Nicetto D, Donahue G, Jain T, Peng T, Sidoli S, Sheng L, et al. H3K9me3-heterochromatin loss at protein-coding genes enables developmental lineage specification. Science. 2019;363:294–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Olcina MM, Leszczynska KB, Senra JM, Isa NF, Harada H, Hammond EM. H3K9me3 facilitates hypoxia-induced p53-dependent apoptosis through repression of APAK. Oncogene. 2016;35:793–9.

    Article  CAS  PubMed  Google Scholar 

  36. Salvi A, Amrine CSM, Austin JR, Kilpatrick K, Russo A, Lantvit D, et al. Verticillin A causes apoptosis and reduces tumor burden in high-grade serous ovarian cancer by inducing DNA damage. Mol Cancer Ther. 2020;19:89–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Aggarwal BB, Gupta SC, Kim JH. Historical perspectives on tumor necrosis factor and its superfamily: 25 years later, a golden journey. Blood. 2012;119:651–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gu Y, Bouwman P, Greco D, Saarela J, Yadav B, Jonkers J, et al. Suppression of BRCA1 sensitizes cells to proteasome inhibitors. Cell Death Dis. 2014;5:e1580.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Iurlaro R, Puschel F, Leon-Annicchiarico CL, O’Connor H, Martin SJ, Palou-Gramon D, et al. Glucose deprivation induces ATF4-mediated apoptosis through TRAIL death receptors. Mol Cell Biol. 2017;37:e00479-16.

  40. Redza-Dutordoir M, Averill-Bates DA. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim Biophys Acta. 2016;1863:2977–92.

    Article  CAS  PubMed  Google Scholar 

  41. Balaban RS, Nemoto S, Finkel T. Mitochondria, oxidants, and aging. Cell. 2005;120:483–95.

    Article  CAS  PubMed  Google Scholar 

  42. Koo SJ, Szczesny B, Wan X, Putluri N, Garg NJ. Pentose phosphate shunt modulates reactive oxygen species and nitric oxide production controlling Trypanosoma cruzi in macrophages. Front Immunol. 2018;9:202.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Tu D, Gao Y, Yang R, Guan T, Hong JS, Gao HM. The pentose phosphate pathway regulates chronic neuroinflammation and dopaminergic neurodegeneration. J Neuroinflammation. 2019;16:255.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hayes JD, Dinkova-Kostova AT, Tew KD. Oxidative stress in cancer. Cancer Cell. 2020;38:167–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Oshima N, Ishida R, Kishimoto S, Beebe K, Brender JR, Yamamoto K, et al. Dynamic imaging of LDH inhibition in tumors reveals rapid in vivo metabolic rewiring and vulnerability to combination therapy. Cell Rep. 2020;30:1798–810 e1794.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A, Deck LM, et al. Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc Natl Acad Sci USA. 2010;107:2037–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Yeung C, Gibson AE, Issaq SH, Oshima N, Baumgart JT, Edessa LD, et al. Targeting glycolysis through inhibition of lactate dehydrogenase impairs tumor growth in preclinical models of ewing sarcoma. Cancer Res. 2019;79:5060–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sena LA, Chandel NS. Physiological roles of mitochondrial reactive oxygen species. Mol Cell. 2012;48:158–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Jian SL, Chen WW, Su YC, Su YW, Chuang TH, Hsu SC, et al. Glycolysis regulates the expansion of myeloid-derived suppressor cells in tumor-bearing hosts through prevention of ROS-mediated apoptosis. Cell Death Dis. 2017;8:e2779.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kuehne A, Emmert H, Soehle J, Winnefeld M, Fischer F, Wenck H, et al. Acute activation of oxidative pentose phosphate pathway as first-line response to oxidative stress in human skin cells. Mol Cell. 2015;59:359–71.

    Article  CAS  PubMed  Google Scholar 

  51. Dick TP, Ralser M. Metabolic remodeling in times of stress: who shoots faster than his shadow? Mol Cell. 2015;59:519–21.

    Article  CAS  PubMed  Google Scholar 

  52. Li W, Kou J, Qin J, Li L, Zhang Z, Pan Y, et al. NADPH levels affect cellular epigenetic state by inhibiting HDAC3-Ncor complex. Nat Metab. 2021;3:75–89.

    Article  CAS  PubMed  Google Scholar 

  53. Monteiro-Reis S, Lameirinhas A, Miranda-Goncalves V, Felizardo D, Dias PC, Oliveira J, et al. Sirtuins’ deregulation in bladder cancer: SIRT7 is implicated in tumor progression through epithelial to mesenchymal transition promotion. Cancers. 2020;12:1066.

  54. Stomper J, Meier R, Ma T, Pfeifer D, Ihorst G, Blagitko-Dorfs N, et al. Integrative study of EZH2 mutational status, copy number, protein expression and H3K27 trimethylation in AML/MDS patients. Clin Epigenet. 2021;13:77.

    Article  CAS  Google Scholar 

  55. Mozzetta C, Pontis J, Fritsch L, Robin P, Portoso M, Proux C, et al. The histone H3 lysine 9 methyltransferases G9a and GLP regulate polycomb repressive complex 2-mediated gene silencing. Mol Cell. 2014;53:277–89.

    Article  CAS  PubMed  Google Scholar 

  56. Paschall AV, Liu K. Epigenetic regulation of apoptosis and cell cycle regulatory genes in human colon carcinoma cells. Genom Data. 2015;5:189–91.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Lu C, Klement JD, Yang D, Albers T, Lebedyeva IO, Waller JL, et al. SUV39H1 regulates human colon carcinoma apoptosis and cell cycle to promote tumor growth. Cancer Lett. 2020;476:87–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Williams D, Fingleton B. Non-canonical roles for metabolic enzymes and intermediates in malignant progression and metastasis. Clin Exp Metastasis. 2019;36:211–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Wong CC, Qian Y, Yu J. Interplay between epigenetics and metabolism in oncogenesis: mechanisms and therapeutic approaches. Oncogene. 2017;36:3359–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. McDonald OG, Li X, Saunders T, Tryggvadottir R, Mentch SJ, Warmoes MO, et al. Epigenomic reprogramming during pancreatic cancer progression links anabolic glucose metabolism to distant metastasis. Nat Genet. 2017;49:367–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Cui W, Popat S. Pleural mesothelioma (PM)—the status of systemic therapy. Cancer Treat Rev. 2021;100:102265.

    Article  CAS  PubMed  Google Scholar 

  62. Pinton G, Wang Z, Balzano C, Missaglia S, Tavian D, Boldorini R, et al. CDKN2A determines mesothelioma cell fate to EZH2 inhibition. Front Oncol. 2021;11:678447.

    Article  PubMed  PubMed Central  Google Scholar 

  63. McCambridge AJ, Napolitano A, Mansfield AS, Fennell DA, Sekido Y, Nowak AK, et al. Progress in the management of malignant pleural mesothelioma in 2017. J Thorac Oncol. 2018;13:606–23.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Hotta K, Fujimoto N. Current evidence and future perspectives of immune-checkpoint inhibitors in unresectable malignant pleural mesothelioma. J Immunother Cancer. 2020;8:e000461.

  65. Nakajima EC, Vellanki PJ, Larkins E, Chatterjee S, Mishra-Kalyani PS, Bi Y, et al. FDA approval summary: nivolumab in combination with ipilimumab for the treatment of unresectable malignant pleural mesothelioma. Clin Cancer Res. 2021;28:446–51.

  66. Lemoine J, Ruella M, Houot R. Overcoming intrinsic resistance of cancer cells to CAR T-cell killing. Clin Cancer Res. 2021;27:6298–306.

Download references

Acknowledgements

We thank Dr. Roni Bollag at the Georgia Cancer Center Biorepository for providing the human mesothelioma specimen and for pathological analysis. We also thank Dr. Natasha Savage for tumor specimen analysis and Dr. Kimya Jones for immunohistochemical analysis of tumor specimen. This work was supported by the National Cancer Institute grants R01 CA227433 (to MWG, YLC, NHO, KL), R01CA133085 (to KL), P01 CA125066 (to NHO), R01CA190429, and R01CA236890 (to H-FD), F30CA236436 (to JDK), and the US Department of Veterans Affairs Award CX001364 (to KL).

Author information

Authors and Affiliations

Authors

Contributions

CL, YLC, NHO, CJP, AHC, MWG, HS, HD, and KL designed the study and wrote or reviewed the manuscript. NHO and CJP provided key study materials. CL, DY, JDK, performed experiments, collected data, and analyzed data. ZL and HS performed bioinformatics analysis and data analysis.

Corresponding authors

Correspondence to Chunwan Lu or Kebin Liu.

Ethics declarations

Competing interests

AHC, MWG, and NHO have ownership interest in Ionic Pharmaceuticals, LLC. KL has an ownership interest in CheMedImmune Inc. NHO is a member of the Scientific Advisory Board of Mycosynthetix, Inc. CJP has an ownership interest in Mycosynthetix, Inc. Other authors have declared that no conflict of interest exists.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lu, C., Yang, D., Klement, J.D. et al. H3K9me3 represses G6PD expression to suppress the pentose phosphate pathway and ROS production to promote human mesothelioma growth. Oncogene 41, 2651–2662 (2022). https://doi.org/10.1038/s41388-022-02283-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-022-02283-0

This article is cited by

Search

Quick links