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:

Myc-mediated SDHA acetylation triggers epigenetic regulation of gene expression and tumorigenesis

Abstract

The transcriptional role of cMyc (or Myc) in tumorigenesis is well appreciated; however, it remains to be fully established how extensively Myc is involved in the epigenetic regulation of gene expression. Here, we show that by deactivating succinate dehydrogenase complex subunit A (SDHA) via acetylation, Myc triggers a regulatory cascade in cancer cells that leads to H3K4me3 activation and gene expression. We find that Myc facilitates the acetylation-dependent deactivation of SDHA by activating the SKP2-mediated degradation of SIRT3 deacetylase. We further demonstrate that Myc inhibition of SDH-complex activity leads to cellular succinate accumulation, which triggers H3K4me3 activation and tumour-specific gene expression. We demonstrate that acetylated SDHA at Lys 335 contributes to tumour growth in vitro and in vivo, and we confirm increased tumorigenesis in clinical samples. This study illustrates a link between acetylation-dependent SDHA deactivation and Myc-driven epigenetic regulation of gene expression, which is critical for cancer progression.

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: Myc inhibits SDH-complex activity and causes cellular succinate accumulation by inducing SDHA acetylation.
Fig. 2: Myc increases SDHA acetylation by accelerating SKP2-mediated proteasomal degradation of SIRT3.
Fig. 3: Myc enhances H3K4me3 via SDHA acetylation-mediated increase in cellular succinate.
Fig. 4: Myc-mediated succinate accumulation and H3K4me3 activation promote its target gene expression.
Fig. 5: SDHA acetylation at Lys 335 contributes to Myc-promoted tumour progression.
Fig. 6: Myc-mediated SDHA acetylation and H3K4me3 activation trigger epigenetic regulation of gene expression and tumorigenesis.

Similar content being viewed by others

Data availability

Data from this study have been deposited in the GEO. Original data from ChIP-seq and RNA-seq in P493 or CA46 cells are available in the NCBI GEO (accession numbers GSE124255 and GSE141227, respectively). The datasets generated and/or analysed during the current study are available from the corresponding author upon reasonable request. Source data for Figs. 1–5 and Extended Data Figs. 1–5 are presented within the paper.

References

  1. Eilers, M. & Eisenman, R. N. Myc’s broad reach. Genes Dev. 22, 2755–2766 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Brooks, T. A. & Hurley, L. H. The role of supercoiling in transcriptional control of MYC and its importance in molecular therapeutics. Nat. Rev. Cancer 9, 849–861 (2009).

    CAS  PubMed  Google Scholar 

  3. Dang, C. V. MYC on the path to cancer. Cell 149, 22–35 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. van Riggelen, J., Yetil, A. & Felsher, D. W. MYC as a regulator of ribosome biogenesis and protein synthesis. Nat. Rev. Cancer 10, 301–309 (2010).

    PubMed  Google Scholar 

  5. Dang, C. V. MYC, metabolism, cell growth, and tumorigenesis. Cold Spring Harb. Perspect. Med. 3, a014217 (2013).

    PubMed  PubMed Central  Google Scholar 

  6. Kim, J. W., Gao, P., Liu, Y. C., Semenza, G. L. & Dang, C. V. Hypoxia-inducible factor 1 and dysregulated c-Myc cooperatively induce vascular endothelial growth factor and metabolic switches hexokinase 2 and pyruvate dehydrogenase kinase 1. Mol. Cell. Biol. 27, 7381–7393 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Zeller, K. I. et al. Global mapping of c-Myc binding sites and target gene networks in human B cells. Proc. Natl Acad. Sci. USA 103, 17834–17839 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Guccione, E. et al. Myc-binding-site recognition in the human genome is determined by chromatin context. Nat. Cell Biol. 8, 764–770 (2006).

    CAS  PubMed  Google Scholar 

  9. Zhang, L., Sokolowski, N., Atmadibrata, B. & Liu, T. Histone demethylase JARID1B promotes cell proliferation but is downregulated by N-Myc oncoprotein. Oncol. Rep. 31, 1935–1939 (2014).

    CAS  PubMed  Google Scholar 

  10. Ullius, A. et al. The interaction of MYC with the trithorax protein ASH2L promotes gene transcription by regulating H3K27 modification. Nucleic Acids Res. 42, 6901–6920 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Santos-Rosa, H. et al. Active genes are tri-methylated at K4 of histone H3. Nature 419, 407–411 (2002).

    CAS  PubMed  Google Scholar 

  12. Schneider, R. et al. Histone H3 lysine 4 methylation patterns in higher eukaryotic genes. Nat. Cell Biol. 6, 73–77 (2004).

    CAS  PubMed  Google Scholar 

  13. Bernstein, B. E. et al. Genomic maps and comparative analysis of histone modifications in human and mouse. Cell 120, 169–181 (2005).

    CAS  PubMed  Google Scholar 

  14. Martinato, F., Cesaroni, M., Amati, B. & Guccione, E. Analysis of Myc-induced histone modifications on target chromatin. PLoS One 3, e3650 (2008).

    PubMed  PubMed Central  Google Scholar 

  15. Kim, S. C. et al. Substrate and functional diversity of lysine acetylation revealed by a proteomics survey. Mol. Cell. 23, 607–618 (2006).

    CAS  PubMed  Google Scholar 

  16. Choudhary, C. et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science 325, 834–840 (2009).

    CAS  PubMed  Google Scholar 

  17. Zhao, S. et al. Regulation of cellular metabolism by protein lysine acetylation. Science 327, 1000–1004 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Wang, Q. et al. Acetylation of metabolic enzymes coordinates carbon source utilization and metabolic flux. Science 327, 1004–1007 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Xiong, Y. & Guan, K. L. Mechanistic insights into the regulation of metabolic enzymes by acetylation. J. Cell Biol. 198, 155–164 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Gil, J., Ramirez-Torres, A. & Encarnacion-Guevara, S. Lysine acetylation and cancer: a proteomics perspective. J. Proteomics 150, 297–309 (2017).

    CAS  PubMed  Google Scholar 

  21. Baysal, B. E. et al. Mutations in SDHD, a mitochondrial complex II gene, in hereditary paraganglioma. Science 287, 848–851 (2000).

    CAS  PubMed  Google Scholar 

  22. Hao, H. X. et al. SDH5, a gene required for flavination of succinate dehydrogenase, is mutated in paraganglioma. Science 325, 1139–1142 (2009).

    CAS  PubMed  Google Scholar 

  23. Hensen, E. F. & Bayley, J. P. Recent advances in the genetics of SDH-related paraganglioma and pheochromocytoma. Fam. Cancer 10, 355–363 (2011).

    CAS  PubMed  Google Scholar 

  24. Oermann, E. K., Wu, J., Guan, K. L. & Xiong, Y. Alterations of metabolic genes and metabolites in cancer. Semin. Cell Dev. Biol. 23, 370–380 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Ricketts, C. et al. Germline SDHB mutations and familial renal cell carcinoma. J. Natl. Cancer Inst. 100, 1260–1262 (2008).

    CAS  PubMed  Google Scholar 

  26. Kim, S., Kim, D. H., Jung, W. H. & Koo, J. S. Succinate dehydrogenase expression in breast cancer. Springerplus 2, 299 (2013).

    PubMed  PubMed Central  Google Scholar 

  27. Cervera, A. M., Bayley, J. P., Devilee, P. & McCreath, K. J. Inhibition of succinate dehydrogenase dysregulates histone modification in mammalian cells. Mol. Cancer 8, 89 (2009).

    PubMed  PubMed Central  Google Scholar 

  28. Kinnaird, A., Zhao, S., Wellen, K. E. & Michelakis, E. D. Metabolic control of epigenetics in cancer. Nat. Rev. Cancer 16, 694–707 (2016).

    CAS  PubMed  Google Scholar 

  29. Letouze, E. et al. SDH mutations establish a hypermethylator phenotype in paraganglioma. Cancer Cell 23, 739–752 (2013).

    CAS  PubMed  Google Scholar 

  30. Xiao, M. et al. Inhibition of alpha-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).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Lv, L. et al. Acetylation targets the M2 isoform of pyruvate kinase for degradation through chaperone-mediated autophagy and promotes tumor growth. Mol. Cell 42, 719–730 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhao, D. et al. Lysine-5 acetylation negatively regulates lactate dehydrogenase A and is decreased in pancreatic cancer. Cancer Cell 23, 464–476 (2013).

    CAS  PubMed  Google Scholar 

  33. Tao, R. et al. Sirt3-mediated deacetylation of evolutionarily conserved lysine 122 regulates MnSOD activity in response to stress. Mol. Cell 40, 893–904 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Inuzuka, H. et al. Acetylation-dependent regulation of Skp2 function. Cell 150, 179–193 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Bretones, G. et al. SKP2 oncogene is a direct MYC target gene and MYC down-regulates p27(KIP1) through SKP2 in human leukemia cells. J. Biol. Chem. 286, 9815–9825 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Hoekstra, A. S. et al. Inactivation of SDH and FH cause loss of 5hmC and increased H3K9me3 in paraganglioma/pheochromocytoma and smooth muscle tumors. Oncotarget 6, 38777–38788 (2015).

    PubMed  PubMed Central  Google Scholar 

  37. Kooistra, S. M. & Helin, K. Molecular mechanisms and potential functions of histone demethylases. Nat. Rev. Mol. Cell Biol. 13, 297–311 (2012).

    CAS  PubMed  Google Scholar 

  38. Liu, P. S. et al. alpha-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat. Immunol. 18, 985–994 (2017).

    CAS  PubMed  Google Scholar 

  39. Nie, Z. et al. Structure-based design and discovery of potent and selective KDM5 inhibitors. Bioorg. Med. Chem. Lett. 28, 1490–1494 (2018).

    CAS  PubMed  Google Scholar 

  40. Kluk, M. J. et al. Immunohistochemical detection of MYC-driven diffuse large B-cell lymphomas. PLoS One 7, e33813 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Johnson, N. A. et al. Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J. Clin. Oncol. 30, 3452–3459 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Green, T. M. et al. High levels of nuclear MYC protein predict the presence of MYC rearrangement in diffuse large B-cell lymphoma. Am. J. Surg. Pathol. 36, 612–619 (2012).

    PubMed  Google Scholar 

  43. Barrans, S. et al. Rearrangement of MYC is associated with poor prognosis in patients with diffuse large B-cell lymphoma treated in the era of rituximab. J. Clin. Oncol. 28, 3360–3365 (2010).

    CAS  PubMed  Google Scholar 

  44. Savage, K. J. et al. MYC gene rearrangements are associated with a poor prognosis in diffuse large B-cell lymphoma patients treated with R-CHOP chemotherapy. Blood 114, 3533–3537 (2009).

    CAS  PubMed  Google Scholar 

  45. Carey, C. D. et al. Molecular classification of MYC-driven B-cell lymphomas by targeted gene expression profiling of fixed biopsy specimens. J. Mol. Diagn. 17, 19–30 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Dang, C. V. c-Myc target genes involved in cell growth, apoptosis, and metabolism. Mol. Cell. Biol. 19, 1–11 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhao, D. et al. NOTCH-induced aldehyde dehydrogenase 1A1 deacetylation promotes breast cancer stem cells. J. Clin. Invest. 124, 5453–5465 (2014).

    PubMed  PubMed Central  Google Scholar 

  48. Yang, H. et al. SIRT3-dependent GOT2 acetylation status affects the malate-aspartate NADH shuttle activity and pancreatic tumor growth. EMBO J. 34, 1110–1125 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Fan, J. et al. Tyr phosphorylation of PDP1 toggles recruitment between ACAT1 and SIRT3 to regulate the pyruvate dehydrogenase complex. Mol. Cell 53, 534–548 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Zhang, M. et al. Sirtinol promotes PEPCK1 degradation and inhibits gluconeogenesis by inhibiting deacetylase SIRT2. Sci. Rep. 7, 7 (2017).

    PubMed  PubMed Central  Google Scholar 

  51. Gao, P. et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature 458, 762–765 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Sun, L. et al. cMyc-mediated activation of serine biosynthesis pathway is critical for cancer progression under nutrient deprivation conditions. Cell Res. 25, 429–444 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Wu, G. et al. Menin enhances c-Myc-mediated transcription to promote cancer progression. Nat. Commun. 8, 15278 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Miettinen, M. et al. Succinate dehydrogenase-deficient GISTs: a clinicopathologic, immunohistochemical, and molecular genetic study of 66 gastric GISTs with predilection to young age. Am. J. Surg. Pathol. 35, 1712–1721 (2011).

    PubMed  PubMed Central  Google Scholar 

  55. Badve, S. et al. Basal-like and triple-negative breast cancers: a critical review with an emphasis on the implications for pathologists and oncologists. Mod. Pathol. 24, 157–167 (2011).

    PubMed  Google Scholar 

  56. Schuhmacher, M. et al. Control of cell growth by c-Myc in the absence of cell division. Curr. Biol. 9, 1255–1258 (1999).

    CAS  PubMed  Google Scholar 

  57. Guan, K. L., Yu, W., Lin, Y., Xiong, Y. & Zhao, S. Generation of acetyllysine antibodies and affinity enrichment of acetylated peptides. Nat. Protoc. 5, 1583–1595 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Lee, M. G., Wynder, C., Norman, J. & Shiekhattar, R. Isolation and characterization of histone H3 lysine 4 demethylase-containing complexes. Methods 40, 327–330 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Lei, Q. et al. NKX3.1 stabilizes p53, inhibits AKT activation, and blocks prostate cancer initiation caused by PTEN loss. Cancer Cell. 9, 367–378 (2006).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank C. V. Dang for critical reading of this manuscript and valuable suggestions. This work is supported in part by National Natural Science Foundation of China (91957203, 81525022, 81930083, 31571472, 81530076, 81821001), National Key R&D Program of China (2018YFA0800300, 2018YFA0107103, 2017YFA0205600), the Chinese Academy of Sciences (XDPB10), the Program for Guangdong Introducing Innovative and Entrepreneurial Teams (2017ZT07S054) and the Program of Development Foundation of Hefei Center for Physical Science and Technology (2017FXZY004). Please address all the correspondence and requests for materials to P.G. (pgao2@ustc.edu.cn).

Author information

Authors and Affiliations

Authors

Contributions

P.G. and H.Z. conceived the study and supervised the experiments. S.-T.L., D.H., P.G., X.D. and H.Z. designed the experiments. X.D. performed and analysed MS mitochondrial protein acetylation. S.-T.L., D.H., S.X., G.W., Z.J., Y.H., M.Y., N.W., R.Y., D.Y., L.W., Z.L. and X.H. performed experiments. S.S., L.Z. and K.Q. analysed ChIP-seq data and RNA-seq data. Y.C. provided clinical specimens. R.Z. and A.L. provided constructive guidance and advice. P.G., H.Z., D.H. and S.-T.L. wrote the paper. All the authors read and approved the manuscript.

Corresponding authors

Correspondence to Xiaotao Duan, Huafeng Zhang or Ping Gao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Primary Handling Editors: Ana Mateus; Elena Bellafante.

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

Extended data

Extended Data Fig. 1 Myc inhibits SDH complex activity and causes cellular succinate accumulation by inducing SDHA acetylation.

a, Western blot analysis of the nuclear marker lamin B1, cytoplasmic marker tubulin, endoplasmic reticulum marker CALR (Calreticulin) and mitochondrial marker COX4 (cytochrome c oxidase subunit IV) using the whole cell lysates or purified subcellular fractions from P493 cells. b, Diagram showing extraction of the mitochondrial fraction after P493 cells were treated with or without tetracycline (tet) for 48 hours. Mitochondrial proteins were digested, and lysine-acetylated peptides were immunoprecipitated using acetylated-lysine antibody. Enriched acetylated peptides were analyzed by nano LC-MS/MS (see Materials and Methods). c, HT1080 cells stably expressing flag-SDHA were further infected with viruses expressing non-targeting control (NTC) or Myc shRNA, followed by immunoprecipitation using anti-flag antibody, IgG (left panel) or using anti-lysine-acetylation antibody (anti-K-Ac) or IgG (right panel). Western blot analysis of Myc, flag-SDHA and SDHA acetylation. d, SDH activity was measured in HT1080 and HeLa cells infected with viruses expressing NTC or Myc shRNA for 48 hours (left panel), or in HT1080 and HeLa cells infected with pMX-GFP empty vector (EV) or pMX-GFP-Myc vector (right panel). Myc and SDHA expression were analyzed by western blotting. Data are presented as the mean ± SD of three independent experiments (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the NTC or EV group. e, SDH activity was measured in HT1080 cells infected with viruses expressing NTC or Myc shRNA (right panel) and further exposed to 10 mM NAM and 1 μM TSA. Immunoprecipitation assay using anti-SDHA antibody was performed using the above conditions. SDH activity was normalized to protein content. Data are presented as the mean ± SD of three independent experiments (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the corresponding control group. f, K335, K547 and K550 acetylated sites of SDHA were determined by LC-MS/MS. g, LC-MS/MS spectrum of the K335 acetylated peptide of SDHA. h, Primary sequence alignment of acetylated peptides from the indicated species. * denotes the acetylated K335 residues detected by nano LC-MS/MS. i, Validation of the acetyl-SDHA (K335) antibody by dot blot using a nitrocellulose membrane spotted with acetylated K335 peptide or unmodified peptide at the indicated concentration. j, Western blot analysis of Myc, SDHA K335 acetylation and endogenous SDHA levels in Raji cells expressing NTC or Myc shRNAs (sh1 and sh2), or in HT1080 cells expressing pMX-GFP empty vector (EV) or pMX-GFP-Myc vector. k, Western blot analysis of SDHA expression in HT1080 cells infected with viruses expressing NTC or SDHA shRNAs (sh1 and sh2). Cellular succinate and fumarate levels were determined using assay kits. Data are presented as the mean ± SD of three independent experiments (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the NTC group. l, HT1080 cells with stable knockdown of endogenous SDHA were subsequently infected with viruses expressing flag-wild-type-SDHA (WT), flag-SDHAK335R (K335R) or flag-SDHAK335Q (K335Q), followed by western blot analysis of SDHA K335 acetylation and measurement of succinate and fumarate. Relative succinate or fumarate levels of triplicate experiments mean ± SD are presented (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the EV group. m, Cellular succinate, fumarate or α-KG levels were determined using the LC-MS in the HT1080 cells used in (Fig. 1i, right panel). The metabolic measurements of LC-MS were normalized to cellular protein. Metabolites level of triplicate experiments mean ± SD are presented (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the indicated group. n and o, Cellular succinate, fumarate or α-KG levels were determined by assay kits in the HT1080 cells used in (Fig. 1i, right panel) (o), then succinate/α-KG ratio or fumarate/α-KG ratio was calculated (n). The metabolic measurements of LC-MS were normalized to cellular protein. Data are presented as the mean ± SD of three independent experiments (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the indicated group. n = 3 independent experiments were repeated with similar results (a, c, i, and j). Two independent LC-MS/MS experiments in P493 cells were repeated with similar results (g).

Source data

Extended Data Fig. 2 Myc increases SDHA acetylation by accelerating SKP2-mediated proteasomal degradation of SIRT3.

a, Mitochondrial proteins from P493 cells treated with or without tetracycline (tet) for 48 hours were cultured in the presence of vehicle or 1 μM TSA for 18 hours, followed by western blot analysis of lysine-acetylation (K-Ac). COX4 served as a loading control. b, Quantitative real-time PCR analysis of SIRT3 mRNA levels in P493 cells treated with or without tetracycline (tet) for 24 hours. Data are presented as the mean ± SD of three independent experiments (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the indicated group. c, Western blot analysis of SIRT3 in P493 cells treated with or without tetracycline (tet) for 48 hours and further cultured in the presence of vehicle or 10 μM lactacystin for 8 hours. Tubulin served as a loading control. d, Immunoprecipitation was performed using anti-flag antibody or IgG in HEK293T cells transfected with flag-SKP2 and SIRT3, followed by western blot analysis of SIRT3 and SKP2 (left panel). Immunoprecipitation was performed using anti-SIRT3 antibody or IgG in HT1080 cells, followed by western blot of SIRT3 and SKP2 (right panel). Tubulin served as a loading control. e, Quantitative real-time PCR and western blot analysis of SKP2 expression in P493 cells treated with or without tetracycline (tet) for 24 and 48 hours. Data are presented as the mean ± SD of three independent experiments (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the indicated group. f, Western blot analysis of SKP2 in HT1080 cells infected with viruses expressing NTC or Myc shRNA (left panel) or in HT1080 cells infected with viruses expressing pMX-GFP empty vector (EV) or pMX-GFP-Myc vector (right panel). Tubulin served as a loading control. g, Western blot analysis of SKP2 and SIRT3 protein in P493 cells (upper left panel), HT1080 cells (bottom left panel) or CA46 cells (right panel) infected with viruses expressing NTC or SKP2 shRNAs (sh1 and sh2), or infected with EV or pSIN-3×flag-SKP2. Tubulin served as a loading control. h, HT1080 cells stably expressing NTC or Myc shRNA were subsequently infected with viruses expressing empty vector (EV) or flag-SKP2 vector, followed by western blot analysis of Myc, flag-SKP2 and SIRT3 expression. i, HT1080 cells stably expressing NTC or SKP2 shRNAs were subsequently infected with viruses of EV or Myc, followed by western blot analysis of Myc, SKP2 and SIRT3 expression. Tubulin served as a loading control. j, Immunoprecipitation was performed using anti-flag antibody or IgG in HEK293T cells transfected with flag-SDHA and SIRT3, followed by western blot analysis of SIRT3 and flag-SDHA. k, CA46 cells stably expressing empty vector (EV) or SIRT3 were subsequently infected with viruses expressing NTC or Myc shRNA (left panel), or CA46 cells stably expressing NTC or SIRT3 shRNA were further infected with viruses expressing NTC or Myc shRNA (right panel), followed by western blot analysis of Myc, SIRT3, SDHA and SDHA K335 acetylation. l, P493 cells stably expressing EV or 3×flag-SIRT3 were treated with or without tetracycline (tet) for 48 hours, (left panel). P493 cells stably expressing NTC or SIRT3 shRNA were treated with or without tetracycline (tet) for 48 hours (right panel), followed by western blot analysis of SDHA K335 acetylation, SDHA, SIRT3 and Myc. Tubulin served as a loading control. m-p, Cellular succinate or α-KG levels were determined by LC-MS in the HT1080 cells used in Fig. 2i, then succinate/α-KG ratio was calculated. The succinate or α-KG levels were normalized to cellular protein. Succinate or α-KG level of triplicate experiments mean ± SD are presented (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the indicated group. q, Measurement of SDH activity in the CA46 cells used in (k, right panel). Relative enzyme activities of triplicate experiments mean ± SD are presented (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the indicated group. r-t, Cellular succinate or α-KG levels were determined with LC-MS in the CA46 cells used in (k, right panel), then succinate/α-KG ratio was calculated (r). The succinate or α-KG levels were normalized to cellular protein (s, t). Succinate or α-KG level of triplicate experiments mean ± SD are presented (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the indicated group. u, Measurement of SDH activity in the P493 cells used in (l, right panel). Relative enzyme activities of triplicate experiments ± SD are presented (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the indicated group. v-x, Cellular succinate or α-KG levels were determined with LC-MS in the P493 cells used in (l, right panel), then succinate/α-KG ratio was calculated (v). The succinate or α-KG levels were normalized to cellular protein (w, x). Succinate or α-KG level of triplicate experiments mean ± SD are presented (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the indicated group. n = 3 independent experiments were repeated with similar results (a, c, d and f-l).

Source data

Extended Data Fig. 3 Myc enhances H3K4me3 via SDHA acetylation-mediated increase in cellular succinate.

a, Western blot analysis of the indicated histone methylation markers and Myc in Raji cells infected with viruses expressing NTC or Myc shRNAs (sh1, sh2). b, P493 cells treated with or without tetracycline (tet) were exposed to 5 mM dimethyl α-KG (DM-αKG) for 8 hours, followed by western blot analysis of H3K4me3 and Myc from nuclear lysates and whole cell lysates, respectively. c, P493 cells treated with or without tetracycline (tet) were exposed to 10 mM dimethyl succinate (DMS) or/and 5 mM dimethyl α-KG (DM-αKG), followed by western blot analysis of H3K4me3 and Myc from nuclear lysates and whole cell lysates, respectively. H3 and tubulin served as loading controls. d, Western blot analysis of the indicated histone methylation markers from nuclear lysates and whole cell lysates in HT1080 cells infected with viruses expressing NTC or SIRT3 shRNAs (sh1 and sh2). Tubulin and H3 served as loading controls. WCL, whole-cell lysate; NL, nuclear lysate. e, HT1080 cells infected with viruses of NTC or Myc shRNA were cultured in the presence of vehicle or 10 mM NAM, followed by western blot analysis of H3K4me3, Myc and SIRT3 from nuclear lysates and whole cell lysates. Tubulin and H3 served as loading controls. WCL, whole-cell lysates; NL, nuclear lysates. f, HT1080 cells stably expressing pSIN-EV empty vector (EV) or pSIN-SIRT3 vector were subsequently infected with viruses expressing pMX-GFP empty vector (EV) or pMX-GFP-Myc vector (left panel), or HT1080 cells stably expressing NTC or SIRT3 shRNA were subsequently infected with viruses expressing NTC or Myc shRNA (right panel), followed by western blot analysis of H3K4me3, Myc and SIRT3 levels. g, HT1080 cells with stable knockdown of endogenous SDHA were infected with viruses expressing flag-wild-type-SDHA (WT) or flag-SDHAK335Q (K335Q), followed by further infection with viruses of NTC or SIRT3 shRNA and NTC or Myc shRNA. Western blot analysis of H3K4me3, Myc and SIRT3 levels. h, Western blot analysis of the in vitro demethylation reactions using H3K4me3 peptide as substrates. Antibodies used are indicated. i, HT1080 cells stably expressing NTC or sh-Myc were subsequently infected with viruses expressing NTC or sh-KDM5A, followed by western blot analysis of Myc, H3K4me3, KDM5A, H3 and tubulin expression. WCL, whole-cell lysates; NL, nuclear lysates. j, P493 cells treated with or without tetracycline (tet) for 48 h were cultured in the presence of vehicle or 10 mM dimethyl succinate (DMS) for 24 h, followed by western blot analysis of H3K4me3 (up right panel) and ChIP-seq analysis of H3K4me3 around gene TSS regions. Heat map showing H3K4me3 levels in TSS regions for the top 2500 gene promoters (left panel). Distribution analysis of H3K4me3 tags around the TSS regions of the top 2500 genes (bottom right panel). n = 3 independent experiments for western blot (up right panel) were repeated with similar results. One replicate (n = 1, each group) was used for ChIP-seq assay of P493 cells. k, H3K4me3 markers of ChIP-sequencing traces in P493 cells for the indicated genes were determined by IGV software analysis. For IGV analysis, one replicate (n = 1, each group) was used for H3K4me3 analysis in ChIP-seq assay of P493 cells. l, ChIP assay analysis of the occupancy of H3K4me3 on the indicated gene promoters in the CA46 cells used in Fig. 3g (left panel) or P493 cells used in Extended Data Fig. 3j (right panel). Data are presented as the mean ± SD of three independent experiments (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the NTC–DMS group or shMyc–DMS group (left panel); P < 0.05 compared with the –tet–DMS group or +tet–DMS group (right panel). n = 3 independent experiments were repeated with similar results (a-i).

Source data

Extended Data Fig. 4 Myc-mediated succinate accumulation and H3K4me3 activation promote its target gene expression.

a, CA46 cells treated with viruses expressing NTC or Myc shRNAs were cultured in the presence of vehicle or 10 mM dimethyl-succinate (DMS) for 24 hours, followed by the extraction of total RNAs for the RNA-Seq experiment. Heat map showing all the regulated genes. b, P493 cells treated with or without tetracycline (tet) were cultured in the presence of vehicle or 10 mM dimethyl-succinate (DMS) for 24 hours, followed by the extraction of total RNAs for the RNA-Seq experiment. Heat map showing all the regulated genes. c, Heat map of the RNA-seq results showing that the genes with down-regulated expression following Myc depletion were subsequently rescued by DMS treatment in P493 cells (fold-change >1.5). d, Venn diagram showing overlapping genes co-regulated by Myc and succinate based on the RNA-Seq data and ChIP-Seq data in P493 cells. Correlation significance between genes with mRNA levels and genes with H3K4me3 that were rescued by DMS based on RNA-seq and ChIP-seq, respectively, were determined using a two-tailed Spearman test, P < 0.001. e, Analysis of 585 overlapping genes by Gene Ontology (GO) term enrichment. P values were analysed with two-tailed Student’s t-test. The sufficient RNA-seq samples (n = 1, each group) were analyzed in Gene Ontology (GO) term enrichment. f, ChIP assay analysis of the occupancy of H3K4me3 on the indicated gene promoters in the CA46 cells used in (a). Data are presented as the mean ± SD of three independent experiments (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the NTC –DMS group; P < 0.05 compared with the shMyc–DMS group. g, ChIP assay analysis of the occupancy of H3K4me3 on the indicated gene promoters in the P493 cells used in (b). Data are presented as the mean ± SD of three independent experiments. Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the –tet –DMS group; P < 0.05 compared with the +tet –DMS group. h, P493 cells treated with or without tetracycline (tet) for 48 hours were cultured in the presence of vehicle or 500 μM 3-nitropropionic acid (3-NPA) for 24 hours, followed by western blot analysis of H3K4me3. i, Heat map of the RNA-seq data showing that the genes with down-regulated expression following Myc depletion were markedly rescued by 3-NPA treatment in P493 cells (fold-change >1.5). The statistical significance between Myc- and succinate-regulated genes was determined using Fisher’s exact probability, P < 0.001. The RNA-seq samples (n = 1, each group) used to analysed for Heatmap. j, ChIP assay studying the occupancy of H3K4me3 on the indicated gene promoters in the P493 cells used in i. Data are presented as the mean ± SD of three independent experiments (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared to the –tet +PBS group; P < 0.05 compared to the +tet +PBS group. k, The mRNA levels of the indicated genes were determined by RT-qPCR in the P493 cells used in b. Data are presented as the mean ± SD of three independent experiments (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the –tet –DMS group; P < 0.05 compared with the +tet –DMS group. l, The indicated proteins were determined by western blotting in the P493 cells used in k. m, P493 cells with endogenous SDHA knockdown were further constructed to stably express flag-wild-type-SDHA (WT) or flag-SDHAK335R (K335R), followed by treatment with tetracycline (tet) for 48 h or washing to remove tet for 48 hours. The mRNA levels of the indicated genes were determined by RT-qPCR. Data are presented as the mean ± SD of three independent experiments (n = 3). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the +tet +WT group; P < 0.05 compared with the wash +WT group. n, The indicated proteins were determined by western blotting in the P493 cells used in m. The statistical significance between Myc- and succinate-regulated genes was determined using the two-sided Fisher’s exact probability, P < 0.001. The sufficient RNA-seq samples (n = 1, each group) used to analysed for Heatmap (a-c). n = 3 independent experiments were repeated with similar results (h, l and n). Tubulin and H3 served as loading controls. WCL, whole-cell lysates; NL, nuclear lysates.

Source data

Extended Data Fig. 5 SDHA acetylation at lysine 335 contributes to Myc-promoted tumour progression.

a, P493 cells with stable knockdown of endogenous SDHA were further infected with viruses expressing flag-wild-type-SDHA (WT), flag-SDHAK335R (K335R) or flag-SDHAK335Q (K335Q). Cell growth curves were determined by trypan blue counting. Data are presented as the mean ± SD (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the corresponding indicated group. b, Endogenous SDHA was knocked down in empty vector (EV) or Myc overexpressing HT1080 cells, followed by further infection with viruses expressing flag-wild-type-SDHA (WT) or flag-SDHA-K335Q (K335Q). The cells were treated with or without 5 mM dimethyl α-KG (DM-αKG) (fresh medium containing fresh DM-αKG was replaced daily), and the cell growth of HT1080 cells was determined by trypan blue staining. Data are presented as the mean ± SD (n = 5, each group). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared between indicated groups. c, Endogenous SDHA was knocked down in empty vector (EV) or Myc overexpressing HT1080 cells, followed by further infection with viruses expressing flag-wild-type-SDHA (WT) or flag-SDHA-K335R (K335R). The cells were treated with or without 5 μM CPI-455, followed by cell growth analysis with trypan blue staining. Data are presented as the mean ± SD (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the indicated group. d, The tumor mass of the extracted tumors in Fig. 5d was measured. Data are presented as mean ± SEM (n = 5, each group). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the corresponding indicated group. e and f, The succinate and α-KG levels were measured using detection kits with cell lysates from xenograft tumor tissues. The succinate and α-KG level were normalized to cellular protein. Data are presented as the mean ± SD (n = 5, each group). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared with the indicated group. g and h, HT1080 cells used in b were injected subcutaneously into BALB/c nude mice (n = 5, each group). Mice were treated with daily i.p injection of Dimethyl α-KG (0.6 g/kg body weight) or PBS, starting one day before tumor cell implantation. Tumor growth curves were measured starting from 5 days after inoculation (g). Tumors were extracted and compared at the end of the experiment (h). Data are presented as the mean ± SD (n = 5, each group). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared between indicated groups. i, Levels of Myc, SIRT3, SDHA, and K335 acetylation of SDHA and H3K4me3 were determined by western blot using the nuclear lysates or whole cell lysates of tumor tissues from each group as in h. n = 5 independent xenograft tumor tissues lysates in each group were repeated by western blot with similar results. Tubulin and H3 served as loading controls. WCL, whole cell lysates; NL, nuclear lysates. j and k, HT1080 cells used in c were injected subcutaneously into BALB/c nude mice (n = 5 for each group). Mice were treated with daily i.p injection of CPI- 455 (20 mg/kg body weight) or vehicle (4%DMSO + 35%PEG 300 + 2%Tween 80+PBS), starting one day before tumor cell implantation. Tumor growth curves were measured starting from 5 days after inoculation (j). Tumors were extracted and compared at the end of the experiment (k). Data are presented as the mean ± SD (n = 5, each group). Group differences are analyzed by the two-tailed Student’s t-test. P < 0.05 compared between indicated groups. l, Levels of Myc, SIRT3, SDHA, and K335 acetylation of SDHA and H3K4me3 were determined by western blot using the nuclear lysates or whole cell lysates of tumor tissues from each group as in k. Tubulin and H3 served as loading controls. WCL, whole-cell lysates; NL, nuclear lysates. n = 5 independent xenograft tumor tissues lysates in each group were repeated by western blot with similar results.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 3–6

Reporting Summary

Supplementary Tables

Supplementary Tables 1 and 2

Source data

Source Data Fig. 1

Unprocessed western blots. Please note that the slight difference between WB images of Fig. 1c (SDHA), 1e (SDHA), 1h (Myc) and the corresponding uncropped images in source data were caused by different exposure time.

Source Data Fig. 2

Unprocessed western blots. Please note that the slight difference between WB images of Fig. 2b (Tubulin), Fig. 2c (SIRT3), Fig. 2e (Myc), Fig. 2g (SDHA and Tubulin) and the corresponding uncropped images in source data were caused by different exposure time.

Source Data Fig. 3

Unprocessed western blots. Please note that the slight difference between WB images of Fig. 3a (Myc) and the corresponding uncropped images in source data were caused by different exposure time.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Unprocessed western blots. Please note that the slight difference between WB images of Fig. 5e (H3K4me3) and the corresponding uncropped images in source data were caused by different exposure time.

Source Data Extended Data Fig. 1

Unprocessed western blots. Please note that the slight difference between WB images of Extended Data Fig. 1c (Tubulin) and the corresponding uncropped images in source data were caused by different exposure time.

Source Data Extended Data Fig. 2

Unprocessed western blots. Please note that the slight difference between WB images of Extended Data Fig. 2g (SKP2) and the corresponding uncropped images in source data were caused by different exposure time.

Source Data Extended Data Fig. 3

Unprocessed western blots. Please note that the slight difference between WB images of Extended Data Fig. 3g (SIRT3) and the corresponding uncropped images in source data were caused by different exposure time.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, ST., Huang, D., Shen, S. et al. Myc-mediated SDHA acetylation triggers epigenetic regulation of gene expression and tumorigenesis. Nat Metab 2, 256–269 (2020). https://doi.org/10.1038/s42255-020-0179-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-0179-8

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