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:

NADPH levels affect cellular epigenetic state by inhibiting HDAC3–Ncor complex

Abstract

NADPH has long been recognized as a key cofactor for antioxidant defence and reductive biosynthesis. Here we report a metabolism-independent function of NADPH in modulating epigenetic status and transcription. We find that the reduction of cellular NADPH levels, achieved by silencing malic enzyme or glucose-6-phosphate dehydrogenase, impairs global histone acetylation and transcription in both adipocytes and tumour cells. These effects can be reversed by supplementation with exogenous NADPH or by inhibition of histone deacetylase 3 (HDAC3). Mechanistically, NADPH directly interacts with HDAC3 and interrupts the association between HDAC3 and its co-activator nuclear receptor corepressor 2 (Ncor2; SMRT) or Ncor1, thereby impairing HDAC3 activation. Interestingly, NADPH and the inositol tetraphosphate molecule Ins(1,4,5,6)P4 appear to bind to the same domains on HDAC3, with NADPH having a higher affinity towards HDAC3 than Ins(1,4,5,6)P4. Thus, while Ins(1,4,5,6)P4 promotes formation of the HDAC3–Ncor complex, NADPH inhibits it. Collectively, our findings uncover a previously unidentified and metabolism-independent role of NADPH in controlling epigenetic change and gene expression by acting as an endogenous inhibitor of HDAC3.

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: Malic enzyme silencing suppresses adipocytic gene transcription and global histone acetylation, resulting in a block to adipocyte differentiation.
Fig. 2: Malic enzymes regulate histone acetylation through HDAC3.
Fig. 3: HDAC3 mediates ME-regulated gene transcription.
Fig. 4: NADPH directly binds and inhibits HDAC3 in vitro.
Fig. 5: NADPH affects histone acetylation through inhibition of HDAC3.
Fig. 6: NADPH binds to HDAC3 and abrogates assembly of HDAC3–DAD complexes.
Fig. 7: NADPH change reprograms histone acetylation and transcription.

Similar content being viewed by others

Data availability

Uncropped versions of blots and source data are provided with this paper. The CHIP–seq data have been deposited in the Gene Expression Omnibus under accession GSE137694. The remaining data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Lunt, S. Y. & Vander Heiden, M. G. Aerobic glycolysis: meeting the metabolic requirements of cell proliferation. Annu. Rev. Cell Dev. Biol. 27, 441–464 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Article  CAS  Google Scholar 

  3. Du, W. et al. TAp73 enhances the pentose phosphate pathway and supports cell proliferation. Nat. Cell Biol. 15, 991–1000 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Fan, J. et al. Quantitative flux analysis reveals folate-dependent NADPH production. Nature 510, 298–302 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Jeon, S. M., Chandel, N. S. & Hay, N. AMPK regulates NADPH homeostasis to promote tumour cell survival during energy stress. Nature 485, 661–665 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Jiang, P., Du, W., Mancuso, A., Wellen, K. E. & Yang, X. Reciprocal regulation of p53 and malic enzymes modulates metabolism and senescence. Nature 493, 689–693 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Liu, L. et al. Malic enzyme tracers reveal hypoxia-induced switch in adipocyte NADPH pathway usage. Nat. Chem. Biol. 12, 345–352 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jiang, P. et al. p53 regulates biosynthesis through direct inactivation of glucose-6- phosphate dehydrogenase. Nat. Cell Biol. 13, 310–316 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  10. Comerford, S. A. et al. Acetate dependence of tumors. Cell 159, 1591–1602 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Montgomery, D. C., Sorum, A. W., Guasch, L., Nicklaus, M. C. & Meier, J. L. Metabolic regulation of histone acetyltransferases by endogenous Acyl-CoA cofactors. Chem. Biol. 22, 1030–1039 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Su, X., Wellen, K. E. & Rabinowitz, J. D. Metabolic control of methylation and acetylation. Curr. Opin. Chem. Biol. 30, 52–60 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Takahashi, H., McCaffery, J. M., Irizarry, R. A. & Boeke, J. D. Nucleocytosolic acetyl-coenzyme a synthetase is required for histone acetylation and global transcription. Mol. Cell 23, 207–217 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. 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 

  15. Cohen, H. Y. et al. Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science 305, 390–392 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Imai, S., Armstrong, C. M., Kaeberlein, M. & Guarente, L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 403, 795–800 (2000).

    Article  CAS  PubMed  Google Scholar 

  17. Lu, C. & Thompson, C. B. Metabolic regulation of epigenetics. Cell Metab. 16, 9–17 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Yang, H. et al. Nutrient-sensitive mitochondrial NAD+ levels dictate cell survival. Cell 130, 1095–1107 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Perissi, V., Jepsen, K., Glass, C. K. & Rosenfeld, M. G. Deconstructing repression: evolving models of co-repressor action. Nat. Rev. Genet. 11, 109–123 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Sun, Z. et al. Deacetylase-independent function of HDAC3 in transcription and metabolism requires nuclear receptor corepressor. Mol. Cell 52, 769–782 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Guenther, M. G., Barak, O. & Lazar, M. A. The SMRT and N-CoR corepressors are activating cofactors for histone deacetylase 3. Mol. Cell. Biol. 21, 6091–6101 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Watson, P. J., Fairall, L., Santos, G. M. & Schwabe, J. W. Structure of HDAC3 bound to co- repressor and inositol tetraphosphate. Nature 481, 335–340 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Rubin, C. S., Hirsch, A., Fung, C. & Rosen, O. M. Development of hormone receptors and hormonal responsiveness in vitro. Insulin receptors and insulin sensitivity in the preadipocyte and adipocyte forms of 3T3-L1 cells. J. Biol. Chem. 253, 7570–7578 (1978).

    Article  CAS  PubMed  Google Scholar 

  24. Cristancho, A. G. & Lazar, M. A. Forming functional fat: a growing understanding of adipocyte differentiation. Nat. Rev. Mol. Cell Biol. 12, 722–734 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Molchadsky, A. et al. p53 is required for brown adipogenic differentiation and has a protective role against diet-induced obesity. Cell Death Differ. 20, 774–783 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Yoo, E. J., Chung, J. J., Choe, S. S., Kim, K. H. & Kim, J. B. Down-regulation of histone deacetylases stimulates adipocyte differentiation. J. Biol. Chem. 281, 6608–6615 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Johnsson, A., Xue-Franzen, Y., Lundin, M. & Wright, A. P. H. Stress-specific role of fission yeast Gcn5 histone acetyltransferase in programming a subset of stress response genes. Eukaryot. Cell 5, 1337–1346 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Menzies, K. J., Zhang, H., Katsyuba, E. & Auwerx, J. Protein acetylation in metabolism—metabolites and cofactors. Nat. Rev. Endocrinol. 12, 43–60 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Shi, L. & Tu, B. P. Acetyl-CoA and the regulation of metabolism: mechanisms and consequences. Curr. Opin. Cell Biol. 33, 125–131 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wood, M., Rymarchyk, S., Zheng, S. & Cen, Y. Trichostatin A inhibits deacetylation of histone H3 and p53 by SIRT6. Arch. Biochem. Biophys. 638, 8–17 (2018).

    Article  CAS  PubMed  Google Scholar 

  31. Yoshida, M. Potent and specific inhibition of mammalian histone deacetylase both in vivo and in vitro by trichostatin A. Tanpakushitsu Kakusan Koso 52, 1788–1789 (2007).

    PubMed  Google Scholar 

  32. Saito, A. et al. A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proc. Natl Acad. Sci. USA 96, 4592–4597 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Li, Z. Y. et al. Histone deacetylase inhibitor RGFP109 overcomes temozolomide resistance by blocking NF-κB-dependent transcription in glioblastoma cell lines. Neurochem. Res. 41, 3192–3205 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Hideshima, T. et al. Discovery of selective small-molecule HDAC6 inhibitor for overcoming proteasome inhibitor resistance in multiple myeloma. Proc. Natl. Acad. Sci. USA 113, 13162–13167 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Marek, L. et al. Histone deacetylase (HDAC) inhibitors with a novel connecting unit linker region reveal a selectivity profile for HDAC4 and HDAC5 with improved activity against chemoresistant cancer cells. J. Med. Chem. 56, 427–436 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Malvaez, M. et al. HDAC3-selective inhibitor enhances extinction of cocaine-seeking behavior in a persistent manner. Proc. Natl Acad. Sci. USA 110, 2647–2652 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Cracan, V., Titov, D. V., Shen, H., Grabarek, Z. & Mootha, V. K. A genetically encoded tool for manipulation of NADP+/NADPH in living cells. Nat. Chem. Biol. 13, 1088–1095 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Trott, O. & Olson, A. J. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading. J. Comput. Chem. 31, 455–461 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Arrar, M., Turnham, R., Pierce, L., de Oliveira, C. A. & McCammon, J. A. Structural insight into the separate roles of inositol tetraphosphate and deacetylase-activating domain in activation of histone deacetylase 3. Protein Sci. 22, 83–92 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Vogelauer, M., Krall, A. S., McBrian, M. A., Li, J. Y. & Kurdistani, S. K. Stimulation of histone deacetylase activity by metabolites of intermediary metabolism. J. Biol. Chem. 287, 32006–32016 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Alenghat, T. et al. Nuclear receptor corepressor and histone deacetylase 3 govern circadian metabolic physiology. Nature 456, 997–1000 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Feng, L. et al. Nonreciprocal light propagation in a silicon photonic circuit. Science 333, 729–733 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Rey, G. et al. The pentose phosphate pathway regulates the circadian clock. Cell Metab. 24, 462–473 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Zhang, Y. et al. Model-based analysis of ChIP–seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ramirez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Su, R. et al. R-2HG exhibits anti-tumor activity by targeting FTO/m6A/MYC/CEBPA signaling. Cell 172, 90–105 (2018).

    Article  CAS  PubMed  Google Scholar 

  50. Case, D. A. et al. The amber biomolecular simulation programs. J. Comput. Chem. 26, 1668–1688 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Maier, J. A. et al. ff14SB: improving the accuracy of protein side chain and backbone parameters from ff99SB. J. Chem. Theory Comput. 11, 3696–3713 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Holmberg, N., Ryde, U. & Bulow, L. Redesign of the coenzyme specificity in l-lactate dehydrogenase from bacillus stearothermophilus using site-directed mutagenesis and media engineering. Protein Eng. 12, 851–856 (1999).

    Article  CAS  PubMed  Google Scholar 

  53. Wang, J., Wolf, R. M., Caldwell, J. W., Kollman, P. A. & Case, D. A. Development and testing of a general amber force field. J. Comput. Chem. 25, 1157–1174 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank X. Wang and H. Liu for helping with analysis of ChIP–seq data. This work was supported by the National Key Research and Development Program of China (2019YFA0802600), CAMS Innovation Fund for Medical Sciences (2016-I2M-4-002), the National Natural Science Foundation of China (81672766), State Key Laboratory Special Fund (2060204), CAMS Basic Research Fund (2019-RC-HL-007, 2016ZX310186 and 2016RC310038) to W.D. and a postdoctoral fellowship at Peking-Tsinghua Center for Life Sciences and National Science Foundation for Young Scholars of China (31601154) and China Postdoctoral Science Foundation (2016M601005; to J.Q.)

Author information

Authors and Affiliations

Authors

Contributions

W.L., J.K. and J.Q. performed all experiments except those mentioned below. Y.P. and Y.X. performed the molecular docking calculation and MD simulations and interpreted the data. L.L. and Z.Z. helped with animal experiments. W.D. designed the experiments. W.D. and W.L. analysed the data. W.D. wrote the manuscript with the help of Y.X. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Yi Xue or Wenjing Du.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Primary Handling Editor: Christoph Schmitt. Nature Metabolism thanks Michael Parker and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 ME silencing impairs adipocyte differentiation, histone acetylation and genes transcription.

a, b, 3T3-L1 preadipocytes transfected with control siRNA, or different sets of ME1 (a) or ME2 siRNAs (b) as indicated were induced to differentiate for 7 days. The accumulation of differentiation-induced lipid droplets was visualized by Oil Red O staining (left). Oil Red O staining was quantified (right, top panel). The protein expression is shown below. c, d, mRNA expression of 3T3-L1 preadipocytes transfected with control siRNA, or different sets of ME1 (c) or ME2 siRNAs (d) as indicated before (Day 0) and after differentiating for 7 days (Day 7). e, mRNA expression of control or ME knockout 3T3-L1 cells using sgRNA CRISPR/Cas9. fh, 3T3-L1 preadipocytes transfected with control, ME1, ME2 and/or p53 siRNAs as indicated were induced to differentiate for 7 days. Cells were stained with Oil Red O (f, top). Oil Red O staining was quantified (f, bottom). Protein expression (g) and mRNA levels (h) are shown. i, Western blots of 3T3-L1 preadipocytes transfected with control siRNA, or different sets of ME1 (left) or ME2 siRNAs (right) as indicated after 7 days of differentiation. j, Protein expression of control or ME knockout 3T3-L1 cells using sgRNA CRISPR/Cas9 after differentiating for 7 days. In a, b, f, n = 3 biological replicates; In c, d, e, h, n = 3 technical replicates of one out of three independent experiments with similar results. Data are mean ± s.d. Statistical significance was determined by two-tailed unpaired t-test. In a, b, f, g, i, j, Oil Red O staining images and western blots represent three independent experiments. For all western blot data, histone expression was analyzed using acid-extracted cell lysates, total H3 was used as loading control; all other protein expression was assessed with total cell lysates, Actin was used as loading control. The samples derived from the same experiment and the blots were processed in parallel.

Source data

Extended Data Fig. 2 MEs inhibition resulted in decreased histone acetylation and glycolytic genes expression.

U2OS cells were transfected with control, ME1 or ME2 siRNA for 48 hours. a, RNA was extracted and expressions of glucose metabolism genes were analyzed by qRT-PCR. b, Acid extracts were analyzed by western blot with antibodies against indicated acetylated histones. Expressions of actin and total H3 were determined as loading controls. Western blots represent three independent experiments. c, Relative glucose consumption and lactate production were determined by YSI2700 Biochemistry Analyzer. In a, n = 3 technical replicates of one out of three independent experiments with similar results; In c, n = 3 biological replicates. Data are mean ± s.d.. Statistical significance was determined by two-tailed unpaired t-test.

Source data

Extended Data Fig. 3 HDAC3 mediates ME-regulated histone acetylation and genes expression in adipocytes and tumour cells.

a–c, 3T3-L1 preadipocytes transfected with control, ME1 or ME2 siRNA were induced to differentiate for 7 days with or without 0.1 μM TSA. (a) Cells were fixed and stained with Oil Red O (left). Oil Red O staining was quantified (right). (b) Relative gene expression was assessed by qRT-PCR. (c) Protein expression was analyzed by western blotting with specific antibodies. d, 3T3-L1 preadipocytes treated with control, ME1 or ME2 siRNA were induced to differentiate for 7 days in the absence or presence of 50 μM NAM. Protein expression was analyzed by western blot. e, Protein expression of U2OS cells transfected with control, ME1 or ME2 siRNA in presence of HDAC1-6 siRNAs respectively as indicated. f, i, 3T3-L1 preadipocytes treated with control (−), ME1 or ME2 siRNA were induced to differentiate for 7 days with or without 10 μM RGFP966. Protein expression (f) and mRNA levels (i) are shown. g, j, U2OS and HCT116 cells were transfected with control (−), ME1 or ME2 siRNA and 2 days later treated with 10 μM RGFP966 for another 24 hours. Protein expression (g) and relative Glut4 expression (j) are shown. h, 3T3-L1 preadipocytes transfected with control or HDAC3 siRNAs were induced to differentiate for 7 days. Relative mRNA expression was assessed by qRT-PCR. In a, n = 3 biological replicates; In b, h, i, j, n = 3 technical replicates of one out of three independent experiments with similar results. Data are mean ± s.d.. Statistical significance was determined by two-tailed unpaired t-test. NS, not significant. In a, c, d, e, f, g, Oil Red O staining images and western blots represent three independent experiments. For all western blot data, histone expression was analyzed using acid-extracted cell lysates, total H3 was used as loading control; all other protein expression was assessed with total cell lysates, Actin was used as loading control. The samples derived from the same experiment and the blots were processed in parallel.

Source data

Extended Data Fig. 4 Malic enzymes affect cellular NADPH levels.

a, 3T3-L1 preadipocytes were induced to differentiate for 7 days. Before (day 0) and after differentiation induction (day 7), cells were fractionated and cytoplasmic (including mitochondria) and nuclear protein extracts were analyzed by western blot using indicated antibodies. Tubulin and total H3 were used as fraction and loading controls. Western blots represent three independent experiments. b, 3T3-L1 preadipocytes transfected with control, ME1 or ME2 siRNA were stimulated to differentiate for 7 days. Cellular NADPH levels were determined. Protein expression was determined by western blot. Actin was used as loading control. Western blots represent three independent experiments. c, Related to Figure. 2a and 2c. 3T3-L1 preadipocytes transfected with control siRNA, or two sets of ME1 or ME2 siRNAs as indicated, NADPH levels were determined. d, Adipose tissues from ME1 or ME2 knock-out mice (n = 3) were prepared for assaying NADPH levels. e, Tissues from ME1 or ME2 knock-out mice (n = 3) were analyzed NADPH levels using NADPH quantification kit. In b, c, d, e, n = 3 biological replicates; Data are mean ± s.d.. Statistical significance was determined by two-tailed unpaired t-test.

Source data

Extended Data Fig. 5 NADPH is an endogenous inhibitor of HDAC3, ROS and acetyl-CoA are not involved in ME-mediated histone acetylation modification.

ac, 3T3-L1 preadipocytes were transfected with increased amount of NADPH using X-tremeGENE HP DNA Transfection reagent for 24h. (a) Cellular NADPH levels were examined. b, Cellular HDAC3 enzymatic activity were analyzed. HDAC3 expression were detected by western blotting. Actin was used as loading control. c, HAT activity of nucleic extracts was examined. d, Nucleic extracts from 3T3-L1 preadipocytes were incubated with increased amount of NADPH as indicated for 6h, and then HAT activity were determined. e, 3T3-L1 preadipocytes transfected with control (−), ME1 or ME2 siRNA were induced to differentiate for 7 days in the absence or presence of 5 mM NAC (N-acetyl-L-cysteine). Protein expression is shown (left). Cellular ROS levels were detected by FACS with DCF (2,7-dichlorodihydrofluorescein diacetate) staining (right). f, g, 3T3-L1 preadipocytes transfected with control (−), ME1 or ME2 siRNA were induced to differentiate for 7 days in the absence or presence of 10mM GSH as indicated. NADPH levels (f right panel) and GSH levels (g) were determined before differentiation (with 10mM GSH for 48h). Protein expression (f left panel) was analyzed after differentiation. h, 3T3-L1 preadipocytes transfected with control (−), ME1 or ME2 siRNA were induced to differentiate for 7 days in the absence or presence of 5 mM acetate combined with or without 10 μM NADPH as indicated. Protein expression was determined using western blot. In a, b, c, d, e, f, g, n = 3 biological replicates. Data are mean ± s.d.. Statistical significance was determined by two-tailed unpaired t-test. NS, not significant. For b (bottom panel), e (left panel), f (left panel), h, western blots represent three independent experiments. Histone expression was analyzed using acid-extracted cell lysates, total H3 was used as loading control; all other protein expression was assessed with total cell lysates, Actin was used as loading control. The samples derived from the same experiment and the blots were processed in parallel.

Source data

Extended Data Fig. 6 NADPH disrupts HDAC3-DAD association.

a, b, 3T3-L1 preadipocytes transfected with control, ME1 or ME2 siRNA as indicated were induced to differentiate for 7 days in the absence or presence of 20 μM NADPH (NADPH were transfected into cells using X-tremeGENE HP DNA Transfection reagent). Cells were then lysed and subjected to immunoprecipitation using anti-Ncor1 (a) or anti-Ncor2 (b) antibody. Input and bound proteins were analyzed by western blot. Actin was used as loading control. c, d, HA-DAD (Ncor1) (c), HA-DAD (Ncor2) (d) or Flag-HDAC3 was expressed in HEK293T cells individually. After 48 h, protein extracts from HA-DAD (Ncor1) (c) / HA-DAD (Ncor2) (d) and Flag-HDAC3 transfected cells were mixed at a ratio of 1:1 (Ncor1/Ncor2:HDAC3) in the presence or absence of NADPH and/or 20μM Ins(1,4,5,6)P4 (overnight) before immunoprecipitation using anti-IgG or anti-Flag antibody. e, f, HEK293T cells co-transfected with Flag-HDAC3 and HA-DAD(Ncor1) (e) or HA-DAD (Ncor2) (f) were treated with NADPH (NADPH were transfected into cells using X-tremeGENE HP DNA Transfection reagent) and/or Ins(1,4,5,6)P4 overnight as indicated. Cells were then lysed and subjected to immunoprecipitation using anti-Flag antibody. gi, The dissociation constant (KD) for immobilized mutant HDAC3 (L1, L6 or Mut-all) from NADPH or Ins(1,4,5,6)P4 were determined by surface plasmon resonance (SPR). Shown are real-time graphs of response units against NADPH or Ins(1,4,5,6)P4 concentrations. In af, western blots represent three independent experiments. The samples derived from the same experiment and the blots were processed in parallel.

Source data

Extended Data Fig. 7 The molecular docking result targeting HDAC3.

a, Electrostatic surface representation of the HDAC3-DAD complex onto which Ins(1,4,5,6)P4 docks. Ins(1,4,5,6)P4 molecules are shown in stick view either in green (the most energetically favored docking pose in the binding pocket) or in yellow (extracted from the crystal structure with PDB ID 4A69). b, The zoom-in structure of (a) showing details of the interaction between HDAC3-DAD complex and docked (green sticks) or native (yellow sticks) pose of Ins(1,4,5,6)P4. Hydrogen bonds and salt bridges are indicated by green or yellow dashed lines, respectively. HDAC3 (light blue) and DAD (magenta) are represented in cartoon view. c, The blind docking result of NADPH onto HDAC3. The pose in the binding interface is colored in green, other representative poses are colored in yellow. d, The blind docking result of Ins(1,4,5,6)P4 onto HDAC3. The pose in the binding interface is colored in green, other representative poses are colored in yellow. e, Clashes are found after adding DAD (magenta cartoon) into the docked structure of HDAC3-NADPH (left), while no clashes are observed when DAD is added into the docked structure of HDAC3-Ins(1,4,5,6)P4 (right). Residues from HDAC3-DAD binding interface are shown in sticks, clashed residues are highlighted by red circles. f, The 2D interaction diagrams are shown for the docking results of HDAC3-NADPH (left) and HDAC3-Ins(1,4,5,6)P4 (right). g, Statistics of binding energies for the docking results of NAD+, NADH, NADP+, NADPH and Ins(1,4,5,6)P4 targeting HDAC3. The objective docking was repeated for nine times, yielding 81 poses for each ligand.

Extended Data Fig. 8 The sidechain dynamics of Tyr 298 as revealed by MD simulations.

The dihedral angle (N-Cα-Cβ-Cγ) probability distributions of Tyr298 shown for MD simulations of HDAC3, HDAC3-Ins(1,4,5,6)P4, HDAC3-DAD-Ins(1,4,5,6)P4 and HDAC3-NADPH.

Extended Data Fig. 9 NADPH promotes adipocyte differentiation, and inhibition of oxidative PPP or folate metabolism reduces histone acetylation.

a, b, Oil Red O staining of 3T3-L1 cells transfected with control, different sets of ME1 (a) or ME2 siRNA (b) in the presence of increasing amounts of NADPH after 7 days of differentiation. Wells and higher magnification images are shown (left). Oil Red O staining was quantified (right). Protein expression is shown below. c, 3T3-L1 preadipocytes treated with or without 6-AN were induced to differentiate for 7 days. (left and middle) Differentiation-induced lipid droplet accumulation was visualized by Oil Red O staining and quantified. (right) Acid extracts were analyzed by western blot with antibodies against acetylated histones. Total H3 was used as loading control. d, 3T3-L1 preadipocytes transfected with control, G6PD or MTHFD2 siRNA were induced to differentiate for 7 days. Cellular NADPH levels were analyzed. eg, NADP+/NADPH ratios in differentiating 3T3-L1 adipocytes (e) U2OS cells (f) and HCT116 cells (g) treated with control, ME1, ME2, G6PD or MTHFD2 siRNA as indicated were determined respectively. The protein expression was determined by western blot. Actin was used as loading control. h, Expressions of acetylated histones in U2OS cells and HCT116 cells treated with control, G6PD or MTHFD2 siRNA as indicated. Actin and total H3 were used as loading controls. In a, b, c, d, e, f, g, n = 3 biological replicates; Data are mean ± s.d.. Statistical significance was determined by two-tailed unpaired t-test. NS, not significant. In a,b (right, below panel), c (right panel), eg (right panel), h, western blots represent three independent experiments. The samples derived from the same experiment and the blots were processed in parallel.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–3.

Reporting Summary

Supplementary Video

The MD simulation movie for the docked HDAC3–NADPH structure. The length of the MD is 500 ns. Colour scheme: grey for HDAC3; red for NADPH; yellow for residues H17, G21, K25, R265 and R301 of HDAC3.

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Unprocessed western blots.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 9

Unprocessed western blots.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, W., Kou, J., Qin, J. et al. NADPH levels affect cellular epigenetic state by inhibiting HDAC3–Ncor complex. Nat Metab 3, 75–89 (2021). https://doi.org/10.1038/s42255-020-00330-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-00330-2

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing