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:

Hepatic Hdac3 promotes gluconeogenesis by repressing lipid synthesis and sequestration

Abstract

Fatty liver disease is associated with obesity and type 2 diabetes, and hepatic lipid accumulation may contribute to insulin resistance. Histone deacetylase 3 (Hdac3) controls the circadian rhythm of hepatic lipogenesis. Here we show that, despite severe hepatosteatosis, mice with liver-specific depletion of Hdac3 have higher insulin sensitivity without any changes in insulin signaling or body weight compared to wild-type mice. Hdac3 depletion reroutes metabolic precursors towards lipid synthesis and storage within lipid droplets and away from hepatic glucose production. Perilipin 2, which coats lipid droplets, is markedly induced upon Hdac3 depletion and contributes to the development of both steatosis and improved tolerance to glucose. These findings suggest that the sequestration of hepatic lipids in perilipin 2–coated droplets ameliorates insulin resistance and establish Hdac3 as a pivotal epigenomic modifier that integrates signals from the circadian clock in the regulation of hepatic intermediary metabolism.

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

Figure 1: Liver-specific depletion of Hdac3 in adult mice causes quick-onset hepatosteatosis that is distinct from that caused by a HFD.
Figure 2: The Hdac3-depleted liver has impaired glucose production and is insulin hypersensitive.
Figure 3: Molecular insulin signaling events are unchanged despite lipid accumulation in the Hdac3-depleted liver.
Figure 4: Loss of Hdac3 reroutes precursors toward lipid synthesis and sequestration within lipid droplets.
Figure 5: Reduced hepatic glucose production in the Hdac3-depleted liver is a result of metabolic rerouting rather than inherent defects of gluconeogenesis.
Figure 6: Lipid sequestration is required for the development of severe hepatosteatosis and improved glucose tolerance in mice without hepatic Hdac3.

Similar content being viewed by others

References

  1. Cohen, J.C., Horton, J.D. & Hobbs, H.H. Human fatty liver disease: old questions and new insights. Science 332, 1519–1523 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Browning, J.D. & Horton, J.D. Molecular mediators of hepatic steatosis and liver injury. J. Clin. Invest. 114, 147–152 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Utzschneider, K.M. & Kahn, S.E. Review: the role of insulin resistance in nonalcoholic fatty liver disease. J. Clin. Endocrinol. Metab. 91, 4753–4761 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Nagle, C.A., Klett, E.L. & Coleman, R.A. Hepatic triacylglycerol accumulation and insulin resistance. J. Lipid Res. 50 (suppl.), S74–S79 (2009).

    Article  PubMed  Google Scholar 

  5. Postic, C. & Girard, J. Contribution of de novo fatty acid synthesis to hepatic steatosis and insulin resistance: lessons from genetically engineered mice. J. Clin. Invest. 118, 829–838 (2008).

    Article  CAS  PubMed  Google Scholar 

  6. Samuel, V.T., Petersen, K.F. & Shulman, G.I. Lipid-induced insulin resistance: unravelling the mechanism. Lancet 375, 2267–2277 (2010).

    Article  CAS  PubMed  Google Scholar 

  7. Lonardo, A., Bellentani, S., Ratziu, V. & Loria, P. Insulin resistance in nonalcoholic steatohepatitis: necessary but not sufficient—death of a dogma from analysis of therapeutic studies? Expert Rev. Gastroenterol. Hepatol. 5, 279–289 (2011).

    Article  PubMed  Google Scholar 

  8. Lass, A., Zimmermann, R., Oberer, M. & Zechner, R. Lipolysis—a highly regulated multi-enzyme complex mediates the catabolism of cellular fat stores. Prog. Lipid Res. 50, 14–27 (2011).

    Article  CAS  PubMed  Google Scholar 

  9. Donnelly, K.L. et al. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J. Clin. Invest. 115, 1343–1351 (2005).

    Article  CAS  PubMed  Google Scholar 

  10. Larter, C.Z. & Yeh, M.M. Animal models of NASH: getting both pathology and metabolic context right. J. Gastroenterol. Hepatol. 23, 1635–1648 (2008).

    Article  PubMed  Google Scholar 

  11. Biddinger, S.B. & Kahn, C.R. From mice to men: insights into the insulin resistance syndromes. Annu. Rev. Physiol. 68, 123–158 (2006).

    Article  CAS  PubMed  Google Scholar 

  12. Taniguchi, C.M. et al. Divergent regulation of hepatic glucose and lipid metabolism by phosphoinositide 3-kinase via Akt and PKCλ/ζ. Cell Metab. 3, 343–353 (2006).

    Article  CAS  PubMed  Google Scholar 

  13. Leavens, K.F., Easton, R.M., Shulman, G.I., Previs, S.F. & Birnbaum, M.J. Akt2 is required for hepatic lipid accumulation in models of insulin resistance. Cell Metab. 10, 405–418 (2009).

    Article  CAS  PubMed  Google Scholar 

  14. Brown, M.S. & Goldstein, J.L. Selective versus total insulin resistance: a pathogenic paradox. Cell Metab. 7, 95–96 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. Stiles, B. et al. Liver-specific deletion of negative regulator Pten results in fatty liver and insulin hypersensitivity. Proc. Natl. Acad. Sci. USA 101, 2082–2087 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Greenberg, A.S. et al. The role of lipid droplets in metabolic disease in rodents and humans. J. Clin. Invest. 121, 2102–2110 (2011).

    Article  CAS  PubMed  Google Scholar 

  17. Matsuzaka, T. et al. Crucial role of a long-chain fatty acid elongase, Elovl6, in obesity-induced insulin resistance. Nat. Med. 13, 1193–1202 (2007).

    Article  CAS  PubMed  Google Scholar 

  18. Brown, J.M. et al. CGI-58 knockdown in mice causes hepatic steatosis but prevents diet-induced obesity and glucose intolerance. J. Lipid Res. 51, 3306–3315 (2010).

    Article  CAS  PubMed  Google Scholar 

  19. Tuomilehto, J. et al. Prevention of type 2 diabetes mellitus by changes in lifestyle among subjects with impaired glucose tolerance. N. Engl. J. Med. 344, 1343–1350 (2001).

    Article  CAS  PubMed  Google Scholar 

  20. Bass, J. & Takahashi, J.S. Circadian integration of metabolism and energetics. Science 330, 1349–1354 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Desvergne, B., Michalik, L. & Wahli, W. Transcriptional regulation of metabolism. Physiol. Rev. 86, 465–514 (2006).

    Article  CAS  PubMed  Google Scholar 

  22. Feng, D. et al. A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism. Science 331, 1315–1319 (2011).

    Article  CAS  PubMed  Google Scholar 

  23. Panda, S. et al. Coordinated transcription of key pathways in the mouse by the circadian clock. Cell 109, 307–320 (2002).

    Article  CAS  PubMed  Google Scholar 

  24. Lawless, M.W., Norris, S., O'Byrne, K.J. & Gray, S.G. Targeting histone deacetylases for the treatment of immune, endocrine & metabolic disorders. Endocr. Metab. Immune Disord. Drug Targets 9, 84–107 (2009).

    Article  CAS  PubMed  Google Scholar 

  25. Sun, Z., Feng, D., Everett, L.J., Bugge, A. & Lazar, M.A. Circadian epigenomic remodeling and hepatic lipogenesis: lessons from HDAC3. Cold Spring Harb. Symp. Quant. Biol. published online, doi:10.1101/sqb.2011.76.011494 (6 September 2011).

  26. Knutson, S.K. et al. Liver-specific deletion of histone deacetylase 3 disrupts metabolic transcriptional networks. EMBO J. 27, 1017–1028 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Mullican, S.E. et al. Histone deacetylase 3 is an epigenomic brake in macrophage alternative activation. Genes Dev. 25, 2480–2488 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Bell, P. et al. Evaluation of adeno-associated viral vectors for liver-directed gene transfer in dogs. Hum. Gene Ther. 22, 985–997 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Samuel, V.T. et al. Inhibition of protein kinase Cɛ prevents hepatic insulin resistance in nonalcoholic fatty liver disease. J. Clin. Invest. 117, 739–745 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Calvo, M.N. et al. PFKFB3 gene silencing decreases glycolysis, induces cell-cycle delay and inhibits anchorage-independent growth in HeLa cells. FEBS Lett. 580, 3308–3314 (2006).

    Article  CAS  PubMed  Google Scholar 

  31. Bolaños, J.P., Almeida, A. & Moncada, S. Glycolysis: a bioenergetic or a survival pathway? Trends Biochem. Sci. 35, 145–149 (2010).

    Article  PubMed  Google Scholar 

  32. Seo, Y.-K. et al. Genome-wide analysis of SREBP-1 binding in mouse liver chromatin reveals a preference for promoter proximal binding to a new motif. Proc. Natl. Acad. Sci. USA 106, 13765–13769 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Seo, Y.-K. et al. Genome-wide localization of SREBP-2 in hepatic chromatin predicts a role in autophagy. Cell Metab. 13, 367–375 (2011).

    Article  CAS  PubMed  Google Scholar 

  34. Wendel, A.A., Lewin, T.M. & Coleman, R.A. Glycerol-3-phosphate acyltransferases: rate limiting enzymes of triacylglycerol biosynthesis. Biochim. Biophys. Acta 1791, 501–506 (2009).

    Article  CAS  PubMed  Google Scholar 

  35. Brasaemle, D.L. Thematic review series: adipocyte biology. The perilipin family of structural lipid droplet proteins: stabilization of lipid droplets and control of lipolysis. J. Lipid Res. 48, 2547–2559 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Gong, J., Sun, Z. & Li, P. CIDE proteins and metabolic disorders. Curr. Opin. Lipidol. 20, 121–126 (2009).

    Article  CAS  PubMed  Google Scholar 

  37. Puri, V. et al. Fat-specific protein 27, a novel lipid droplet protein that enhances triglyceride storage. J. Biol. Chem. 282, 34213–34218 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Samuel, V.T. et al. Mechanism of hepatic insulin resistance in non-alcoholic fatty liver disease. J. Biol. Chem. 279, 32345–32353 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Hammond, L.E. et al. Mitochondrial glycerol-3-phosphate acyltransferase-1 is essential in liver for the metabolism of excess acyl-CoAs. J. Biol. Chem. 280, 25629–25636 (2005).

    Article  CAS  PubMed  Google Scholar 

  40. Nilsson, A., Thomassen, M.S. & Christiansen, E. Long-chain Acyl-CoA levels in liver from rats fed high-fat diets: is it of significance for an increased peroxisomal β-oxidation? Lipids 19, 187–194 (1984).

    Article  CAS  PubMed  Google Scholar 

  41. Mihaylova, M.M. et al. Class IIa histone deacetylases are hormone-activated regulators of FOXO and mammalian glucose homeostasis. Cell 145, 607–621 (2011).

    Article  CAS  PubMed  Google Scholar 

  42. Chang, B.H.-J. et al. Protection against fatty liver but normal adipogenesis in mice lacking adipose differentiation-related protein. Mol. Cell. Biol. 26, 1063–1076 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Imai, Y. et al. Reduction of hepatosteatosis and lipid levels by an adipose differentiation-related protein antisense oligonucleotide. Gastroenterology 132, 1947–1954 (2007).

    Article  CAS  PubMed  Google Scholar 

  44. Mutel, E. et al. Control of blood glucose in the absence of hepatic glucose production during prolonged fasting in mice: induction of renal and intestinal gluconeogenesis by glucagon. Diabetes 60, 3121–3131 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Vogt, P.K., Jiang, H. & Aoki, M. Triple layer control: phosphorylation, acetylation and ubiquitination of FOXO proteins. Cell Cycle 4, 908–913 (2005).

    Article  CAS  PubMed  Google Scholar 

  46. Zhang, W. et al. FoxO1 regulates multiple metabolic pathways in the liver: effects on gluconeogenic, glycolytic, and lipogenic gene expression. J. Biol. Chem. 281, 10105–10117 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Banks, A.S. et al. Dissociation of the glucose and lipid regulatory functions of FoxO1 by targeted knockin of acetylation-defective alleles in mice. Cell Metab. 14, 587–597 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Haeusler, R.A., Han, S. & Accili, D. Hepatic FoxO1 ablation exacerbates lipid abnormalities during hyperglycemia. J. Biol. Chem. 285, 26861–26868 (2010).

    Article  CAS  PubMed  Google Scholar 

  49. Tao, R. et al. Hepatic FoxOs regulate lipid metabolism via modulation of expression of the nicotinamide phosphoribosyltransferase gene. J. Biol. Chem. 286, 14681–14690 (2011).

    Article  CAS  PubMed  Google Scholar 

  50. Kadereit, B. et al. Evolutionarily conserved gene family important for fat storage. Proc. Natl. Acad. Sci. USA 105, 94–99 (2008).

    Article  CAS  PubMed  Google Scholar 

  51. Yang, X. et al. The G(0)/G(1) switch gene 2 regulates adipose lipolysis through association with adipose triglyceride lipase. Cell Metab. 11, 194–205 (2010).

    Article  CAS  PubMed  Google Scholar 

  52. Bell, M. et al. Consequences of lipid droplet coat protein downregulation in liver cells: abnormal lipid droplet metabolism and induction of insulin resistance. Diabetes 57, 2037–2045 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Kantartzis, K. et al. Dissociation between fatty liver and insulin resistance in humans carrying a variant of the patatin-like phospholipase 3 gene. Diabetes 58, 2616–2623 (2009).

    Article  CAS  PubMed  Google Scholar 

  54. Minehira, K. et al. Blocking VLDL secretion causes hepatic steatosis but does not affect peripheral lipid stores or insulin sensitivity in mice. J. Lipid Res. 49, 2038–2044 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Schonfeld, G., Yue, P., Lin, X. & Chen, Z. Fatty liver and insulin resistance: not always linked. Trans. Am. Clin. Climatol. Assoc. 119, 217–223, discussion 223–224 (2008).

    PubMed Central  PubMed  Google Scholar 

  56. Chang, B.H.-J., Li, L., Saha, P. & Chan, L. Absence of adipose differentiation related protein upregulates hepatic VLDL secretion, relieves hepatosteatosis, and improves whole body insulin resistance in leptin-deficient mice. J. Lipid Res. 51, 2132–2142 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Gerstein, H.C. et al. Effects of intensive glucose lowering in type 2 diabetes. N. Engl. J. Med. 358, 2545–2559 (2008).

    Article  CAS  PubMed  Google Scholar 

  58. Sapiro, J.M., Mashek, M.T., Greenberg, A.S. & Mashek, D.G. Hepatic triacylglycerol hydrolysis regulates peroxisome proliferator-activated receptor α activity. J. Lipid Res. 50, 1621–1629 (2009).

    Article  CAS  PubMed  Google Scholar 

  59. Kumashiro, N. et al. Cellular mechanism of insulin resistance in nonalcoholic fatty liver disease. Proc. Natl. Acad. Sci. USA 108, 16381–16385 (2011).

    Article  CAS  PubMed  Google Scholar 

  60. Sztalryd, C. et al. Functional compensation for adipose differentiation-related protein (ADFP) by Tip47 in an ADFP null embryonic cell line. J. Biol. Chem. 281, 34341–34348 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank J. Millar and the Metabolic Tracer Resource at the Penn Institute for Diabetes, Obesity and Metabolism for the lipogenesis flux assay. We thank the Mouse Metabolic Phenotyping Core and the Viral Vector Core at Penn Diabetes Research Center (P30 DK19525) and the Penn Digestives Disease Center Morphology Core (P30 DK050306) at the University of Pennsylvania. We thank the Vanderbilt (DK59637) and Yale (U24 DK059635) Mouse Metabolic Phenotyping Centers and the Children's Hospital of Philadelphia Pathology Core. We thank B. Agarwal and S. Mullican for helpful discussion. This work was supported by US National Institutes of Health grants R37 DK43806 (M.A.L.), P01 DK49210 (M.A.L., M.J. Birnbaum and R.S.A.) and R01 DK40936 (G.I.S.), R01 DK075017 (C.S.), the Department of Veteran Affairs Merit Review Program (T.G.U.), grant F32 DK079572 (R.A.M.), the JPB Foundation and the Cox Institute for Medical Research.

Author information

Authors and Affiliations

Authors

Contributions

Z.S. and M.A.L. conceived of the hypothesis and designed the experiments. Z.S., R.A.M., R.T.P., J.C., R.D., H.W. and D.Z. performed the experiments. Z.S., R.A.M., R.D., T.G.U., G.I.S., C.S., M.J. Bennett, R.S.A., M.J. Birnbaum and M.A.L. analyzed and interpreted the data. M.J.G. provided reagents. Z.S. and M.A.L. wrote the manuscript.

Corresponding author

Correspondence to Mitchell A Lazar.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 742 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sun, Z., Miller, R., Patel, R. et al. Hepatic Hdac3 promotes gluconeogenesis by repressing lipid synthesis and sequestration. Nat Med 18, 934–942 (2012). https://doi.org/10.1038/nm.2744

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2744

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