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:

Activation of intestinal hypoxia-inducible factor 2α during obesity contributes to hepatic steatosis

Abstract

Nonalcoholic fatty liver disease is becoming the most common chronic liver disease in Western countries, and limited therapeutic options are available. Here we uncovered a role for intestinal hypoxia-inducible factor (HIF) in hepatic steatosis. Human-intestine biopsies from individuals with or without obesity revealed that intestinal HIF-2α signaling was positively correlated with body-mass index and hepatic toxicity. The causality of this correlation was verified in mice with an intestine-specific disruption of Hif2a, in which high-fat-diet-induced hepatic steatosis and obesity were substantially lower as compared to control mice. PT2385, a HIF-2α-specific inhibitor, had preventive and therapeutic effects on metabolic disorders that were dependent on intestine HIF-2α. Intestine HIF-2α inhibition markedly reduced intestine and serum ceramide levels. Mechanistically, intestine HIF-2α regulates ceramide metabolism mainly from the salvage pathway, by positively regulating the expression of Neu3, the gene encoding neuraminidase 3. These results suggest that intestinal HIF-2α could be a viable target for hepatic steatosis therapy.

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: Increased HIF-2α signaling in human ileum biopsies is correlated with obesity.
Figure 2: Intestine-specific HIF-2α disruption ameliorates the development of hepatic steatosis.
Figure 3: Intestinal HIF-2α deficiency reduces ceramide synthesis in the small intestine.
Figure 4: The ceramide-synthesis-related gene Neu3 is a novel HIF-2α target gene in the small intestine.
Figure 5: Administration of ceramide reverses the protective effects of intestinal HIF-2α inhibition on the development of HFD-induced hepatic steatosis.
Figure 6: PT2385 reverses HFD-induced hepatic steatosis.

Similar content being viewed by others

References

  1. Ray, K. NAFLD-the next global epidemic. Nat. Rev. Gastroenterol. Hepatol. 10, 621 (2013).

    Article  PubMed  Google Scholar 

  2. Chalasani, N. et al. The diagnosis and management of non-alcoholic fatty liver disease: practice guideline by the American Gastroenterological Association, American Association for the Study of Liver Diseases, and American College of Gastroenterology. Gastroenterology 142, 1592–1609 (2012).

    Article  PubMed  Google Scholar 

  3. Rotman, Y. & Sanyal, A.J. Current and upcoming pharmacotherapy for non-alcoholic fatty liver disease. Gut 66, 180–190 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Ju, C., Colgan, S.P. & Eltzschig, H.K. Hypoxia-inducible factors as molecular targets for liver diseases. J. Mol. Med. (Berl.) 94, 613–627 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ivan, M. et al. HIFα targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 292, 464–468 (2001).

    Article  CAS  PubMed  Google Scholar 

  7. Jaakkola, P. et al. Targeting of HIF-α to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 292, 468–472 (2001).

    Article  CAS  PubMed  Google Scholar 

  8. Minamishima, Y.A. et al. A feedback loop involving the Phd3 prolyl hydroxylase tunes the mammalian hypoxic response in vivo. Mol. Cell. Biol. 29, 5729–5741 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Qu, A. et al. Hypoxia-inducible transcription factor 2α promotes steatohepatitis through augmenting lipid accumulation, inflammation, and fibrosis. Hepatology 54, 472–483 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Ramakrishnan, S.K. et al. HIF2α is an essential molecular brake for postprandial hepatic glucagon response independent of insulin signaling. Cell Metab. 23, 505–516 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Taniguchi, C.M. et al. Cross-talk between hypoxia and insulin signaling through Phd3 regulates hepatic glucose and lipid metabolism and ameliorates diabetes. Nat. Med. 19, 1325–1330 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wei, K. et al. A liver Hif-2α-Irs2 pathway sensitizes hepatic insulin signaling and is modulated by Vegf inhibition. Nat. Med. 19, 1331–1337 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gonzalez, F.J., Jiang, C. & Patterson, A.D. An intestinal microbiota-farnesoid X receptor axis modulates metabolic disease. Gastroenterology 151, 845–859 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Singh, V. et al. Microbiota-dependent hepatic lipogenesis mediated by stearoyl CoA desaturase 1 (SCD1) promotes metabolic syndrome in TLR5-deficient mice. Cell Metab. 22, 983–996 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Jiang, C. et al. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J. Clin. Invest. 125, 386–402 (2015).

    Article  PubMed  Google Scholar 

  16. Safran, M. et al. Mouse model for noninvasive imaging of HIF prolyl hydroxylase activity: assessment of an oral agent that stimulates erythropoietin production. Proc. Natl. Acad. Sci. USA 103, 105–110 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Pagadala, M., Kasumov, T., McCullough, A.J., Zein, N.N. & Kirwan, J.P. Role of ceramides in nonalcoholic fatty liver disease. Trends Endocrinol. Metab. 23, 365–371 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kitatani, K., Idkowiak-Baldys, J. & Hannun, Y.A. The sphingolipid salvage pathway in ceramide metabolism and signaling. Cell. Signal. 20, 1010–1018 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Jornayvaz, F.R. & Shulman, G.I. Diacylglycerol activation of protein kinase Cɛ and hepatic insulin resistance. Cell Metab. 15, 574–584 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Xue, X. et al. Endothelial PAS domain protein 1 activates the inflammatory response in the intestinal epithelium to promote colitis in mice. Gastroenterology 145, 831–841 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Wallace, E.M. et al. A small-molecule antagonist of HIF2α is efficacious in preclinical models of renal cell carcinoma. Cancer Res. 76, 5491–5500 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Zou, Y., Albohy, A., Sandbhor, M. & Cairo, C.W. Inhibition of human neuraminidase 3 (NEU3) by C9-triazole derivatives of 2,3-didehydro-N-acetyl-neuraminic acid. Bioorg. Med. Chem. Lett. 20, 7529–7533 (2010).

    Article  CAS  PubMed  Google Scholar 

  23. Yoshinaga, A. et al. NEU3 inhibitory effect of naringin suppresses cancer cell growth by attenuation of EGFR signaling through GM3 ganglioside accumulation. Eur. J. Pharmacol. 782, 21–29 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. Triner, D. & Shah, Y.M. Hypoxia-inducible factors: a central link between inflammation and cancer. J. Clin. Invest. 126, 3689–3698 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Kelly, C.J. et al. Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF augments tissue barrier function. Cell Host Microbe 17, 662–671 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Rivera-Chávez, F. et al. Depletion of butyrate-producing clostridia from the gut microbiota drives an aerobic luminal expansion of salmonella. Cell Host Microbe 19, 443–454 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Hubbard, T.D., Murray, I.A. & Perdew, G.H. Indole and tryptophan metabolism: endogenous and dietary routes to Ah receptor activation. Drug Metab. Dispos. 43, 1522–1535 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Kawano, Y. et al. Colonic pro-inflammatory macrophages cause insulin resistance in an intestinal Ccl2/Ccr2-dependent manner. Cell Metab. 24, 295–310 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Rahman, K. et al. Loss of junctional adhesion molecule A promotes severe steatohepatitis in mice on a diet high in saturated fat, fructose, and cholesterol. Gastroenterology 151, 733–746 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Ramakrishnan, S.K. & Shah, Y.M. Role of intestinal HIF-2α in health and disease. Annu. Rev. Physiol. 78, 301–325 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Glover, L.E., Lee, J.S. & Colgan, S.P. Oxygen metabolism and barrier regulation in the intestinal mucosa. J. Clin. Invest. 126, 3680–3688 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Furuta, G.T. et al. Hypoxia-inducible factor 1-dependent induction of intestinal trefoil factor protects barrier function during hypoxia. J. Exp. Med. 193, 1027–1034 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Synnestvedt, K. et al. Ecto-5′-nucleotidase (CD73) regulation by hypoxia-inducible factor-1 mediates permeability changes in intestinal epithelia. J. Clin. Invest. 110, 993–1002 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Robinson, A. et al. Mucosal protection by hypoxia-inducible factor prolyl hydroxylase inhibition. Gastroenterology 134, 145–155 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Karhausen, J. et al. Epithelial hypoxia-inducible factor-1 is protective in murine experimental colitis. J. Clin. Invest. 114, 1098–1106 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Glover, L.E. et al. Control of creatine metabolism by HIF is an endogenous mechanism of barrier regulation in colitis. Proc. Natl. Acad. Sci. USA 110, 19820–19825 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Xie, L. et al. Hypoxia-inducible factor/MAZ-dependent induction of caveolin-1 regulates colon permeability through suppression of occludin, leading to hypoxia-induced inflammation. Mol. Cell. Biol. 34, 3013–3023 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Cummins, E.P. et al. The hydroxylase inhibitor dimethyloxalylglycine is protective in a murine model of colitis. Gastroenterology 134, 156–165 (2008).

    Article  CAS  PubMed  Google Scholar 

  39. Shah, Y.M. et al. Hypoxia-inducible factor augments experimental colitis through an MIF-dependent inflammatory signaling cascade. Gastroenterology 134, 2036–2048.e3 (2008).

    Article  PubMed  Google Scholar 

  40. Summers, S.A. & Goodpaster, B.H. CrossTalk proposal: Intramyocellular ceramide accumulation does modulate insulin resistance. J. Physiol. (Lond.) 594, 3167–3170 (2016).

    Article  CAS  Google Scholar 

  41. Chavez, J.A. & Summers, S.A. A ceramide-centric view of insulin resistance. Cell Metab. 15, 585–594 (2012).

    Article  CAS  PubMed  Google Scholar 

  42. Gorden, D.L. et al. Biomarkers of NAFLD progression: a lipidomics approach to an epidemic. J. Lipid Res. 56, 722–736 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Haus, J.M. et al. Plasma ceramides are elevated in obese subjects with type 2 diabetes and correlate with the severity of insulin resistance. Diabetes 58, 337–343 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chaurasia, B. & Summers, S.A. Ceramides—lipotoxic inducers of metabolic disorders. Trends Endocrinol. Metab. 26, 538–550 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Xia, J.Y. et al. Targeted induction of ceramide degradation leads to improved systemic metabolism and reduced hepatic steatosis. Cell Metab. 22, 266–278 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Turpin, S.M. et al. Obesity-induced CerS6-dependent C16:0 ceramide production promotes weight gain and glucose intolerance. Cell Metab. 20, 678–686 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Raichur, S. et al. CerS2 haploinsufficiency inhibits β-oxidation and confers susceptibility to diet-induced steatohepatitis and insulin resistance. Cell Metab. 20, 687–695 (2014).

    Article  CAS  PubMed  Google Scholar 

  48. Holland, W.L. et al. Inhibition of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-, and obesity-induced insulin resistance. Cell Metab. 5, 167–179 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Chaurasia, B. et al. Adipocyte ceramides regulate subcutaneous adipose browning, inflammation, and metabolism. Cell Metab. 24, 820–834 (2016).

    Article  CAS  PubMed  Google Scholar 

  50. Jiang, C. et al. Intestine-selective farnesoid X receptor inhibition improves obesity-related metabolic dysfunction. Nat. Commun. 6, 10166 (2015).

    Article  CAS  PubMed  Google Scholar 

  51. Perry, R.J., Samuel, V.T., Petersen, K.F. & Shulman, G.I. The role of hepatic lipids in hepatic insulin resistance and type 2 diabetes. Nature 510, 84–91 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Samuel, V.T. & Shulman, G.I. Mechanisms for insulin resistance: common threads and missing links. Cell 148, 852–871 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Giussani, P., Tringali, C., Riboni, L., Viani, P. & Venerando, B. Sphingolipids: key regulators of apoptosis and pivotal players in cancer drug resistance. Int. J. Mol. Sci. 15, 4356–4392 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Yoshizumi, S. et al. Increased hepatic expression of ganglioside-specific sialidase, NEU3, improves insulin sensitivity and glucose tolerance in mice. Metabolism 56, 420–429 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Scaringi, R. et al. NEU3 sialidase is activated under hypoxia and protects skeletal muscle cells from apoptosis through the activation of the epidermal growth factor receptor signaling pathway and the hypoxia-inducible factor (HIF)-1α. J. Biol. Chem. 288, 3153–3162 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Cho, H. et al. On-target efficacy of a HIF2α antagonist in preclinical kidney cancer models. Nature 539, 107–111 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Chen, W. et al. Targeting renal cell carcinoma with a HIF-2 antagonist. Nature 539, 112–117 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Haase, V.H., Glickman, J.N., Socolovsky, M. & Jaenisch, R. Vascular tumors in livers with targeted inactivation of the von Hippel-Lindau tumor suppressor. Proc. Natl. Acad. Sci. USA 98, 1583–1588 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Anderson, E.R. et al. The hypoxia-inducible factor-C/EBPα axis controls ethanol-mediated hepcidin repression. Mol. Cell. Biol. 32, 4068–4077 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Hu, C.J., Sataur, A., Wang, L., Chen, H. & Simon, M.C. The N-terminal transactivation domain confers target gene specificity of hypoxia-inducible factors HIF-1α and HIF-2α. Mol. Biol. Cell 18, 4528–4542 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank L.G. Byrd, Y. Zhang, X. Gao, W. Liu, X. Gong, and T. Yan (National Cancer Institute) for assistance with the mouse studies, and B. Liu and X. Wu (Chinese Academy of Sciences) for help with the immunohistochemistry. This project was funded in part by the National Cancer Institute Intramural Research Program to F.J.G., grants from the National Key Research and Development Program of China (2016YFC0903100, 2016YFC0903102) to Changtao Jiang, the National Natural Science Foundation of China (31401011 and 81522007 to CT. J., and 81403007 to X.C.), National Institutes of Health grants ES022186 to A.D.P., and CA148828 and DK095201 to Y.M.S. S.K.R. was supported by NIDDK (K99DK110537). Q.W. was supported by the Peak Talent Foundation of Jiangsu Province Hospital of Chinese Medicine (Y2014RC18) and Jiangsu Government Scholarship for Overseas Studies. D.S. and J.Z. were supported by fellowships from the Chinese Scholarship Council.

Author information

Authors and Affiliations

Authors

Contributions

C.X., T.Y., Y.L., X.L., T.C., Q.W., D.S., J.Z., S.K.R., L.S., Chunmei Jiang, X.X., Y.T., and K.W.K. performed the research and analyzed the data. C.X., A.D.P., Y.M.S., Y.W., Changtao Jiang and F.J.G. designed and supervised the research. C.X., Changtao Jiang and F.J.G. wrote the manuscript.

Corresponding authors

Correspondence to Yue Wu, Changtao Jiang or Frank J Gonzalez.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Figures and Tables

Supplementary Figures 1–12 and Supplementary Tables 1–3 (PDF 5390 kb)

Life Sciences Reporting Summary (PDF 130 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xie, C., Yagai, T., Luo, Y. et al. Activation of intestinal hypoxia-inducible factor 2α during obesity contributes to hepatic steatosis. Nat Med 23, 1298–1308 (2017). https://doi.org/10.1038/nm.4412

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4412

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research