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:

Inhibition of Notch uncouples Akt activation from hepatic lipid accumulation by decreasing mTorc1 stability

Abstract

Increased hepatic lipid content is an early correlate of insulin resistance and can be caused by nutrient-induced activation of mammalian target of rapamycin (mTor). This activation of mTor increases basal Akt activity, leading to a self-perpetuating lipogenic cycle. We have previously shown that the developmental Notch pathway has metabolic functions in adult mouse liver. Acute or chronic inhibition of Notch dampens hepatic glucose production and increases Akt activity and may therefore be predicted to increase hepatic lipid content. Here we now show that constitutive liver-specific ablation of Notch signaling, or its acute inhibition with a decoy Notch1 receptor, prevents hepatosteatosis by blocking mTor complex 1 (mTorc1) activity. Conversely, Notch gain of function causes fatty liver through constitutive activation of mTorc1, an effect that is reversible by treatment with rapamycin. We demonstrate that Notch signaling increases mTorc1 complex stability, augmenting mTorc1 function and sterol regulatory element binding transcription factor 1c (Srebp1c)-mediated lipogenesis. These data identify Notch as a therapeutically actionable branch point of metabolic signaling at which Akt activation in the liver can be uncoupled from hepatosteatosis.

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: Regulation of hepatic Notch activity.
Figure 2: Lower hepatic triglyceride concentrations in HFD-fed L-Rbpj mice.
Figure 3: Notch1 decoy increases insulin sensitivity and decreases hepatic lipid content.
Figure 4: Activation of hepatic Notch increases mTorc1 activity, lipogenic gene expression and steatosis in chow-fed mice.
Figure 5: mTor inhibition prevents Notch-induced fatty liver.
Figure 6: Notch induces mTorc1 complex stability.

Similar content being viewed by others

References

  1. Wang, Y.C., McPherson, K., Marsh, T., Gortmaker, S.L. & Brown, M. Health and economic burden of the projected obesity trends in the USA and the UK. Lancet 378, 815–825 (2011).

    Article  PubMed  Google Scholar 

  2. Hay, N. & Sonenberg, N. Upstream and downstream of mTOR. Genes Dev. 18, 1926–1945 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Li, S., Brown, M.S. & Goldstein, J.L. Bifurcation of insulin signaling pathway in rat liver: mTORC1 required for stimulation of lipogenesis, but not inhibition of gluconeogenesis. Proc. Natl. Acad. Sci. USA 107, 3441–3446 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sabatini, D.M. mTOR and cancer: insights into a complex relationship. Nat. Rev. Cancer 6, 729–734 (2006).

    Article  CAS  PubMed  Google Scholar 

  5. Peterson, T.R. et al. mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway. Cell 146, 408–420 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Matsumoto, M., Pocai, A., Rossetti, L., Depinho, R.A. & Accili, D. Impaired regulation of hepatic glucose production in mice lacking the forkhead transcription factor Foxo1 in liver. Cell Metab. 6, 208–216 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Matsumoto, M., Han, S., Kitamura, T. & Accili, D. Dual role of transcription factor Foxo1 in controlling hepatic insulin sensitivity and lipid metabolism. J. Clin. Invest. 116, 2464–2472 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Savage, D.B. & Semple, R.K. Recent insights into fatty liver, metabolic dyslipidaemia and their links to insulin resistance. Curr. Opin. Lipidol. 21, 329–336 (2010).

    Article  CAS  PubMed  Google Scholar 

  9. Bolós, V., Grego-Bessa, J. & de la Pompa, J.L. Notch signaling in development and cancer. Endocr. Rev. 28, 339–363 (2007).

    Article  PubMed  CAS  Google Scholar 

  10. Fortini, M.E. Notch signaling: the core pathway and its posttranslational regulation. Dev. Cell 16, 633–647 (2009).

    Article  CAS  PubMed  Google Scholar 

  11. Dufraine, J., Funahashi, Y. & Kitajewski, J. Notch signaling regulates tumor angiogenesis by diverse mechanisms. Oncogene 27, 5132–5137 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Swiatek, P.J., Lindsell, C.E., del Amo, F.F., Weinmaster, G. & Gridley, T. Notch1 is essential for postimplantation development in mice. Genes Dev. 8, 707–719 (1994).

    Article  CAS  PubMed  Google Scholar 

  13. Oka, C. et al. Disruption of the mouse RBP-Jκ gene results in early embryonic death. Development 121, 3291–3301 (1995).

    Article  CAS  PubMed  Google Scholar 

  14. Shen, J. et al. Skeletal and CNS defects in Presenilin-1–deficient mice. Cell 89, 629–639 (1997).

    Article  CAS  PubMed  Google Scholar 

  15. Rizzo, P. et al. Rational targeting of Notch signaling in cancer. Oncogene 27, 5124–5131 (2008).

    Article  CAS  PubMed  Google Scholar 

  16. Weinmaster, G. & Kopan, R. A garden of Notch-ly delights. Development 133, 3277–3282 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Pajvani, U.B. et al. Inhibition of Notch signaling ameliorates insulin resistance in a Foxo1-dependent manner. Nat. Med. 17, 961–967 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chan, S.M., Weng, A.P., Tibshirani, R., Aster, J.C. & Utz, P.J. Notch signals positively regulate activity of the mTOR pathway in T-cell acute lymphoblastic leukemia. Blood 110, 278–286 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Efferson, C.L. et al. Downregulation of Notch pathway by a γ-secretase inhibitor attenuates AKT/mammalian target of rapamycin signaling and glucose uptake in an ERBB2 transgenic breast cancer model. Cancer Res. 70, 2476–2484 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Postic, C. & Magnuson, M.A. DNA excision in liver by an albumin-Cre transgene occurs progressively with age. Genesis 26, 149–150 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Kitamura, T. et al. A Foxo/Notch pathway controls myogenic differentiation and fiber type specification. J. Clin. Invest. 117, 2477–2485 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Haeusler, R.A., Pratt-Hyatt, M., Welch, C.L., Klaassen, C.D. & Accili, D. Impaired generation of 12-hydroxylated bile acids links hepatic insulin signaling with dyslipidemia. Cell Metab. 15, 65–74 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. 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  PubMed Central  Google Scholar 

  24. 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  PubMed Central  Google Scholar 

  25. Kim, J.B., Wright, H.M., Wright, M. & Spiegelman, B.M. ADD1/SREBP1 activates PPARγ through the production of endogenous ligand. Proc. Natl. Acad. Sci. USA 95, 4333–4337 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhang, Y.L. et al. Aberrant hepatic expression of PPARγ2 stimulates hepatic lipogenesis in a mouse model of obesity, insulin resistance, dyslipidemia, and hepatic steatosis. J. Biol. Chem. 281, 37603–37615 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Gingras, A.C. et al. Regulation of 4E-BP1 phosphorylation: a novel two-step mechanism. Genes Dev. 13, 1422–1437 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Chiang, G.G. & Abraham, R.T. Phosphorylation of mammalian target of rapamycin (mTOR) at Ser-2448 is mediated by p70S6 kinase. J. Biol. Chem. 280, 25485–25490 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Weng, Q.P. et al. Regulation of the p70 S6 kinase by phosphorylation in vivo. Analysis using site-specific anti-phosphopeptide antibodies. J. Biol. Chem. 273, 16621–16629 (1998).

    Article  CAS  PubMed  Google Scholar 

  30. Funahashi, Y. et al. A notch1 ectodomain construct inhibits endothelial notch signaling, tumor growth, and angiogenesis. Cancer Res. 68, 4727–4735 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Funahashi, Y. et al. Notch modulates VEGF action in endothelial cells by inducing matrix metalloprotease activity. Vasc. Cell 3, 2 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Peterson, T.R. et al. DEPTOR is an mTOR inhibitor frequently overexpressed in multiple myeloma cells and required for their survival. Cell 137, 873–886 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Blättler, S.M. et al. Yin Yang 1 deficiency in skeletal muscle protects against rapamycin-induced diabetic-like symptoms through activation of insulin/IGF signaling. Cell Metab. 15, 505–517 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Kim, D.H. et al. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 110, 163–175 (2002).

    Article  CAS  PubMed  Google Scholar 

  35. Foster, K.G. et al. Regulation of mTOR complex 1 (mTORC1) by raptor Ser863 and multisite phosphorylation. J. Biol. Chem. 285, 80–94 (2010).

    Article  CAS  PubMed  Google Scholar 

  36. Kaizuka, T. et al. Tti1 and Tel2 are critical factors in mammalian target of rapamycin complex assembly. J. Biol. Chem. 285, 20109–20116 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu, H. et al. Wnt signaling regulates hepatic metabolism. Sci. Signal. 4, ra6 (2011).

    PubMed  PubMed Central  Google Scholar 

  38. Haeusler, R.A., Kaestner, K.H. & Accili, D. FoxOs function synergistically to promote glucose production. J. Biol. Chem. 285, 35245–35248 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Sun, Z. et al. Hepatic Hdac3 promotes gluconeogenesis by repressing lipid synthesis and sequestration. Nat. Med. 18, 934–942 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hagiwara, A. et al. Hepatic mTorc2 activates glycolysis and lipogenesis through Akt, glucokinase, and SREBP1c. Cell Metab. 15, 725–738 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Fukuda, D. et al. Notch ligand Delta-like 4 blockade attenuates atherosclerosis and metabolic disorders. Proc. Natl. Acad. Sci. USA 109, E1868–E1877 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kim-Muller, J.Y. & Accili, D. Cell biology. Selective insulin sensitizers. Science 331, 1529–1531 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Howell, J.J. & Manning, B.D. mTOR couples cellular nutrient sensing to organismal metabolic homeostasis. Trends Endocrinol. Metab. 22, 94–102 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Yecies, J.L. et al. Akt stimulates hepatic SREBP1c and lipogenesis through parallel mTORC1-dependent and independent pathways. Cell Metab. 14, 21–32 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Gwinn, D.M. et al. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol. Cell 30, 214–226 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Noguera-Troise, I. et al. Blockade of Dll4 inhibits tumour growth by promoting non-productive angiogenesis. Nature 444, 1032–1037 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Wu, Y. et al. Therapeutic antibody targeting of individual Notch receptors. Nature 464, 1052–1057 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. Houde, V.P. et al. Chronic rapamycin treatment causes glucose intolerance and hyperlipidemia by upregulating hepatic gluconeogenesis and impairing lipid deposition in adipose tissue. Diabetes 59, 1338–1348 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. van Es, J.H. et al. Notch/γ-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature 435, 959–963 (2005).

    Article  CAS  PubMed  Google Scholar 

  50. Fujikura, J. et al. Notch/Rbp-j signaling prevents premature endocrine and ductal cell differentiation in the pancreas. Cell Metab. 3, 59–65 (2006).

    Article  CAS  PubMed  Google Scholar 

  51. Paik, J.H. et al. FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis. Cell 128, 309–323 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Folch, J., Lees, M. & Sloane Stanley, G.H. A simple method for the isolation and purification of total lipides from animal tissues. J. Biol. Chem. 226, 497–509 (1957).

    Article  CAS  PubMed  Google Scholar 

  53. Millar, J.S., Cromley, D.A., McCoy, M.G., Rader, D.J. & Billheimer, J.T. Determining hepatic triglyceride production in mice: comparison of poloxamer 407 with Triton WR-1339. J. Lipid Res. 46, 2023–2028 (2005).

    Article  CAS  PubMed  Google Scholar 

  54. Li, G., Hernandez-Ono, A., Crooke, R.M., Graham, M.J. & Ginsberg, H.N. Effects of antisense-mediated inhibition of 11β-hydroxysteroid dehydrogenase type 1 on hepatic lipid metabolism. J. Lipid Res. 52, 971–981 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Nakae, J. et al. The forkhead transcription factor Foxo1 regulates adipocyte differentiation. Dev. Cell 4, 119–129 (2003).

    Article  CAS  PubMed  Google Scholar 

  56. Kim, J.B. et al. Nutritional and insulin regulation of fatty acid synthetase and leptin gene expression through ADD1/SREBP1. J. Clin. Invest. 101, 1–9 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Qiang, L. et al. Brown remodeling of white adipose tissue by SirT1-dependent deacetylation of Pparγ. Cell 150, 620–632 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by US National Institutes of Health grants DK093604 (U.B.P.), DK57539 (D.A.), HL062454 (J.K.) and DK63608 (Columbia Diabetes Research Center). We thank D. Conlon, C. Eng, I. Goldberg, R. Haeusler and I. Tabas, as well as members of the Accili, Kitajewski and Ginsberg laboratories, for insightful discussion of the data. We acknowledge excellent technical support from A. Flete, T. Kolar and J. Lee, as well as plasmids from D. Sabatini (Whitehead Institute) and B. Spiegelman (Dana-Farber Cancer Institute).

Author information

Authors and Affiliations

Authors

Contributions

U.B.P. designed and performed experiments, analyzed data and wrote the manuscript. L.Q. and T.K. designed and performed experiments and analyzed data. J.K., H.N.G. and D.A. designed the studies, analyzed the data and wrote the manuscript.

Corresponding authors

Correspondence to Utpal B Pajvani or Domenico Accili.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 925 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Pajvani, U., Qiang, L., Kangsamaksin, T. et al. Inhibition of Notch uncouples Akt activation from hepatic lipid accumulation by decreasing mTorc1 stability. Nat Med 19, 1054–1060 (2013). https://doi.org/10.1038/nm.3259

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3259

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