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:

The regulatory subunits of PI3K, p85α and p85β, interact with XBP-1 and increase its nuclear translocation

Abstract

Despite the fact that X-box binding protein-1 (XBP-1) is one of the main regulators of the unfolded protein response (UPR), the modulators of XBP-1 are poorly understood. Here, we show that the regulatory subunits of phosphotidyl inositol 3-kinase (PI3K), p85α (encoded by Pik3r1) and p85β (encoded by Pik3r2) form heterodimers that are disrupted by insulin treatment. This disruption of heterodimerization allows the resulting monomers of p85 to interact with, and increase the nuclear translocation of, the spliced form of XBP-1 (XBP-1s). The interaction between p85 and XBP-1s is lost in ob/ob mice, resulting in a severe defect in XBP-1s translocation to the nucleus and thus in the resolution of endoplasmic reticulum (ER) stress. These defects are ameliorated when p85α and p85β are overexpressed in the liver of ob/ob mice. Our results define a previously unknown insulin receptor signaling pathway and provide new mechanistic insight into the development of ER stress during obesity.

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: p85α and p85β interact with XBP-1s.
Figure 2: p85α and p85β increase nuclear translocation of XBP-1s.
Figure 3: Insulin increases nuclear transport of XBP-1s.
Figure 4: XBP-1s import to the nucleus is impaired in the ob/ob mice.
Figure 5: Overexpression of p85α and p85β in the liver of ob/ob mice increases glucose tolerance and establishes euglycemia.
Figure 6: Nuclear translocation of XBP-1s is impaired in the liver-specific Pik3r1−/−;Pik3r2−/− mice.

Similar content being viewed by others

References

  1. Marciniak, S.J. & Ron, D. Endoplasmic reticulum stress signaling in disease. Physiol. Rev. 86, 1133–1149 (2006).

    Article  CAS  Google Scholar 

  2. Schröder, M. & Kaufman, R.J. The mammalian unfolded protein response. Annu. Rev. Biochem. 74, 739–789 (2005).

    Article  Google Scholar 

  3. Bernales, S., Papa, F.R. & Walter, P. Intracellular signaling by the unfolded protein response. Annu. Rev. Cell Dev. Biol. 22, 487–508 (2006).

    Article  CAS  Google Scholar 

  4. Ron, D. & Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat. Rev. Mol. Cell Biol. 8, 519–529 (2007).

    Article  CAS  Google Scholar 

  5. Zhang, K. & Kaufman, R.J. From endoplasmic-reticulum stress to the inflammatory response. Nature 454, 455–462 (2008).

    Article  CAS  Google Scholar 

  6. Harding, H.P., Zhang, Y. & Ron, D. Protein translation and folding are coupled by an endoplasmic-reticulum–resident kinase. Nature 397, 271–274 (1999).

    Article  CAS  Google Scholar 

  7. Harding, H.P., Zhang, Y., Bertolotti, A., Zeng, H. & Ron, D. Perk is essential for translational regulation and cell survival during the unfolded protein response. Mol. Cell 5, 897–904 (2000).

    Article  CAS  Google Scholar 

  8. Cox, J.S., Shamu, C.E. & Walter, P. Transcriptional induction of genes encoding endoplasmic reticulum resident proteins requires a transmembrane protein kinase. Cell 73, 1197–1206 (1993).

    Article  CAS  Google Scholar 

  9. Mori, K., Ma, W., Gething, M.J. & Sambrook, J. A transmembrane protein with a cdc2+/CDC28-related kinase activity is required for signaling from the ER to the nucleus. Cell 74, 743–756 (1993).

    Article  CAS  Google Scholar 

  10. Urano, F. et al. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 287, 664–666 (2000).

    Article  CAS  Google Scholar 

  11. Nishitoh, H. et al. ASK1 is essential for endoplasmic reticulum stress-induced neuronal cell death triggered by expanded polyglutamine repeats. Genes Dev. 16, 1345–1355 (2002).

    Article  CAS  Google Scholar 

  12. Calfon, M. et al. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature 415, 92–96 (2002).

    Article  CAS  Google Scholar 

  13. Lee, K. et al. IRE1-mediated unconventional mRNA splicing and S2P-mediated ATF6 cleavage merge to regulate XBP1 in signaling the unfolded protein response. Genes Dev. 16, 452–466 (2002).

    Article  CAS  Google Scholar 

  14. Yoshida, H., Matsui, T., Yamamoto, A., Okada, T. & Mori, K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 107, 881–891 (2001).

    Article  CAS  Google Scholar 

  15. Lee, A.H., Iwakoshi, N.N. & Glimcher, L.H. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol. Cell. Biol. 23, 7448–7459 (2003).

    Article  CAS  Google Scholar 

  16. Sriburi, R. et al. Coordinate regulation of phospholipid biosynthesis and secretory pathway gene expression in XBP-1(S)–induced endoplasmic reticulum biogenesis. J. Biol. Chem. 282, 7024–7034 (2007).

    Article  CAS  Google Scholar 

  17. Sriburi, R., Jackowski, S., Mori, K. & Brewer, J.W. XBP1: a link between the unfolded protein response, lipid biosynthesis and biogenesis of the endoplasmic reticulum. J. Cell Biol. 167, 35–41 (2004).

    Article  CAS  Google Scholar 

  18. Ozcan, U. et al. Endoplasmic reticulum stress links obesity, insulin action and type 2 diabetes. Science 306, 457–461 (2004).

    Article  Google Scholar 

  19. Ozcan, L. et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 9, 35–51 (2009).

    Article  CAS  Google Scholar 

  20. Saltiel, A.R. & Pessin, J.E. Insulin signaling pathways in time and space. Trends Cell Biol. 12, 65–71 (2002).

    Article  CAS  Google Scholar 

  21. Nandi, A., Kitamura, Y., Kahn, C.R. & Accili, D. Mouse models of insulin resistance. Physiol. Rev. 84, 623–647 (2004).

    Article  CAS  Google Scholar 

  22. Engelman, J.A., Luo, J. & Cantley, L.C. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat. Rev. Genet. 7, 606–619 (2006).

    Article  CAS  Google Scholar 

  23. Taniguchi, C.M., Emanuelli, B. & Kahn, C.R. Critical nodes in signalling pathways: insights into insulin action. Nat. Rev. Mol. Cell Biol. 7, 85–96 (2006).

    Article  CAS  Google Scholar 

  24. Bader, A.G., Kang, S., Zhao, L. & Vogt, P.K. Oncogenic PI3K deregulates transcription and translation. Nat. Rev. Cancer 5, 921–929 (2005).

    Article  CAS  Google Scholar 

  25. Zheng, Y., Bagrodia, S. & Cerione, R.A. Activation of phosphoinositide 3-kinase activity by Cdc42Hs binding to p85. J. Biol. Chem. 269, 18727–18730 (1994).

    CAS  PubMed  Google Scholar 

  26. Okkenhaug, K. & Vanhaesebroeck, B. New responsibilities for the PI3K regulatory subunit p85α. Sci. STKE 2001, pe1 (2001).

    CAS  PubMed  Google Scholar 

  27. Furuya, F., Ying, H., Zhao, L. & Cheng, S.Y. Novel functions of thyroid hormone receptor mutants: beyond nucleus-initiated transcription. Steroids 72, 171–179 (2007).

    Article  CAS  Google Scholar 

  28. Tsuboi, N. et al. The tyrosine phosphatase CD148 interacts with the p85 regulatory subunit of phosphoinositide 3-kinase. Biochem. J. 413, 193–200 (2008).

    Article  CAS  Google Scholar 

  29. Déléris, P., Gayral, S. & Breton-Douillon, M. Nuclear Ptdlns(3,4,5)P3 signaling: an ongoing story. J. Cell. Biochem. 98, 469–485 (2006).

    Article  Google Scholar 

  30. Sephton, C.F. & Mousseau, D.D. Dephosphorylation of Akt in C6 cells grown in serum-free conditions corresponds with redistribution of p85/PI3K to the nucleus. J. Neurosci. Res. 86, 675–682 (2008).

    Article  CAS  Google Scholar 

  31. Dai, Y. et al. Haloperidol induces the nuclear translocation of phosphatidylinositol 3′-kinase to disrupt Akt phosphorylation in PC12 cells. J. Psychiatry Neurosci. 32, 323–330 (2007).

    PubMed  PubMed Central  Google Scholar 

  32. Oyadomari, S., Harding, H.P., Zhang, Y., Oyadomari, M. & Ron, D. Dephosphorylation of translation initiation factor 2α enhances glucose tolerance and attenuates hepatosteatosis in mice. Cell Metab. 7, 520–532 (2008).

    Article  CAS  Google Scholar 

  33. Lin, J.H., Walter, P. & Yen, T.S. Endoplasmic reticulum stress in disease pathogenesis. Annu. Rev. Pathol. 3, 399–425 (2008).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Ozcan laboratory for their contributions to this project. We are grateful to L. Cantley (Harvard Medical School) for kindly providing us with the Pik3r1f/f;Pik3r2−/− mice. We would like to thank the Children's Hospital Boston Proteomics Core Facility and H. Steen for the tandem mass spectroscopy analysis. This study was supported by junior faculty start-up funds provided to U.O. by Children's Hospital Boston, Translational Research Award, an RO1 grant (R01DK081009) provided to U.O., and Timothy Murphy funds provided to Division of Endocrinology, Children's Hospital Boston.

Author information

Authors and Affiliations

Authors

Contributions

S.W.P. came up with the hypothesis, designed and performed the experiments, analyzed the data and wrote the manuscript. Y.Z., J.L., A.L., C.S. and J.C. performed the experiments. K.U. came up with the hypothesis and provided reagents. U.O. came up with the hypothesis, designed and performed the experiments, analyzed the data and wrote the manuscript.

Corresponding authors

Correspondence to Kohjiro Ueki or Umut Ozcan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Methods (PDF 9192 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Park, S., Zhou, Y., Lee, J. et al. The regulatory subunits of PI3K, p85α and p85β, interact with XBP-1 and increase its nuclear translocation. Nat Med 16, 429–437 (2010). https://doi.org/10.1038/nm.2099

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2099

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