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:

NR4A orphan nuclear receptors are transcriptional regulators of hepatic glucose metabolism

Abstract

Hepatic glucose production is crucial for glucose homeostasis, and its dysregulation contributes to the pathogenesis of diabetes. Here, we show that members of the NR4A family of ligand-independent orphan nuclear receptors are downstream mediators of cAMP action in the hormonal control of gluconeogenesis. Hepatic expression of Nur77, Nurr1 and NOR1 is induced by the cAMP axis in response to glucagon and fasting in vivo and is increased in diabetic mice that exhibit elevated gluconeogenesis. Adenoviral expression of Nur77 induces genes involved in gluconeogenesis, stimulates glucose production both in vitro and in vivo, and raises blood glucose levels. Conversely, expression of an inhibitory mutant Nur77 receptor antagonizes gluconeogenic gene expression and lowers blood glucose levels in db/db mice. These results outline a previously unrecognized role for orphan nuclear receptors in the transcriptional control of glucose homeostasis.

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: Induction of NR4A receptor expression in liver in response to the glucagon-cAMP axis and fasting.
Figure 2: Complementary regulation of glucose metabolism by Nur77 and PGC-1α in primary hepatocytes.
Figure 3: NR4A proteins are direct regulators of genes involved in glucose metabolism.
Figure 4: Activation of gluconeogenesis by Nur77 in vivo.
Figure 5: Inhibition of NR4A receptor activity lowers blood glucose in physiology and diabetes.

Similar content being viewed by others

References

  1. Saltiel, A.R. & Kahn, C.R. Insulin signalling and the regulation of glucose and lipid metabolism. Nature 414, 799–806 (2001).

    Article  CAS  Google Scholar 

  2. DeFronzo, R.A., Bonadonna, R.C. & Ferrannini, E. Pathogenesis of NIDDM. A balanced overview. Diabetes Care 15, 318–368 (1992).

    Article  CAS  Google Scholar 

  3. Saltiel, A.R. New perspectives into the molecular pathogenesis and treatment of type 2 diabetes. Cell 104, 517–529 (2001).

    Article  CAS  Google Scholar 

  4. Trinh, K.Y., O'Doherty, R.M., Anderson, P., Lange, A.J. & Newgard, C.B. Perturbation of fuel homeostasis caused by overexpression of the glucose-6-phosphatase catalytic subunit in liver of normal rats. J. Biol. Chem. 273, 31615–31620 (1998).

    Article  CAS  Google Scholar 

  5. Valera, A., Pujol, A., Pelegrin, M. & Bosch, F. Transgenic mice overexpressing phosphoenolpyruvate carboxykinase develop non-insulin-dependent diabetes mellitus. Proc. Natl. Acad. Sci. USA 91, 9151–9154 (1994).

    Article  CAS  Google Scholar 

  6. Pilkis, S.J. & Granner, D.K. Molecular physiology of the regulation of hepatic gluconeogenesis and glycolysis. Annu. Rev. Physiol. 54, 885–909 (1992).

    Article  CAS  Google Scholar 

  7. Montminy, M. Transcriptional regulation by cyclic AMP. Annu. Rev. Biochem. 66, 807–822 (1997).

    Article  CAS  Google Scholar 

  8. Montminy, M., Koo, S.H. & Zhang, X. The CREB family: key regulators of hepatic metabolism. Ann. Endocrinol. (Paris) 65, 73–75 (2004).

    Article  CAS  Google Scholar 

  9. Herzig, S. et al. CREB regulates hepatic gluconeogenesis through the coactivator PGC-1. Nature 413, 179–183 (2001).

    Article  CAS  Google Scholar 

  10. Yoon, J.C. et al. Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1. Nature 413, 131–138 (2001).

    Article  CAS  Google Scholar 

  11. Koo, S.H. et al. The CREB coactivator TORC2 is a key regulator of fasting glucose metabolism. Nature 437, 1109–1111 (2005).

    Article  CAS  Google Scholar 

  12. Lin, J. et al. Defects in adaptive energy metabolism with CNS-linked hyperactivity in PGC-1α null mice. Cell 119, 121–135 (2004).

    Article  CAS  Google Scholar 

  13. Zhang, L., Rubins, N.E., Ahima, R.S., Greenbaum, L.E. & Kaestner, K.H. Foxa2 integrates the transcriptional response of the hepatocyte to fasting. Cell Metab. 2, 141–148 (2005).

    Article  Google Scholar 

  14. Puigserver, P. et al. Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1α interaction. Nature 423, 550–555 (2003).

    Article  CAS  Google Scholar 

  15. Wang, Z. et al. Structure and function of Nurr1 identifies a class of ligand-independent nuclear receptors. Nature 423, 555–560 (2003).

    Article  CAS  Google Scholar 

  16. Baker, K.D. et al. The Drosophila orphan nuclear receptor DHR38 mediates an atypical ecdysteroid signaling pathway. Cell 113, 731–742 (2003).

    Article  CAS  Google Scholar 

  17. Philips, A. et al. Novel dimeric Nur77 signaling mechanism in endocrine and lymphoid cells. Mol. Cell. Biol. 17, 5946–5951 (1997).

    Article  CAS  Google Scholar 

  18. Wilson, T.E., Fahrner, T.J., Johnston, M. & Milbrandt, J. Identification of the DNA binding site for NGFI-B by genetic selection in yeast. Science 252, 1296–1300 (1991).

    Article  CAS  Google Scholar 

  19. Winoto, A. & Littman, D.R. Nuclear hormone receptors in T lymphocytes. Cell 109, Suppl, S57–S66 (2002).

    Article  CAS  Google Scholar 

  20. Pei, L., Castrillo, A., Chen, M., Hoffmann, A. & Tontonoz, P. Induction of NR4A orphan nuclear receptor expression in macrophages in response to inflammatory stimuli. J. Biol. Chem. 280, 29256–29262 (2005).

    Article  CAS  Google Scholar 

  21. Milbrandt, J. Nerve growth factor induces a gene homologous to the glucocorticoid receptor gene. Neuron 1, 183–188 (1988).

    Article  CAS  Google Scholar 

  22. Woronicz, J.D., Calnan, B., Ngo, V. & Winoto, A. Requirement for the orphan steroid receptor Nur77 in apoptosis of T-cell hybridomas. Nature 367, 277–281 (1994).

    Article  CAS  Google Scholar 

  23. Liu, Z.G., Smith, S.W., McLaughlin, K.A., Schwartz, L.M. & Osborne, B.A. Apoptotic signals delivered through the T-cell receptor of a T-cell hybrid require the immediate-early gene nur77. Nature 367, 281–284 (1994).

    Article  CAS  Google Scholar 

  24. Zetterstrom, R.H. et al. Dopamine neuron agenesis in Nurr1-deficient mice. Science 276, 248–250 (1997).

    Article  CAS  Google Scholar 

  25. Saucedo-Cardenas, O. et al. Nurr1 is essential for the induction of the dopaminergic phenotype and the survival of ventral mesencephalic late dopaminergic precursor neurons. Proc. Natl. Acad. Sci. USA 95, 4013–4018 (1998).

    Article  CAS  Google Scholar 

  26. Maxwell, M.A. et al. Nur77 regulates lipolysis in skeletal muscle cells. Evidence for cross-talk between the beta-adrenergic and an orphan nuclear hormone receptor pathway. J. Biol. Chem. 280, 12573–12584 (2005).

    Article  CAS  Google Scholar 

  27. Kovalovsky, D. et al. Activation and induction of NUR77/NURR1 in corticotrophs by CRH/cAMP: involvement of calcium, protein kinase A, and MAPK pathways. Mol. Endocrinol. 16, 1638–1651 (2002).

    Article  CAS  Google Scholar 

  28. Koo, S.H. et al. PGC-1 promotes insulin resistance in liver through PPAR-α–dependent induction of TRB-3. Nat. Med. 10, 530–534 (2004).

    Article  CAS  Google Scholar 

  29. Shepherd, P.R. & Kahn, B.B. Glucose transporters and insulin action–implications for insulin resistance and diabetes mellitus. N. Engl. J. Med. 341, 248–257 (1999).

    Article  CAS  Google Scholar 

  30. Rhee, J. et al. Regulation of hepatic fasting response by PPARγ coactivator-1alpha (PGC-1): requirement for hepatocyte nuclear factor 4α in gluconeogenesis. Proc. Natl. Acad. Sci. USA 100, 4012–4017 (2003).

    Article  CAS  Google Scholar 

  31. Arkenbout, E.K. et al. Protective function of transcription factor TR3 orphan receptor in atherogenesis: decreased lesion formation in carotid artery ligation model in TR3 transgenic mice. Circulation 106, 1530–1535 (2002).

    Article  CAS  Google Scholar 

  32. Spiegelman, B.M. & Heinrich, R. Biological control through regulated transcriptional coactivators. Cell 119, 157–167 (2004).

    Article  CAS  Google Scholar 

  33. Conkright, M.D. et al. Genome-wide analysis of CREB target genes reveals a core promoter requirement for cAMP responsiveness. Mol. Cell 11, 1101–1108 (2003).

    Article  CAS  Google Scholar 

  34. Law, S.W., Conneely, O.M., DeMayo, F.J. & O'Malley, B.W. Identification of a new brain-specific transcription factor, NURR1. Mol. Endocrinol. 6, 2129–2135 (1992).

    CAS  PubMed  Google Scholar 

  35. Ohkura, N., Hijikuro, M., Yamamoto, A. & Miki, K. Molecular cloning of a novel thyroid/steroid receptor superfamily gene from cultured rat neuronal cells. Biochem. Biophys. Res. Commun. 205, 1959–1965 (1994).

    Article  CAS  Google Scholar 

  36. Murphy, E.P. & Conneely, O.M. Neuroendocrine regulation of the hypothalamic pituitary adrenal axis by the nurr1/nur77 subfamily of nuclear receptors. Mol. Endocrinol. 11, 39–47 (1997).

    Article  CAS  Google Scholar 

  37. Fahrner, T.J., Carroll, S.L. & Milbrandt, J. The NGFI-B protein, an inducible member of the thyroid/steroid receptor family, is rapidly modified posttranslationally. Mol. Cell. Biol. 10, 6454–6459 (1990).

    Article  CAS  Google Scholar 

  38. Erion, M.D. et al. MB06322 (CS-917): A potent and selective inhibitor of fructose 1,6-bisphosphatase for controlling gluconeogenesis in type 2 diabetes. Proc. Natl. Acad. Sci. USA 102, 7970–7975 (2005).

    Article  CAS  Google Scholar 

  39. Wansa, K.D., Harris, J.M., Yan, G., Ordentlich, P. & Muscat, G.E. The AF-1 domain of the orphan nuclear receptor NOR-1 mediates trans-activation, coactivator recruitment, and activation by the purine anti-metabolite 6-mercaptopurine. J. Biol. Chem. 278, 24776–24790 (2003).

    Article  CAS  Google Scholar 

  40. Ordentlich, P., Yan, Y., Zhou, S. & Heyman, R.A. Identification of the antineoplastic agent 6-mercaptopurine as an activator of the orphan nuclear hormone receptor Nurr1. J. Biol. Chem. 278, 24791–24799 (2003).

    Article  CAS  Google Scholar 

  41. Xu, J. et al. Peroxisome proliferator-activated receptor α (PPARα) influences substrate utilization for hepatic glucose production. J. Biol. Chem. 277, 50237–50244 (2002).

    Article  CAS  Google Scholar 

  42. Szafranek, J., Pfaffenberger, C.D. & Horning, E.C. The mass spectra of some per-O-acetylaldononitriles. Carbohydr. Res. 38, 97–105 (1974).

    Article  CAS  Google Scholar 

  43. Katz, J., Lee, W.N., Wals, P.A. & Bergner, E.A. Studies of glycogen synthesis and the Krebs cycle by mass isotopomer analysis with [U-13C]glucose in rats. J. Biol. Chem. 264, 12994–13004 (1989).

    CAS  PubMed  Google Scholar 

  44. Lee, W.N., Byerley, L.O., Bergner, E.A. & Edmond, J. Mass isotopomer analysis: theoretical and practical considerations. Biol. Mass Spectrom. 20, 451–458 (1991).

    Article  CAS  Google Scholar 

  45. Laffitte, B.A. et al. Activation of liver X receptor improves glucose tolerance through coordinate regulation of glucose metabolism in liver and adipose tissue. Proc. Natl. Acad. Sci. USA 100, 5419–5424 (2003).

    Article  CAS  Google Scholar 

  46. Tontonoz, P. et al. Adipocyte-specific transcription factor ARF6 is a heterodimeric complex of two nuclear hormone receptors, PPAR γ and RXR α. Nucleic Acids Res. 22, 5628–5634 (1994).

    Article  CAS  Google Scholar 

  47. Castrillo, A., Diaz-Guerra, M.J., Hortelano, S., Martin-Sanz, P. & Bosca, L. Inhibition of IκB kinase and IκB phosphorylation by 15-deoxy-Δ(12,14)-prostaglandin J(2) in activated murine macrophages. Mol. Cell. Biol. 20, 1692–1698 (2000).

    Article  CAS  Google Scholar 

  48. Castrillo, A., Joseph, S.B., Marathe, C., Mangelsdorf, D.J. & Tontonoz, P. Liver X receptor-dependent repression of matrix metalloproteinase-9 expression in macrophages. J. Biol. Chem. 278, 10443–10449 (2003).

    Article  CAS  Google Scholar 

  49. Pei, L., Castrillo, A. & Tontonoz, P. Regulation of macrophage inflammatory gene expression by the orphan nuclear receptor Nur77. Mol. Endocrinol. 20, 786–794 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank O. Conneely and S. Mullican for discussions and for sharing unpublished data. We are grateful to B. Spiegelman for PGC-1α adenovirus; M. Montminy for A-CREB, PGC-1α RNAi and control scrambled RNAi adenovirus; S. Tetradis for Nur77 and Nurr1 adenovirus; and T. Perlmann for Nurr1 adenovirus. We thank L. Wu, M. Johnson and the UCLA viral vector core (N. Kasahara, D. Cohen and E. Faure, funded by US National Institutes of Health (NIH) grant P30 DK041301) for help with adenovirus experiments. We also thank E. Saez, L. Chao, S. Beaven, A. Castrillo, M. Chen, Y. Zhang, and S. Zhang for their input. P.T is an Investigator of the Howard Hughes Medical Institute. This work was supported by grants from the NIH (HL30568 to P.T. and DK58132 to I.J.K.) and a Bristol-Myers Squibb Freedom to Discover Award.

Author information

Authors and Affiliations

Authors

Contributions

L.P. designed and performed experiments and wrote the manuscript. H.W. and B.V. designed and performed experiments. D.C.W. performed experiments. I.J.K. designed experiments, performed data analysis and edited the manuscript. P.T. designed experiments and wrote the manuscript.

Corresponding author

Correspondence to Peter Tontonoz.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Fig. 1

Induction of NR4A expression by the cAMP axis requires CREB family proteins but not PGC-1α. (PDF 289 kb)

Supplementary Fig. 2

Regulation of gene expression by Nur77 does not require PGC-1α. (PDF 247 kb)

Supplementary Fig. 3

Sequences and locations of putative NBREs in promoters of gluconeogenic genes. (PDF 302 kb)

Supplementary Fig. 4

Effects of Nur77 expression on key genes involved in control of glucose metabolism. (PDF 175 kb)

Supplementary Fig. 5

A mutant Nur77 receptor antagonizes NR4A action on target promoters. (PDF 212 kb)

Supplementary Table 1

Real-time PCR (SyberGreen) primers used. (PDF 15 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Pei, L., Waki, H., Vaitheesvaran, B. et al. NR4A orphan nuclear receptors are transcriptional regulators of hepatic glucose metabolism. Nat Med 12, 1048–1055 (2006). https://doi.org/10.1038/nm1471

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm1471

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