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:

Pten deletion in RIP-Cre neurons protects against type 2 diabetes by activating the anti-inflammatory reflex

Subjects

Abstract

Inflammation has a critical role in the development of insulin resistance. Recent evidence points to a contribution by the central nervous system in the modulation of peripheral inflammation through the anti-inflammatory reflex. However, the importance of this phenomenon remains elusive in type 2 diabetes pathogenesis. Here we show that rat insulin-2 promoter (Rip)-mediated deletion of Pten, a gene encoding a negative regulator of PI3K signaling, led to activation of the cholinergic anti-inflammatory pathway that is mediated by M2 activated macrophages in peripheral tissues. As such, Rip-cre+ Ptenflox/flox mice showed lower systemic inflammation and greater insulin sensitivity under basal conditions compared to littermate controls, which were abolished when the mice were treated with an acetylcholine receptor antagonist or when macrophages were depleted. After feeding with a high-fat diet, the Pten-deleted mice remained markedly insulin sensitive, which correlated with massive subcutaneous fat expansion. They also exhibited more adipogenesis with M2 macrophage infiltration, both of which were abolished after disruption of the anti-inflammatory efferent pathway by left vagotomy. In summary, we show that Pten expression in Rip+ neurons has a critical role in diabetes pathogenesis through mediating the anti-inflammatory reflex.

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: Rip-cre–mediated Pten deletion leads to PI3K activation and neuronal activation in hypothalamic and brainstem nuclei.
Figure 2: Rip-cre+ Ptenflox/flox mice exhibit predominant M2 macrophage polarization and decreased inflammation.
Figure 3: Rip-cre+ Ptenflox/flox mice have enhanced whole-body and metabolic tissue insulin sensitivity on chow and HFD.
Figure 4: Improved insulin sensitivity in Rip-cre+ Ptenflox/flox mice is dependent on cholinergic activation and is present after HFD.
Figure 5: HFD-fed Rip-cre+ Ptenflox/flox mice demonstrate greater M2 macrophage infiltration and adipogenesis in the visceral compartment.
Figure 6: Macrophage depletion or vagotomy attenuates visceral adipogenesis and peripheral insulin sensitivity.

Similar content being viewed by others

References

  1. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 444, 860–867 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Shoelson, S.E., Lee, J. & Goldfine, A.B. Inflammation and insulin resistance. J. Clin. Invest. 116, 1793–1801 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Osborn, O. & Olefsky, J.M. The cellular and signaling networks linking the immune system and metabolism in disease. Nat. Med. 18, 363–374 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Xu, H. et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Invest. 112, 1821–1830 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Weisberg, S.P. et al. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Invest. 112, 1796–1808 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kanda, H. et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J. Clin. Invest. 116, 1494–1505 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Weisberg, S.P. et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J. Clin. Invest. 116, 115–124 (2006).

    Article  CAS  PubMed  Google Scholar 

  8. Gordon, S. & Taylor, P.R. Monocyte and macrophage heterogeneity. Nat. Rev. Immunol. 5, 953–964 (2005).

    Article  CAS  PubMed  Google Scholar 

  9. Mantovani, A. et al. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 25, 677–686 (2004).

    Article  CAS  PubMed  Google Scholar 

  10. Bronte, V. & Zanovello, P. Regulation of immune responses by l-arginine metabolism. Nat. Rev. Immunol. 5, 641–654 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Van Ginderachter, J.A. et al. Classical and alternative activation of mononuclear phagocytes: picking the best of both worlds for tumor promotion. Immunobiology 211, 487–501 (2006).

    Article  CAS  PubMed  Google Scholar 

  12. Tracey, K.J. Reflex control of immunity. Nat. Rev. Immunol. 9, 418–428 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Andersson, U. & Tracey, K.J. Reflex principles of immunological homeostasis. Annu. Rev. Immunol. 30, 313–335 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Sternberg, E.M. Neural regulation of innate immunity: a coordinated nonspecific host response to pathogens. Nat. Rev. Immunol. 6, 318–328 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Maier, S.F., Goehler, L.E., Fleshner, M. & Watkins, L.R. The role of the vagus nerve in cytokine-to-brain communication. Ann. NY Acad. Sci. 840, 289–300 (1998).

    Article  CAS  PubMed  Google Scholar 

  16. Groves, D.A. & Brown, V.J. Vagal nerve stimulation: a review of its applications and potential mechanisms that mediate its clinical effects. Neurosci. Biobehav. Rev. 29, 493–500 (2005).

    Article  PubMed  Google Scholar 

  17. Borovikova, L.V. et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 405, 458–462 (2000).

    Article  CAS  PubMed  Google Scholar 

  18. Bernik, T.R. et al. Pharmacological stimulation of the cholinergic antiinflammatory pathway. J. Exp. Med. 195, 781–788 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Saeed, R.W. et al. Cholinergic stimulation blocks endothelial cell activation and leukocyte recruitment during inflammation. J. Exp. Med. 201, 1113–1123 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Huston, J.M. et al. Splenectomy inactivates the cholinergic antiinflammatory pathway during lethal endotoxemia and polymicrobial sepsis. J. Exp. Med. 203, 1623–1628 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. de Jonge, W.J. et al. Stimulation of the vagus nerve attenuates macrophage activation by activating the Jak2-STAT3 signaling pathway. Nat. Immunol. 6, 844–851 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Kreier, F. et al. Selective parasympathetic innervation of subcutaneous and intra-abdominal fat—functional implications. J. Clin. Invest. 110, 1243–1250 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Watkins, L.R. & Maier, S.F. Immune regulation of central nervous system functions: from sickness responses to pathological pain. J. Intern. Med. 257, 139–155 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. The, F. et al. Central activation of the cholinergic anti-inflammatory pathway reduces surgical inflammation in experimental post-operative ileus. Br. J. Pharmacol. 163, 1007–1016 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Choudhury, A.I. et al. The role of insulin receptor substrate 2 in hypothalamic and beta cell function. J. Clin. Invest. 115, 940–950 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang, L. et al. Deletion of Pten in pancreatic β-cells protects against deficient β-cell mass and function in mouse models of type 2 diabetes. Diabetes 59, 3117–3126 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Song, J., Xu, Y., Hu, X., Choi, B. & Tong, Q. Brain expression of Cre recombinase driven by pancreas-specific promoters. Genesis 48, 628–634 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wicksteed, B. et al. Conditional gene targeting in mouse pancreatic β-cells: analysis of ectopic Cre transgene expression in the brain. Diabetes 59, 3090–3098 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Backman, S.A. et al. Deletion of Pten in mouse brain causes seizures, ataxia and defects in soma size resembling Lhermitte-Duclos disease. Nat. Genet. 29, 396–403 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Nguyen, K.T. et al. Essential role of Pten in body size determination and pancreatic beta-cell homeostasis in vivo. Mol. Cell. Biol. 26, 4511–4518 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Petersen, A.M. & Pedersen, B.K. The anti-inflammatory effect of exercise. J. Appl. Physiol. 98, 1154–1162 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Petersen, A.M. & Pedersen, B.K. The role of IL-6 in mediating the anti-inflammatory effects of exercise. J. Physiol. Pharmacol. 57 (suppl. 10), 43–51 (2006).

    PubMed  Google Scholar 

  33. Kubota, N. et al. Insulin receptor substrate 2 plays a crucial role in beta cells and the hypothalamus. J. Clin. Invest. 114, 917–927 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Stanger, B.Z. et al. Pten constrains centroacinar cell expansion and malignant transformation in the pancreas. Cancer Cell 8, 185–195 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Cantley, J. et al. Pancreatic deletion of insulin receptor substrate 2 reduces beta and alpha cell mass and impairs glucose homeostasis in mice. Diabetologia 50, 1248–1256 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Lumeng, C.N., Bodzin, J.L. & Saltiel, A.R. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J. Clin. Invest. 117, 175–184 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Bourlier, V. et al. Remodeling phenotype of human subcutaneous adipose tissue macrophages. Circulation 117, 806–815 (2008).

    Article  CAS  PubMed  Google Scholar 

  38. Plum, L. et al. Enhanced PIP3 signaling in POMC neurons causes KATP channel activation and leads to diet-sensitive obesity. J. Clin. Invest. 116, 1886–1901 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Münzberg, H. & Myers, M.G. Jr. Molecular and anatomical determinants of central leptin resistance. Nat. Neurosci. 8, 566–570 (2005).

    Article  PubMed  CAS  Google Scholar 

  40. Rulifson, E.J., Kim, S.K. & Nusse, R. Ablation of insulin-producing neurons in flies: growth and diabetic phenotypes. Science 296, 1118–1120 (2002).

    Article  CAS  PubMed  Google Scholar 

  41. Kodama, E. et al. Insulin-like signaling and the neural circuit for integrative behavior in C. elegans. Genes Dev. 20, 2955–2960 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Gannon, M., Shiota, C., Postic, C., Wright, C.V. & Magnuson, M. Analysis of the Cre-mediated recombination driven by rat insulin promoter in embryonic and adult mouse pancreas. Genesis 26, 139–142 (2000).

    Article  CAS  PubMed  Google Scholar 

  43. Plum, L. et al. Enhanced leptin-stimulated Pi3k activation in the CNS promotes white adipose tissue transdifferentiation. Cell Metab. 6, 431–445 (2007).

    Article  CAS  PubMed  Google Scholar 

  44. Odegaard, J.I. et al. Macrophage-specific PPARγ controls alternative activation and improves insulin resistance. Nature 447, 1116–1120 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Odegaard, J.I. et al. Alternative M2 activation of Kupffer cells by PPARδ ameliorates obesity-induced insulin resistance. Cell Metab. 7, 496–507 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Bouhlel, M.A. et al. PPARγ activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab. 6, 137–143 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Winer, S. et al. Normalization of obesity-associated insulin resistance through immunotherapy. Nat. Med. 15, 921–929 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Winer, D.A. et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat. Med. 17, 610–617 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Feuerer, M. et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat. Med. 15, 930–939 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Mauer, J. et al. Myeloid cell–restricted insulin receptor deficiency protects against obesity-induced inflammation and systemic insulin resistance. PLoS Genet. 6, e1000938 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Andersson, K. & Arner, P. Cholinoceptor-mediated effects on glycerol output from human adipose tissue using in situ microdialysis. Br. J. Pharmacol. 115, 1155–1162 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Olofsson, P.S. et al. α7 nicotinic acetylcholine receptor (α7nAChR) expression in bone marrow–derived non-T cells is required for the inflammatory reflex. Mol. Med. 18, 539–543 (2012).

    Article  CAS  PubMed  Google Scholar 

  53. Kim, J.Y. et al. Obesity-associated improvements in metabolic profile through expansion of adipose tissue. J. Clin. Invest. 117, 2621–2637 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Primeau, V. et al. Characterizing the profile of obese patients who are metabolically healthy. Int. J. Obes. (Lond.) 35, 971–981 (2011).

    Article  CAS  Google Scholar 

  55. Karelis, A.D. et al. The metabolically healthy but obese individual presents a favorable inflammation profile. J. Clin. Endocrinol. Metab. 90, 4145–4150 (2005).

    Article  CAS  PubMed  Google Scholar 

  56. Postic, C. et al. Dual roles for glucokinase in glucose homeostasis as determined by liver and pancreatic beta cell–specific gene knock-outs using Cre recombinase. J. Biol. Chem. 274, 305–315 (1999).

    Article  CAS  PubMed  Google Scholar 

  57. Wijesekara, N. et al. Muscle-specific Pten deletion protects against insulin resistance and diabetes. Mol. Cell. Biol. 25, 1135–1145 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Nguyen, K.T. et al. Essential role of Pten in body size determination and pancreatic beta-cell homeostasis in vivo. Mol. Cell. Biol. 26, 4511–4518 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Choi, D. et al. Erythropoietin protects against diabetes through direct effects on pancreatic beta cells. J. Exp. Med. 207, 2831–2842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Münzberg, H. et al. Appropriate inhibition of orexigenic hypothalamic arcuate nucleus neurons independently of leptin receptor/STAT3 signaling. J. Neurosci. 27, 69–74 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Franklin, K.B.J. & Paxinos, G. The Mouse Brain in Sterotaxic Coordinates (Academic Press, New York, 2008).

Download references

Acknowledgements

We thank Y. Dor (Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School) for providing the Pdx1-Cre-ER mice. This work was supported by grants to M.W. from the Canadian Institutes of Health Research (CIHR; MOP-93707) and the Canadian Diabetes Association (CDA). M.W. holds a Canada Research Chair in Signal Transduction in Diabetes Pathogenesis. M.G.M.Jr. was supported by a grant from the US National Institutes of Health (DK57768). Support was also received from grants to D.A.W. from the CIHR (MOP-119414) and CDA. L.W. was supported by Frederick Banting and Charles Best Canada Graduate Scholarship from CIHR, a Novo Nordisk Graduate Scholarship from the Banting and Best Diabetes Centre, a Canada Graduate Scholarship from the Natural Sciences and Engineering Research Council of Canada (NSERC) and a Comprehensive Research Experience for Medical Students (CREMS) Scholarship from the Faculty of Medicine, University of Toronto. S.T. was supported by a Toronto General Research Institute Postdoctoral Fellowship. C.T.L. is supported by the Eliot Phillipson Clinician Scientist Training Program and a Banting and Best Diabetes Centre Postdoctoral Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

L.W. performed experiments, analyzed data, wrote the manuscript and contributed to the study concept. D.O., S.T., M.B.A., A.J.E. and C.F. performed experiments, analyzed data and prepared data for presentation. C.T.L. analyzed data and edited the manuscript. S.A.S. developed methods and animal models and performed experiments. A.S. and T.W.M. developed animal models. C.J.P., D.A.W. and M.G.M.Jr. contributed to study design, discussion and the manuscript. M.W. supervised the project, developed the study concept and edited the manuscript.

Corresponding author

Correspondence to Minna Woo.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 4660 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, L., Opland, D., Tsai, S. et al. Pten deletion in RIP-Cre neurons protects against type 2 diabetes by activating the anti-inflammatory reflex. Nat Med 20, 484–492 (2014). https://doi.org/10.1038/nm.3527

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3527

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