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.

  • Letter
  • Published:

Macrophage-specific PPARγ controls alternative activation and improves insulin resistance

Abstract

Obesity and insulin resistance, the cardinal features of metabolic syndrome, are closely associated with a state of low-grade inflammation1,2. In adipose tissue chronic overnutrition leads to macrophage infiltration, resulting in local inflammation that potentiates insulin resistance3,4. For instance, transgenic expression of Mcp1 (also known as chemokine ligand 2, Ccl2) in adipose tissue increases macrophage infiltration, inflammation and insulin resistance5,6. Conversely, disruption of Mcp1 or its receptor Ccr2 impairs migration of macrophages into adipose tissue, thereby lowering adipose tissue inflammation and improving insulin sensitivity5,7. These findings together suggest a correlation between macrophage content in adipose tissue and insulin resistance. However, resident macrophages in tissues display tremendous heterogeneity in their activities and functions, primarily reflecting their local metabolic and immune microenvironment8. While Mcp1 directs recruitment of pro-inflammatory classically activated macrophages to sites of tissue damage5,8, resident macrophages, such as those present in the adipose tissue of lean mice, display the alternatively activated phenotype9. Despite their higher capacity to repair tissue10, the precise role of alternatively activated macrophages in obesity-induced insulin resistance remains unknown. Using mice with macrophage-specific deletion of the peroxisome proliferator activated receptor-γ (PPARγ), we show here that PPARγ is required for maturation of alternatively activated macrophages. Disruption of PPARγ in myeloid cells impairs alternative macrophage activation, and predisposes these animals to development of diet-induced obesity, insulin resistance, and glucose intolerance. Furthermore, gene expression profiling revealed that downregulation of oxidative phosphorylation gene expression in skeletal muscle and liver leads to decreased insulin sensitivity in these tissues. Together, our findings suggest that resident alternatively activated macrophages have a beneficial role in regulating nutrient homeostasis and suggest that macrophage polarization towards the alternative state might be a useful strategy for treating type 2 diabetes.

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: PPARγ regulates alternative macrophage activation.
Figure 2: Mac-PPARγ KO mice are less susceptible to infection by Leishmania major.
Figure 3: Alterations in adipose tissue mass and function in Mac-PPARγ KO mice.
Figure 4: Impaired glucose homeostasis in male Mac-PPARγ KO mice fed a high fat diet.

Similar content being viewed by others

References

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

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

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

    Article  MathSciNet  CAS  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  Google Scholar 

  5. 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  Google Scholar 

  6. Kamei, N. et al. Overexpression of monocyte chemoattractant protein-1 in adipose tissues causes macrophage recruitment and insulin resistance. J. Biol. Chem. 281, 26602–26614 (2006)

    Article  CAS  Google Scholar 

  7. Weisberg, S. P. et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J. Clin. Invest. 116, 115–124 (2005)

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. 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  Google Scholar 

  10. Gordon, S. Alternative activation of macrophages. Nature Rev. Immunol. 3, 23–35 (2003)

    Article  CAS  Google Scholar 

  11. Lehrke, M. & Lazar, M. A. The many faces of PPARγ. Cell 123, 993–999 (2005)

    Article  CAS  Google Scholar 

  12. Huang, J. T. et al. Interleukin-4-dependent production of PPAR-gamma ligands in macrophages by 12/15-lipoxygenase. Nature 400, 378–382 (1999)

    Article  ADS  CAS  Google Scholar 

  13. Vats, D. et al. Oxidative metabolism and PGC-1beta attenuate macrophage-mediated inflammation. Cell Metab. 4, 13–24 (2006)

    Article  MathSciNet  CAS  Google Scholar 

  14. Herbert, D. R. et al. Alternative macrophage activation is essential for survival during schistosomiasis and downmodulates T helper 1 responses and immunopathology. Immunity 20, 623–635 (2004)

    Article  CAS  Google Scholar 

  15. Pauleau, A. L. et al. Enhancer-mediated control of macrophage-specific arginase I expression. J. Immunol. 172, 7565–7573 (2004)

    Article  CAS  Google Scholar 

  16. Evans, R. M., Barish, G. D. & Wang, Y.-X. PPARs and the complex journey to obesity. Nature Med. 10, 355–361 (2004)

    Article  CAS  Google Scholar 

  17. Alexander, J., Satoskar, A. R. & Russell, D. G. Leishmania species: models of intracellular parasitism. J. Cell Sci. 112, 2993–3002 (1999)

    Article  CAS  Google Scholar 

  18. Holscher, C., Arendse, B., Schwegmann, A., Myburgh, E. & Brombacher, F. Impairment of alternative macrophage activation delays cutaneous leishmaniasis in nonhealing BALB/c mice. J. Immunol. 176, 1115–1121 (2006)

    Article  Google Scholar 

  19. Rosen, E. D. & Spiegelman, B. M. Adipocytes as regulators of energy balance and glucose homeostasis. Nature 444, 847–853 (2006)

    Article  ADS  CAS  Google Scholar 

  20. Rutschman, R. et al. Cutting edge: Stat6-dependent substrate depletion regulates nitric oxide production. J. Immunol. 166, 2173–2177 (2001)

    Article  CAS  Google Scholar 

  21. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nature Genet. 34, 267–273 (2003)

    Article  ADS  CAS  Google Scholar 

  22. Patti, M. E. et al. Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: Potential role of PGC1 and NRF1. Proc. Natl Acad. Sci. USA 100, 8466–8471 (2003)

    Article  ADS  CAS  Google Scholar 

  23. Petersen, K. F. et al. Mitochondrial dysfunction in the elderly: possible role in insulin resistance. Science 300, 1140–1142 (2003)

    Article  ADS  CAS  Google Scholar 

  24. Vianna, C. R. et al. Hypomorphic mutation of PGC-1β causes mitochondrial dysfunction and liver insulin resistance. Cell Metab. 4, 453–464 (2006)

    Article  CAS  Google Scholar 

  25. Kelly, D. P. & Scarpulla, R. C. Transcriptional regulatory circuits controlling mitochondrial biogenesis and function. Genes Dev. 18, 357–368 (2004)

    Article  CAS  Google Scholar 

  26. Lin, J., Handschin, C. & Spiegelman, B. M. Metabolic control through the PGC-1 family of transcription coactivators. Cell Metab. 1, 361–370 (2005)

    Article  Google Scholar 

  27. Lazar, M. A. How obesity causes diabetes: not a tall tale. Science 307, 373–375 (2005)

    Article  ADS  CAS  Google Scholar 

  28. Civitarese, A. E. et al. Role of adiponectin in human skeletal muscle bioenergetics. Cell Metab. 4, 75–87 (2006)

    Article  CAS  Google Scholar 

  29. Akiyama, T. E. et al. Conditional disruption of the peroxisome proliferator-activated receptor gamma gene in mice results in lowered expression of ABCA1, ABCG1, and apoE in macrophages and reduced cholesterol efflux. Mol. Cell. Biol. 22, 2607–2619 (2002)

    Article  CAS  Google Scholar 

  30. Lumeng, C. N., Deyoung, S. M. & Saltiel, A. R. Macrophages block insulin action in adipocytes by altering expression of signaling and glucose transport proteins. Am. J. Physiol. Endocrinol. Metab. 292, E166–E174 (2007)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Chawla laboratory for comments, and A. Loh and C. H. Lee for critiquing the manuscript. We also thank P. Murray, J. McKerrow and O. McGuinness for providing key reagents and technical guidance. This work was supported by grants made available to A.C. from the NIH, the Astellas Foundation, Takeda Pharmaceuticals North America, the Rockefeller Brothers Fund and by Goldman Philanthropic Partnerships; and to A.W.F. from the NIH and Columbia DERC. A.C. is a Charles E. Culpeper Medical Scholar. Support was provided by Stanford MSTP (to J.I.O. and A.R.E.), the AHA (to J.I.O.), a HHMI Gilliam fellowship (to A.R.E.), the NRSA (to R.R.R.-G.), and an NIH Training Grant (to L.M.). All animal care was in accordance with Stanford University’s A-PLAC committee guidelines.

Author Contributions J.I.O and R.R.R.-G were involved in project planning, experimental work and data analysis; M.H.G, C.R.M, V.S, L.M., D.V. and A.R.E. performed experimental work; F.B. was involved in project planning; and A.W.F. and A.C. were involved in project planning, data analysis and manuscript preparation.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ajay Chawla.

Ethics declarations

Competing interests

Reprints and permissions information is available at www.nature.com/reprints. The authors declare no competing financial interests.

Supplementary information

Supplementary Information 1

This file contains Supplementary Figures S1-S6 with Legends and Supplementary Table S1. (PDF 2865 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Odegaard, J., Ricardo-Gonzalez, R., Goforth, M. et al. Macrophage-specific PPARγ controls alternative activation and improves insulin resistance. Nature 447, 1116–1120 (2007). https://doi.org/10.1038/nature05894

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature05894

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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