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:

Depletion of fat-resident Treg cells prevents age-associated insulin resistance

Abstract

Age-associated insulin resistance (IR) and obesity-associated IR are two physiologically distinct forms of adult-onset diabetes. While macrophage-driven inflammation is a core driver of obesity-associated IR1,2,3,4,5,6, the underlying mechanisms of the obesity-independent yet highly prevalent age-associated IR7 are largely unexplored. Here we show, using comparative adipo-immune profiling in mice, that fat-resident regulatory T cells, termed fTreg cells, accumulate in adipose tissue as a function of age, but not obesity. Supporting the existence of two distinct mechanisms underlying IR, mice deficient in fTreg cells are protected against age-associated IR, yet remain susceptible to obesity-associated IR and metabolic disease. By contrast, selective depletion of fTreg cells via anti-ST2 antibody treatment increases adipose tissue insulin sensitivity. These findings establish that distinct immune cell populations within adipose tissue underlie ageing- and obesity-associated IR, and implicate fTreg cells as adipo-immune drivers and potential therapeutic targets in the treatment of age-associated IR.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: fTreg cells are selectively enriched in aged mice.
Figure 2: fTreg knockout mice are protected from general hallmarks of metabolic ageing.
Figure 3: Loss of fTreg cells protects against the clinical hallmarks of age-associated IR.
Figure 4: fTreg cell depletion improves adipose glucose uptake.

Similar content being viewed by others

Accession codes

Primary accessions

Sequence Read Archive

Data deposits

RNA-Seq data can be accessed in the NCBI Sequence Read Archive under the accession SRP053799.

References

  1. Ferrante, A. W. Jr. Macrophages, fat, and the emergence of immunometabolism. J. Clin. Invest. 123, 4992–4993 (2013)

    Article  CAS  Google Scholar 

  2. Lumeng, C. N. & Saltiel, A. R. Inflammatory links between obesity and metabolic disease. J. Clin. Invest. 121, 2111–2117 (2011)

    Article  CAS  Google Scholar 

  3. Mathis, D. Immunological goings-on in visceral adipose tissue. Cell Metab. 17, 851–859 (2013)

    Article  CAS  Google Scholar 

  4. Osborn, O. & Olefsky, J. M. The cellular and signaling networkslinking the immune system and metabolism in disease. Nature Med. 18, 363–374 (2012)

    Article  CAS  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  Google Scholar 

  6. Xu, H. 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 

  7. CDC. 2014 National Diabetes Statistics Report; http://www.cdc.gov/diabetes/data/statistics/2014statisticsreport.html (2014)

  8. Wu, D. et al. Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis. Science 332, 243–247 (2011)

    Article  CAS  ADS  Google Scholar 

  9. Qiu, Y. et al. Eosinophils and type 2 cytokine signaling in macrophages orchestrate development of functional beige fat. Cell 157, 1292–1308 (2014)

    Article  CAS  Google Scholar 

  10. Rao, R. R. et al. Meteorin-like is a hormone that regulates immune-adipose interactions to increase beige fat thermogenesis. Cell 157, 1279–1291 (2014)

    Article  CAS  Google Scholar 

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

    Article  CAS  ADS  Google Scholar 

  12. Fujisaka, S. et al. Regulatory mechanisms for adipose tissue m1 and m2 macrophages in diet-induced obese mice. Diabetes 58, 2574–2582 (2009)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  14. Lumeng, C. N. et al. Aging is associated with an increase in T cells and inflammatory macrophages in visceral adipose tissue. J. Immunol. 187, 6208–6216 (2011)

    Article  CAS  Google Scholar 

  15. Cipolletta, D. et al. PPAR-γ is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature 486, 549–553 (2012)

    Article  CAS  ADS  Google Scholar 

  16. Houtkooper, R. H. et al. The metabolic footprint of aging in mice. Sci. Rep. 1, 134 (2011)

    Article  Google Scholar 

  17. Sun, K. et al. Dichotomous effects of VEGF-A on adipose tissue dysfunction. Proc. Natl Acad. Sci. USA 109, 5874–5879 (2012)

    Article  CAS  ADS  Google Scholar 

  18. Khan, T. et al. Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol. Cell. Biol. 29, 1575–1591 (2009)

    Article  CAS  Google Scholar 

  19. Steppan, C. M. et al. The hormone resistin links obesity to diabetes. Nature 409, 307–312 (2001)

    Article  CAS  ADS  Google Scholar 

  20. Schwartz, D. R. & Lazar, M. A. Human resistin: found in translation from mouse to man. Trends Endocrinol. Metab. 22, 259–265 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Webster, K. E. et al. In vivo expansion of T reg cells with IL-2-mAb complexes: induction of resistance to EAE and long-term acceptance of islet allografts without immunosuppression. J. Exp. Med. 206, 751–760 (2009)

    Article  CAS  Google Scholar 

  22. Feuerer, M. et al. Genomic definition of multiple ex vivo regulatory T cell subphenotypes. Proc. Natl Acad. Sci. USA 107, 5919–5924 (2010)

    Article  CAS  ADS  Google Scholar 

  23. Josefowicz, S. Z., Lu, L.-F. & Rudensky, A. Y. Regulatory T cells: mechanisms of differentiation and function. Annu. Rev. Immunol. 30, 531–564 (2012)

    Article  CAS  Google Scholar 

  24. Ohkura, N., Kitagawa, Y. & Sakaguchi, S. Development and maintenance of regulatory T cells. Immunity 38, 414–423 (2013)

    Article  CAS  Google Scholar 

  25. Wing, K. et al. CTLA-4 control over Foxp3+ regulatory T cell function. Science 322, 271–275 (2008)

    Article  CAS  ADS  Google Scholar 

  26. Fontenot, J. D. et al. A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nature Immunol. 6, 1142–1151 (2005)

    Article  CAS  Google Scholar 

  27. Vasanthakumar, A. et al. The transcriptional regulators IRF4, BATF and IL-33 orchestrate development and maintenance of adipose tissue–resident regulatory T cells. Nature Immunol. 16, 276–285 (2015)

    Article  CAS  Google Scholar 

  28. Schiering, C. et al. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature 513, 564–568 (2014)

    Article  CAS  ADS  Google Scholar 

  29. Asterholm, I. W. et al. Adipocyte inflammation is essential for healthy adipose tissue expansion and remodeling. Cell Metab. 20, 103–118 (2014)

    Article  Google Scholar 

  30. Rubtsov, Y. P. et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28, 546–558 (2008)

    Article  CAS  Google Scholar 

  31. He, W. et al. Adipose-specific peroxisome proliferator-activated receptor gamma knockout causes insulin resistance in fat and liver but not in muscle. Proc. Natl Acad. Sci. USA 100, 15712–15717 (2003)

    Article  CAS  ADS  Google Scholar 

  32. Liston, A. et al. Differentiation of regulatory Foxp3+ T cells in the thymic cortex. Proc. Natl Acad. Sci. USA 105, 11903–11908 (2008)

    Article  CAS  ADS  Google Scholar 

  33. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013)

    Article  CAS  Google Scholar 

  34. Trapnell, C. et al. Differential analysis of gene regulation at transcript resolution with RNA-seq. Nature Biotechnol. 31, 46–53 (2012)

    Article  Google Scholar 

  35. Roberts, A., Pimentel, H., Trapnell, C. & Pachter, L. Identification of novel transcripts in annotated genomes using RNA-Seq. Bioinformatics 27, 2325–2329 (2011)

    Article  CAS  Google Scholar 

  36. Monticelli, L. A. et al. Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nature Immunol. 12, 1045–1054 (2011)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank L. Chong, J. Alvarez, Y. Dai, S. Kaufman and B. Collins for technical assistance, L. Ong and C. Brondos for administrative assistance, and J. Simon for assistance with graphics. S.P.B. is supported by National Institutes of Health (NIH) grants F30 DK096828 and T32 GM007198. C.L. and M.D. are funded by grants from the National Health and Medical Research Council of Australia Project grants 512354, 632886 and 1043199. R.M.E. is an Investigator of the Howard Hughes Medical Institute (HHMI) at the Salk Institute and March of Dimes Chair in Molecular and Developmental Biology, and is supported by NIH grants DK057978, DK090962, HL088093, HL105278 and ES010337, the Glenn Foundation for Medical Research, the Leona M. and Harry B. Helmsley Charitable Trust, Ipsen/Biomeasure, California Institute for Regenerative Medicine and The Ellison Medical Foundation. Y. Zheng is supported by the Nomis Foundation, the Rita Allen Foundation, the Emerald Foundation, the Hearst Foundation, the National Multiple Sclerosis Society, and National Institutes of Health (AI099295 and AI107027). This work was also supported by National Cancer Institute funded Salk Institute Cancer Center core facilities (CA014195) and the James B. Pendleton Charitable Trust.

Author information

Authors and Affiliations

Authors

Contributions

S.P.B., J.M.S., M.D., R.M.E. and Y. Zheng designed and supervised the research. S.P.B., J.M.S., S.F., S.L., Y. Zhang, A.C., C.Z. and Y.L. performed research. S.P.B., J.M.S., M.L., C.L., A.R.A., R.T.Y., M.D., R.M.E. and Y. Zheng analysed data. S.P.B., J.M.S., S.L., A.R.A., R.T.Y., M.D., R.M.E. and Y. Zheng wrote the manuscript.

Corresponding authors

Correspondence to Ronald M. Evans or Ye Zheng.

Ethics declarations

Competing interests

S.B., R.M.E., Y. Zheng and M.D. are co-inventors of technologies related to methods of targeting fat Treg cells to prevent insulin resistance.

Extended data figures and tables

Extended Data Figure 1 Schematic outlining study premise and selected gating strategies used to generate AIPs.

a, Schematic outlining study premise. bd, AIPs were generated through the use of several distinct antibody cocktails. Here, using Foxp3Cre (Foxp3-IRES-YFP-Cre) reporter mice, we show how the stromal vascular fraction of VAT was analysed by flow cytometry to identify several T cell subtypes (b), macrophage subsets (c) and eosinophils and neutrophils (d).

Extended Data Figure 2 AIPs of aged fTreg knockout and control mice in VAT, SAT and spleen.

a, AIPs of aged (~14 months) fTreg knockout and control male mice depicting immune cell abundance, expressed as percentage of CD45.2+ cells. Entirety of immune compartment (top) is further divided into pan-macrophage (middle) and non-macrophage (bottom) pie charts (n = 9 mice per group). bd, Immune cell abundance between fTreg knockout and control mice, expressed as cells per gram of VAT (b), SAT (c) and spleen (d) (n = 9 mice per group). Data are mean ± s.e.m. #, false discovery rate < 2%.

Extended Data Figure 3 Aged fTreg knockout mice do not show signs of systemic autoimmunity or breakdown in peripheral tolerance.

a, Percentage of splenic naive CD4+ T cells as defined by CD62hi CD44lo relative to total CD4+ CD25 Foxp3Cre population (n = 9 mice per group). b, Representative histology of gastrointestinal tract—duodenum, jejunum, ileum and colon (left to right) (n = 3 mice per group). There were no significant lesions observed or differences in inflammation, epithelial changes, or mucosal architecture between the two groups (H&E, original magnification, ×100). Scale bar, 50 μm. c, Histopathology score in the small intestine and colon of fTreg knockout and control mice. The severity and extent of inflammation and epithelial changes as well as mucosal architecture were each graded on a score of 1 (minimal) to 5 (severe) and added to obtain an overall score over 20. There were minimal inflammatory changes with no significant differences between groups. d, Multiplex inflammation panel of serum from fTreg knockout and control mice (n = 4 pooled samples (3 mice per sample) per group). Data are mean ± s.e.m. *P < 0.05, ***P < 0.001, Student’s t-test.

Extended Data Figure 4 Weight-matched cohorts’ body weights and adipocyte size frequency in VAT of aged control and fTreg knockout mice.

a, Body weights of fTreg knockout and control male mice used in weight-matched metabolic studies in young (12 week; control n = 9; fTreg KO n = 9), aged (36 week; control, n = 9 mice; fTreg KO, n = 11 mice) and obese (diet-induced obese, 12 weeks of HFD starting at 12 weeks; control n = 10; fTreg KO n = 10) settings. b, Frequency of small (0–5,000 μm2), medium (5,000–10,000 μm2) and large (>10,000 μm2) adipocytes in VAT of aged control and fTreg knockout mice (n = 3 mice per group, 850 adipocytes counted from control mice, 269 adipocytes counted from fTreg knockout adipose). Data are mean ± s.e.m.

Extended Data Figure 5 VAT AIPs of obese fTreg knockout and control mice.

a, AIPs of diet-induced obese (16 weeks high fat diet started at 12 weeks) control (n = 6 mice) and fTreg knockout (n = 8 mice) male mice depicting immune cell abundance, expressed as percentage of CD45.2+ cells. Entirety of immune compartment (top) is further divided into pan-macrophage (middle) and non-macrophage (bottom) pie charts. b, Immune cell abundance between fTreg knockout and control mice, expressed as cells per gram of VAT (n = 9 mice per group). Data are mean ± s.e.m.

Extended Data Figure 6 fTreg cells are dispensable for TZDs to exert their therapeutic insulin-sensitizing effect.

a, Scheme used for longitudinal interventional study of control and fTreg knockout mice which indicates when particular assays were conducted and whose results are described in bg, in which rosiglitazone (Rosi) was introduced in diet after firmly establishing obesity with a HFD alone for 12 weeks (n = 8 mice per group). b, Cohort weights during course of study. Black arrow indicates introduction of rosiglitazone to the diet. c, Homeostatic model assessment of IR (HOMA-IR). d, e, Glucose tolerance test (d) and glucose excursions of glucose tolerance test (e) described as area under curve (AUC). f, g, Insulin tolerance test (f) and bar-graph quantitation of relative serum glucose decrease during insulin tolerance test (g) described as area above curve (AAC). h, Scheme used for parallel prophylactic study of control and fTreg knockout mice, the results of which are described in il, in which mice were placed on a HFD or HFD with rosiglitazone for 12 weeks (n = 8 mice per group). i, Cohort weights at end of study. j, HOMA-IR. k, l, Glucose and insulin tolerance tests of control (k) or fTreg knockout (l) mice fed HFD or HFD with rosiglitazone. m, Scheme used to determine temporal relationship of TZD-induced fTreg expansion and TZD-induced insulin-sensitization in wild-type mice, the results of which are described in nq, where mice were fed HFD or HFD with rosiglitazone for up to 11 weeks (n = 10 mice per group, 5 mice of each group were euthanized at 5 weeks after diet introduction and remaining 5 mice were euthanized at 11 weeks). n, HOMA-IR at 4 weeks. o, p, Glucose (o) and insulin (p) tolerance tests at 5 weeks. q, Relative fTreg cell enrichment of mice fed HFD with rosiglitazone versus mice fed HFD alone at 5 and at 11 weeks. Data are mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, Student’s t-test.

Extended Data Figure 7 Increased TNFα levels and gene expression pattern of aged fTreg knockout adipose tissue is consistent with an improved adipose remodelling capacity.

a, TNFα levels quantified by ELISA of whole adipose lysate (~40 weeks, n = 6 per group). bd, FPKM values of all differentially expressed genes (b), differentially expressed collagens (c) and differentially expressed extracellular matrix proteases (d) in VAT from aged fTreg knockout and control mice (~40 weeks, n = 3 mice per group). Data are mean ± s.e.m. ***P < 0.001, Student’s t-test.

Extended Data Figure 8 fTreg cell gene expression and depletion with anti-ST2 antibody treatment.

a, Expression of several canonical Treg cell genes across fat and splenic Treg and fat and splenic Tconv cells. Cells were pooled from 3 and 4 mice before isolating RNA for subsequent RNA-Seq analysis. b, Expression of St2 across all haematopoietic cells catalogued in the ImmGen database. Position of adipose CD4+ CD25+ T cells is marked. c, Total weight before beginning course of anti-ST2 or isotype control antibodies (day 0) and after terminal analysis (day 3) (n = 4 mice per group). d, e, Spleen weight (d) and percentage of splenic naive CD4+ T cells as defined by CD62hi CD44lo relative to total splenic CD45+ CD4+ CD25 T cell population (e) of mice after terminal analysis (day 3, n = 4 mice per group). Data are mean ± s.e.m.

Extended Data Figure 9 Aged fTreg knockout mice are resistant to short-term, but not persistent, HFD-induced weight gain and IR.

ae, Aged control and fTreg knockout mice were placed on HFD and monitored throughout course of diet for weight (a), fasting glucose levels (b), fasting serum insulin levels (c), performance on glucose tolerance test (d), and on insulin tolerance test (e). (Control, n = 10; fTreg KO, n = 11; mice were aged 27–29 weeks and weight-matched before HFD was introduced.) Data are mean ± s.e.m. ns, non-significant, *P < 0.05, **P < 0.01, Student’s t-test.

Extended Data Table 1 Antibodies used to identify the given immune cell type molecularly

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bapat, S., Myoung Suh, J., Fang, S. et al. Depletion of fat-resident Treg cells prevents age-associated insulin resistance. Nature 528, 137–141 (2015). https://doi.org/10.1038/nature16151

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature16151

This article is cited by

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