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:

NK cells link obesity-induced adipose stress to inflammation and insulin resistance

Subjects

Abstract

An important cause of obesity-induced insulin resistance is chronic systemic inflammation originating in visceral adipose tissue (VAT). VAT inflammation is associated with the accumulation of proinflammatory macrophages in adipose tissue, but the immunological signals that trigger their accumulation remain unknown. We found that a phenotypically distinct population of tissue-resident natural killer (NK) cells represented a crucial link between obesity-induced adipose stress and VAT inflammation. Obesity drove the upregulation of ligands of the NK cell–activating receptor NCR1 on adipocytes; this stimulated NK cell proliferation and interferon-γ (IFN-γ) production, which in turn triggered the differentiation of proinflammatory macrophages and promoted insulin resistance. Deficiency of NK cells, NCR1 or IFN-γ prevented the accumulation of proinflammatory macrophages in VAT and greatly ameliorated insulin sensitivity. Thus NK cells are key regulators of macrophage polarization and insulin resistance in response to obesity-induced adipocyte stress.

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: A high-fat diet increases IFN-γ production by NK cells in VAT.
Figure 2: VAT-resident NK cells are activated by HFD feeding and promote insulin resistance.
Figure 3: VAT NK cells are required and sufficient for the induction of insulin resistance.
Figure 4: VAT-resident NK cells expand locally in response to an HFD.
Figure 5: Obese VAT induces expression of NKp46-l and stimulates IFN-γ production by NK cells.
Figure 6: NCR1 deficiency delays the development of insulin resistance after DIO.
Figure 7: NK cell–derived IFN-γ induces M1 macrophage formation in response to NCR1 activation.

Similar content being viewed by others

References

  1. Stevens, G.A. et al. National, regional, and global trends in adult overweight and obesity prevalences. Popul. Health Metr. 10, 22 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Danaei, G. et al. National, regional, and global trends in fasting plasma glucose and diabetes prevalence since 1980: systematic analysis of health examination surveys and epidemiological studies with 370 country-years and 2.7 million participants. Lancet 378, 31–40 (2011).

    Article  CAS  PubMed  Google Scholar 

  3. 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 

  4. 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 

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

    Article  CAS  PubMed  Google Scholar 

  6. Lynch, L. et al. Adipose tissue invariant NKT cells protect against diet-induced obesity and metabolic disorder through regulatory cytokine production. Immunity 37, 574–587 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Molofsky, A.B. et al. Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages. J. Exp. Med. 210, 535–549 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Schipper, H.S. et al. Natural killer T cells in adipose tissue prevent insulin resistance. J. Clin. Invest. 122, 3343–3354 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cao, H. Adipocytokines in obesity and metabolic disease. J. Endocrinol. 220, T47–T59 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Shi, H. et al. TLR4 links innate immunity and fatty acid-induced insulin resistance. J. Clin. Invest. 116, 3015–3025 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ye, J., Gao, Z., Yin, J. & He, Q. Hypoxia is a potential risk factor for chronic inflammation and adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. Am. J. Physiol. Endocrinol. Metab. 293, E1118–E1128 (2007).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  14. 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 

  15. Talukdar, S. et al. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nat. Med. 18, 1407–1412 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Nishimura, S. et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat. Med. 15, 914–920 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Borst, S.E. The role of TNF-alpha in insulin resistance. Endocrine 23, 177–182 (2004).

    Article  CAS  PubMed  Google Scholar 

  18. Patsouris, D. et al. Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab. 8, 301–309 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Vivier, E. et al. Innate or adaptive immunity? The example of natural killer cells. Science 331, 44–49 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Shi, F.D., Ljunggren, H.G., La Cava, A. & Van Kaer, L. Organ-specific features of natural killer cells. Nat. Rev. Immunol. 11, 658–671 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Gazit, R. et al. Lethal influenza infection in the absence of the natural killer cell receptor gene Ncr1. Nat. Immunol. 7, 517–523 (2006).

    Article  CAS  PubMed  Google Scholar 

  22. Gur, C. et al. The activating receptor NKp46 is essential for the development of type 1 diabetes. Nat. Immunol. 11, 121–128 (2010).

    Article  CAS  PubMed  Google Scholar 

  23. Mosser, D.M. & Edwards, J.P. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 8, 958–969 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Goldszmid, R.S. et al. NK cell-derived interferon-gamma orchestrates cellular dynamics and the differentiation of monocytes into dendritic cells at the site of infection. Immunity 36, 1047–1059 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Han, M.S. et al. JNK expression by macrophages promotes obesity-induced insulin resistance and inflammation. Science 339, 218–222 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. O'Rourke, R.W. et al. Systemic inflammation and insulin sensitivity in obese IFN-gamma knockout mice. Metabolism 61, 1152–1161 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wong, N. et al. Deficiency in interferon-gamma results in reduced body weight and better glucose tolerance in mice. Endocrinology 152, 3690–3699 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Rocha, V.Z. et al. Interferon-gamma, a Th1 cytokine, regulates fat inflammation: a role for adaptive immunity in obesity. Circ. Res. 103, 467–476 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hayakawa, Y. & Smyth, M.J. CD27 dissects mature NK cells into two subsets with distinct responsiveness and migratory capacity. J. Immunol. 176, 1517–1524 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. Klose, C.S. et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell 157, 340–356 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. 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 

  32. Waggoner, S.N., Cornberg, M., Selin, L.K. & Welsh, R.M. Natural killer cells act as rheostats modulating antiviral T cells. Nature 481, 394–398 (2012).

    Article  CAS  Google Scholar 

  33. Feng, B. et al. Clodronate liposomes improve metabolic profile and reduce visceral adipose macrophage content in diet-induced obese mice. PLoS ONE 6, e24358 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Blunt, T. et al. Defective DNA-dependent protein kinase activity is linked to V(D)J recombination and DNA repair defects associated with the murine scid mutation. Cell 80, 813–823 (1995).

    Article  CAS  PubMed  Google Scholar 

  35. Wong, R.H. et al. A role of DNA-PK for the metabolic gene regulation in response to insulin. Cell 136, 1056–1072 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ballak, D.B. et al. Combined B- and T-cell deficiency does not protect against obesity-induced glucose intolerance and inflammation. Cytokine 62, 96–103 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Amano, S.U. et al. Local proliferation of macrophages contributes to obesity-associated adipose tissue inflammation. Cell Metab. 19, 162–171 (2014).

    Article  CAS  PubMed  Google Scholar 

  38. Zafirova, B., Wensveen, F.M., Gulin, M. & Polic, B. Regulation of immune cell function and differentiation by the NKG2D receptor. Cell. Mol. Life Sci. 68, 3519–3529 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Inzucchi, S.E. et al. Management of hyperglycaemia in type 2 diabetes: a patient-centered approach. Position statement of the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia 55, 1577–1596 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. Johnson, A.R., Milner, J.J. & Makowski, L. The inflammation highway: metabolism accelerates inflammatory traffic in obesity. Immunol. Rev. 249, 218–238 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  42. Houstis, N., Rosen, E.D. & Lander, E.S. Reactive oxygen species have a causal role in multiple forms of insulin resistance. Nature 440, 944–948 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Haus, J.M. et al. Plasma ceramides are elevated in obese subjects with type 2 diabetes and correlate with the severity of insulin resistance. Diabetes 58, 337–343 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Aouadi, M. et al. Gene silencing in adipose tissue macrophages regulates whole-body metabolism in obese mice. Proc. Natl. Acad. Sci. USA 110, 8278–8283 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Tripp, C.S., Wolf, S.F. & Unanue, E.R. Interleukin 12 and tumor necrosis factor alpha are costimulators of interferon gamma production by natural killer cells in severe combined immunodeficiency mice with listeriosis, and interleukin 10 is a physiologic antagonist. Proc. Natl. Acad. Sci. USA 90, 3725–3729 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Bryceson, Y.T., March, M.E., Ljunggren, H.G. & Long, E.O. Synergy among receptors on resting NK cells for the activation of natural cytotoxicity and cytokine secretion. Blood 107, 159–166 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ouchi, N., Parker, J.L., Lugus, J.J. & Walsh, K. Adipokines in inflammation and metabolic disease. Nat. Rev. Immunol. 11, 85–97 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Wang, Q.A., Tao, C., Gupta, R.K. & Scherer, P.E. Tracking adipogenesis during white adipose tissue development, expansion and regeneration. Nat. Med. 19, 1338–1344 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Huebner, L. et al. Human NK cell subset functions are differentially affected by adipokines. PLoS ONE 8, e75703 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Polić, B. et al. Hierarchical and redundant lymphocyte subset control precludes cytomegalovirus replication during latent infection. J. Exp. Med. 188, 1047–1054 (1998).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Slavić-Stupac, M. Samsa and K. Miklić for technical assistance and D. Jurišić-Eržen and M. Zelić for help with study setup. Supported by the European Foundation for the Study of Diabetes (New Horizons Program), the Unity through Knowledge Fund (15/13 to B.P.), the University of Rijeka (13.06.1.1.03 to B.P.), the Netherlands Organization for Scientific Research (91614029 to F.M.W.) and the European Commission (PCIG14-GA-2013-630827 to F.M.W.).

Author information

Authors and Affiliations

Authors

Contributions

F.M.W. designed and carried out most of the experiments and analyzed data. V.J., S.V., M.Š., T.T.W. and V.S. performed and analyzed experiments. A.G. generated key research reagents. D.M. obtained informed consent from patients and obtained human samples. D.Š. supervised the setup of the clinical study. F.T.W. and J.C.B. coordinated the metabolic studies. O.M. helped design studies with NCRGFP mice. B.P. directed the research and wrote the paper with F.M.W., with input from all coauthors.

Corresponding author

Correspondence to Bojan Polić.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Phenotype and function of VAT-resident NK cells.

(a) VAT leukocytes were stimulated in vitro with the indicated stimuli, and IFN-γ production was assessed in NK cells and macrophages (n = 3). (b) VAT leukocytes were stimulated in vitro with PMA and ionomycin, and IFN-γ production was assessed (n = 3). (c) Splenic and VAT-resident NK cells were isolated, and the relative number of cells expressing the indicated markers was determined by flow cytometry. Gated for CD3NK1.1+NCR1+ cells, except for the markers NK1.1 (CD3NCR1+) and NCR1 (CD3NK1.1+). Representative plots are shown of one of seven independent experiments, with three to five mice used per experiment. (d) Total splenocytes or VAT-derived leukocytes were stimulated with PMA and ionomycin, and the expression of CD107a on NK cells was determined by flow cytometry (n = 5). (e) The relative number of NK cells in total splenocytes or VAT-derived NK cells was determined and cells were mixed in the indicated ratio with CFSE-labeled target cells. After 5 h, effector cell–induced cell death (Topro3+) of target cells was determined by flow cytometry (n = 5). (f) Splenic and VAT-resident NK cells were stimulated in vitro with IL-15 or agonistic NCR1 antibodies, and expression of Ki67 was analyzed after 48 h. Gated for CD3NK1.1+ cells (n = 3). (g) Kinetics of the absolute number of γδ T cells in VAT after 4 weeks of NCD or HFD feeding (n = 5). (h) Kinetics of the absolute number of the indicated immune cell subsets in the spleens of mice after 12 weeks of NCD or HFD feeding (n = 5). (i) Relative number of regulatory T cells (CD3+CD4+FoxP3+) in VAT after 12 weeks of NCD or HFD feeding (n = 5). Shown are data from one of two to seven experiments with similar results (mean ± s.e.m., n = 4–5, *P < 0.05, **P < 0.01, ***P < 0.001). FMO, fluorescence minus one.

Supplementary Figure 2 VAT-resident NK cells are activated upon HFD feeding and promote M1 macrophage formation.

(a) Mice were fed an NCD or an HFD for 4 weeks, after which VAT leukocytes were stimulated in vitro with PMA and ionomycin and cytokine production was assessed in NK cells and CD4+ TH cells (n = 5). (b) Mice were fed an NCD or an HFD and received PBS or α-NK1.1 antibodies once every 5 d. After 12 weeks, numbers of NK (CD3NK1.1+NCR1+), NKT (CD3+NK1.1+NCR1) and CD8+ T cells (CD8+CD3+) were quantified (n = 4–5). (c) Gating strategy for adipose tissue macrophages and NK cells. M1 macrophages (M1) are defined as CD19CD11b+F4/80+GR1–/DimCD11c+CD86Dim. M2 macrophages (M2) are defined as CD19CD11b+F4/80+GR1–/DimCD11cCD86+. NK cells are defined as CD3NK1.1+NCR1+. (d) Macrophage subsets (as defined in c) were analyzed for the expression of M1 and M2 markers under homeostatic conditions (arginase, IL-4Rα) and after 2 d of in vitro stimulation with LPS (iNOS, n = 3). (e) Mice were fed an NCD or an HFD and received PBS or α-NK1.1 antibodies once every 5 d. After 6 weeks, the ratio between M1 and M2 macrophages in VAT was determined (n = 4–5). (f,g) Mice were HFD-fed for 2 weeks and injected with PBS or neutralizing α-IFN-γ antibodies once every other day (n = 5). (f) Ratio between M1 and M2 macrophages in VAT. (g) Phenotype of NK cells in VAT. (hk) Mice were fed an NCD or an HFD for 12 weeks and received depleting α-NK1.1 or isotype control antibodies every 5 d. (h) Mice were fasted overnight and injected i.p. with either PBS (–) or 1.0 U/kg insulin (+). Immunoblot was used to quantify phosphorylated p70 (Thr389) in tissue samples of liver and VAT. β-Actin was used as a loading control. (i) Quantification shows pooled data from two independent experiments (n = 6). (j) Body weight and (k) visceral fat pad weight of indicated groups after 12 weeks of feeding (n = 5). Shown are data from one of two to five experiments with similar results (mean + s.e.m., n = 5, *P < 0.05, **P < 0.01, ***P < 0.001).

Supplementary Figure 3 NK cells do not require the adaptive immune system to promote HFD-induced M1 macrophage formation in VAT.

(a,b) Mice underwent either sham operation or removal of abdominal fat (VATectomy; n = 5). (a) 12 weeks after surgery, mice were subjected to GTT. (b) Weight of subcutaneous fat pads. (c) Wild-type and Prkdcscid/scid (SCID) mice were fed an NCD or an HFD for 12 weeks and subjected to GTT. Shown are plasma glucose levels and AUC (n = 3–5). (d,e) NCD- or HFD-fed SCID mice were injected with α-NK1.1 antibodies or PBS once every 5 d. After 14 weeks, (d) the phenotype of NK cells and (n = 4–5) (e) the number of total and M1 macrophages in VAT were determined (n = 3–5). (f,g) SCID mice were fed an NCD or an HFD for 12 weeks. Mice received depleting α-NK1.1 or isotype control antibodies every 5 d. After 12 weeks the (f) body weight and (g) visceral fat pad weight of mice were determined (n = 4–5). Shown are data from one of two experiments with similar results (mean ± s.e.m., n = 3–5, *P < 0.05, **P < 0.01). AUC, area under the curve.

Supplementary Figure 4 HFD promotes proliferation of NK cells in VAT.

(a) Fraction of leukocytes as a percentage of the stromal vascular fraction (SVF) in spleen, brown adipose tissue (BAT), VAT and subcutaneous (SC) fat (n = 14). (b) Fraction of NK cells as a percentage of leukocytes in indicated organs under homeostatic conditions (n = 14). (c) Mice were HFD-fed for 4 weeks and subsequently injected with 2 mg BrdU. BrdU incorporation in VAT NK cells was determined after 4 h (n = 5). Shown are pooled data from three independent experiments (a,b) or one experiment (c).

Supplementary Figure 5 VAT-resident NK cells are activated by stress ligands in adipose tissue upon HFD feeding.

(a) Wild-type mice were fed an NCD or an HFD for 4 weeks. VAT sections were stained with mNCR1-immunoglobulin or mNKG2D-immunoglobulin fusion proteins and visualized with DAB. Irrelevant fusion proteins were used as negative controls. As a positive control for NCR1-L staining, 3T3-L1 cells were included. Arrow shows fat capsule. Scale bar, 100 μm. (b) Concentration-dependent IFN-γ production in NK cells after in vitro NCR1 stimulation (n = 3). (c) Splenic NK cells from Ncr1gfp/gfp mice were stimulated in vitro with plate-bound NK1.1 or NCR1 antibodies, and IFN-γ production was assessed (n = 3–4). (d) The SVF of VAT was stimulated for 48 h with agonistic NCR1 antibodies in the presence or absence of neutralizing IFN-γ antibodies. After 48 h, the fraction of iNOS+ and CD11c+ macrophages was determined (n = 3). (e) NK cells were activated in vitro for 48 h with agonistic NCR1 antibodies in the presence or absence of neutralizing IFN-γ antibodies. Subsequently, supernatants of these cells were taken and used to stimulate macrophages. After 48 h, iNOS production in macrophages was assessed (n = 3). Shown are data from one of two to four experiments with similar results (mean ± s.e.m., n = 5, *P < 0.05, **P < 0.01, ***P < 0.001). n.d., not determined.

Supplementary Figure 6 NCR1 deficiency or NK cell depletion reduces HFD-induced insulin resistance but does not affect weight gain.

(ac) Mice were fed an NCD or an HFD and received PBS or α-NK1.1 antibodies once every 5 d (n = 4–5). (a) After 5 weeks, the number of M1 macrophages was determined in VAT. (b,c) After 12 weeks, (b) total body weight and (c) visceral adipose fat pad weight were determined. (d) Mice were fasted overnight and injected i.p. with either PBS (–) or 1.0 U/kg insulin (+). Immunoblot was used to quantify p-Akt (Ser473), p-GSK3α (Ser21/9), p-p70 (Thr389), pERK1/2 (Thr202/Tyr204) and total ERK1/2 in tissue samples of liver and VAT. Calnexin was used as a loading control (n = 2–3). Shown are data from one of two experiments with similar results (mean ± s.e.m., n = 4–5, *P < 0.05, **P < 0.01, ***P < 0.001).

Supplementary Figure 7 Therapeutic blocking of NCR1 prevents M1 macrophage formation.

(a) HFD-fed Ifng–/– mice (CD45.2) received PBS, wild-type (CD45.1+) or NCR1GFP NK cells (4 × 105) every 4 d. Shown are representative plots of the experiment shown in Figure 7a,b. Donor cells were identified based on their expression of CD45.1 or GFP. (b) Indicated groups were fed an NCD or an HFD for 12 weeks and subjected to GTT (n = 3–5). (c,d) HFD-fed Ifng–/– mice received PBS or wild-type GFP+ NK cells (5 × 105) once every 5 d and were compared with PBS-injected, NCD-fed animals. After 2 weeks, the phenotypes of (c) donor and recipient NK cells and (d) macrophages in VAT were analyzed (n = 3–4). (e) NK cells were stimulated in vitro with plate-bound α-NCR1, in the presence or absence of the same clone of α-NCR1 antibodies soluble in the medium. (f) Wild-type NK cells, NCR1 NK cells, BW cells and BW cells transgenically expressing NCR1ζ were stained with the antibodies mNCR15 and mNCR16 to demonstrate the specificity of these reagents for NCR1. (g) Six cell lines that are known to express NCR1 ligands were stained with Ncr1-immunoglobulin alone or in combination with NCR1-specific antibodies. NCR1-specific antibodies blocked binding of NCR1-immunoglobulin to target cell lines, indicating that this reagent shows target binding that is identical to that of NCR1. (h) NK cells were stimulated with PMA and ionomycin or agonistic Ncr1 antibodies in the presence or absence (control) of Ncr1-immunoglobulin. NCR1-immunoglobulin is able to reduce NK cell activation upon NCR1 stimulation, again demonstrating the specificity of NCR1-immunoglobulin fusion protein for NCR1 ligands. (ik) Mice were fed an NCD or an HFD for 2 weeks. Animals received PBS or NCR1 ligand–blocking NCR1-immunoglobulin fusion proteins twice per week. Ncr1GFP/GFP mice are genetically deficient for NCR1. M1 macrophage and NK cell numbers were analyzed after 2 weeks (n = 5). (i) NK cell numbers and (j) M1 macrophage numbers in VAT. (k) M1/M2 ratio in VAT showed that NCR1 deficiency or its blocking prevented the absolute (Fig. 7e) and relative increase of M1 cells in VAT. (l) Fat pad weight of mice after 12 weeks of feeding (n = 5). Shown are data from one of one to two experiments with similar results (mean ± s.e.m., n = 5, *P < 0.05, **P < 0.01, **P < 0.001).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 692 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wensveen, F., Jelenčić, V., Valentić, S. et al. NK cells link obesity-induced adipose stress to inflammation and insulin resistance. Nat Immunol 16, 376–385 (2015). https://doi.org/10.1038/ni.3120

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3120

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