LACK OF ST2 ENHANCES HIGH-FAT DIET-INDUCED VISCERAL ADIPOSITY AND INFLAMMATION IN BALB/c MICE

  • Jelena Pantic Faculty of Medical Sciences University of Kragujevac
  • Nada Pejnovic Faculty of Medical Sciences University of Kragujevac
  • Gordana Radosavljevic Faculty of Medical Sciences University of Kragujevac
  • Ivan Jovanovic Faculty of Medical Sciences University of Kragujevac
  • Aleksandar Djukic Faculty of Medical Sciences University of Kragujevac
  • Nebojsa Arsenijevic Faculty of Medical Sciences University of Kragujevac
  • Miodrag Lukic Faculty of Medical Sciences University of Kragujevac

Abstract


ABSTRACT

Obesity and obesity related disorders are strongly associated with chronic low-grade inflammation originating from growing visceral adipose tissue during nutrient excess. Although interleukin (IL)-33 may have protective role in obesity and atherosclerosis, the impact of IL-33/ST2 axis in metabolic disorders needs to be elucidated.

In this study, we investigated the role of IL-33/ST2 pathway in high-fat diet (HFD)-induced obesity using ST2-deficient (ST2-/-) and wild type mice on BALB/c background.

The deletion of ST2 enhanced systemic and visceral adipose tissue (VAT) inflammation and was associated with significantly higher weight gain and amount of total VAT in ST2-/- mice fed with HFD for 18 weeks. More numerous classically activated M1 macrophages and markedly decreased alternatively activated M2 macrophages were observed in VAT of HFD-fed ST2-/- mice. Additionally, VAT of ST2-/- mice fed with HFD had increased percentage of CD3+ T cells with lower incidence of CD4+CD25+FoxP3+ T regulatory cells in comparison with low-fat diet fed controls. The incidence of CD3+IL-17+ and IL-5 positive VAT-derived mononuclear cells was significantly lower in HFD-fed ST2-/- mice. Serum levels of proinflammatory cytokines IL-1β and IFN-γ were increased in HFD-fed ST2-/- mice, while the levels of IL-6 and CRP did not differ among groups. Importantly, the levels of anti-inflammatory IL-10 and IL-13 were significantly lower in the sera of ST2-/- mice compared with wild-type controls.

Our findings suggest that protective role of IL33/ST2 signaling in high-fat diet-induced adipose tissue inflammation. ST2 deficiency related to nutrient excess is associated with polarization of macrophages toward M1 phenotype and the induction Th1-mediated immune response.

Key words: ST2, obesity, high-fat diet, visceral adipose tissue, inflammation

References

Hotamisligil GS. Inflammation and metabolic disorders. Nature 2006;444:860-867.

Lumeng CN, Saltiel AR. Inflammatory links between obesity and metabolic disease. J Clin Invest 2011;121:2111-2117.

Strissel KJ, DeFuria J, Shaul ME, Bennett G, Greenberg AS, Obin MS. T-cell recruitment and Th1 polarization in adipose tissue during diet-induced obesity in C57BL/6 mice. Obesity (Silver Spring) 2010; 18:1918-25.

Ohmura K, Ishimori N, Ohmura Y, Tokuhara S, Nozawa A, Horii S, et al. Natural killer T cells are involved in adipose tissues inflammation and glucose intolerance in diet-induced obese mice. Arterioscler Thromb Vasc Biol 2010; 30:193-9.

Pejnovic N, Pantic J, Jovanovic I, Radosavljevic G, Milovanovic M, Nikolic I, et al. Galectin-3 deficiency accelerates high-fat diet induced obesity and amplifies inflammation in adipose tissue and pancreatic islets. Diabetes 2013;62:1932-44.

Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 2007;117:175-184.

Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, Chen H. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest 2003;112(12):1821–1830.

Feuerer M, Herrero L, Cipolletta D, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med 2009;15:930–939.

Liew FY, Pitman NI and McInnes IB. Disease-associated function of IL-33: the new kid in the IL-1 family. Nat Rev Immunol 2010; 10: 103-10.

Chackerian AA, Oldham ER, Murphy EE, Schmitz J, Pflanz S and Kastelein RA. IL-1 receptor accessory protein and ST2 comprise the IL-33 receptor complex. J Immunol 2007; 179: 2551-55.

Schmitz J, Owyang A, Oldham E, Song Y, Murphy E, McClanahan TK, et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity 2005; 23: 479-90.

Bourgeois E, Van LP, Samson M, Diem S, Barra A, Roga S, et al. The pro-Th2 cytokine IL-33 directly interacts with invariant NKT and NK cells to induce IFN-γ production. Eur J Immunol 2009; 39: 1046-55.

Kurowska-Stolarska M, Stolarski B, Kewin P, Murphy G, Corrigan CJ, Ying S, et al. IL-33 amplifies the polarization of alternatively activated macrophages that contribute to airway inflammation. J Immunol 2009; 183: 6469-77.

Volarevic V, Mitrovic M, Milovanovic M, Zelen I, Nikolic I, Mitrovic S, et al. Protective role of IL-33/ST2 axis in Con A-induced hepatitis. J Hepatol 2012; 56: 26-33.

Milovanovic M, Volarevic V, Ljujic B, Radosavljevic G, Jovanovic I, Arsenijevic N, Lukic ML. Deletion of IL-33R (ST2) Abrogates Resistance to EAE in BALB/C Mice by Enhancing Polarization of APC to Inflammatory Phenotype. PLoS One 2012; 7 (9): e45225.

Zdravkovic N, Shahin A, Arsenijevic N, Lukic ML, Mensah-Brown EP. Regulatory T cells and ST2 signaling control diabetes induction with multiple low doses of streptozotocin. Mol Immunol. 2009 Nov;47(1):28-36.

Jovanovic I, Radosavljevic G, Mitrovic M, Lisnic Juranic V, McKenzie ANJ, Arsenijevic N, et al. ST2 Deletion Enhances Innate and Acquired Immunity to Murine Mammary Carcinoma. Eur J Immunol 2011; 41: 1902-12.

Miller AM, Asquith DL, Hueber AJ, Anderson LA, Holmes WM, McKenzie AN, et al. Interleukin-33 Induces Protective Effects in Adipose Tissue Inflammation During Obesity in Mice. Circ Res 2010; 107: 650-58.

Miller AM, Xu D, Asquith DL, Denby L, Li Y, Sattar N, et al. IL-33 reduces the development of atherosclerosis. J Exp Med 2008; 205: 339-46.

Kintscher U, Hartge M, Hess K, Foryst-Ludwig A, Clemenz M, et al. T-lymphocyte infiltration in visceral adipose tissue: a primary event in adipose tissue inflammation and the development of obesity-mediated insulin resistance. Arterioscler Thromb Vasc Biol 2008;28:1304-1310.

Kang K, Reilly SM, Karabacak V, Gangl MR, Fitzgerald K, Hatano B, Lee CH. Adipocyte-derived Th2 cytokines and myeloid PPARdelta regulate macrophage polarization and insulin sensitivity. Cell Metab. 2008;7:485– 495.

Zuniga LA, Shen WJ, Joyce-Shaikh B, Pyatnova EA, Richards AG, Thom C, Andrade SM, Cua DJ, Kraemer FB, Butcher EC. IL-17 Regulates Adipogenesis, Glucose Homeostasis, and Obesity. J Immunol 2010;185:6947-6959.

Zorzanelli Rocha V, Folco EJ, Sukhova G, Shimizu K, Gotsman I, Vernon AH, Libby P. Interferon-γ, a Th1 Cytokine, Regulates Fat Inflammation A Role for Adaptive Immunity in Obesity. Circ Res 2008;103:467-476.

Ilan Y, Maron R, Tukpah AM, et al. Induction of regulatory T cells decreases adipose inflammation and alleviates insulin resistance in ob/ob mice. Proc Natl Acad Sci USA 2010;107:9765–9770.

Nguyen MTA, Favelyukis S, Nguyen A-K, et al. A subpopulation of macrophages infiltrates hypertrophic adipose tissue and is activated by free fatty acids via Toll-like receptors 2 and 4 and JNK-dependent pathways. J Biol Chem 2007;282:35279–35292.

Fujisaka S, Usui I, Bukhari A, Ikutani M, et al. Regulatory mechanisms for adipose tissue M1 and M2 macrophages in diet-induced obese mice. Diabetes 2009; 58:2574-2582.

Hong EG, Ko HJ, Cho YR, et al. Interleukin-10 prevents diet-induced insulin resistance by attenuating macrophage and cytokine response in skeletal muscle. Diabetes 2009;58:2525–2535.

Stanya KJ, Jacobi D, Liu S, Bhargava P, Dai L, Gangl MR, et al. Direct control of hepatic glucose production by interleukin-13 in mice. J Clin Invest 2013; 123:261-71.

Published
2014/02/26
Section
Original Scientific Paper