The role of the immune system in obesity and insulin resistance

Payal S Patel, Eric D Buras, Ashok Balasubramanyam, Payal S Patel, Eric D Buras, Ashok Balasubramanyam

Abstract

The innate immune system provides organisms with rapid and well-coordinated protection from foreign pathogens. However, under certain conditions of metabolic dysfunction, components of the innate immune system may be activated in the absence of external pathogens, leading to pathologic consequences. Indeed, there appears to be an intimate relationship between metabolic diseases and immune dysfunction; for example, macrophages are prime players in the initiation of a chronic inflammatory state in obesity which leads to insulin resistance. In response to increases in free fatty acid release from obese adipose depots, M1-polarized macrophages infiltrate adipose tissues. These M1 macrophages trigger inflammatory signaling and stress responses within cells that signal through JNK or IKK β pathways, leading to insulin resistance. If overnutrition persists, mechanisms that counteract inflammation (such as M2 macrophages and PPAR signaling) are suppressed, and the inflammation becomes chronic. Although macrophages are a principal constituent of obese adipose tissue inflammation, other components of the immune system such as lymphocytes and mast cells also contribute to the inflammatory cascade. Thus it is not merely an increased mass of adipose tissue that directly leads to attenuation of insulin action, but rather adipose tissue inflammation activated by the immune system in obese individuals that leads to insulin resistance.

Figures

Figure 1
Figure 1
Role of the immune system in lean versus obese adipose tissue. In lean adipose tissue, T-helper type 2 (TH2) cells produce anti-inflammatory cytokines such as interleukin (IL)-4, 10, and 13 which promote alternatively activated M2 macrophage polarization. M2 polarization is also induced by regulatory T cells (Tregs) and eosinophils via IL-4. M2 macrophages secrete other anti-inflammatory signals such as IL-10 which maintain insulin sensitivity within lean adipose tissue. Conversely, TH1 type cytokines such as interferon (IFN)-γ stimulate M1 macrophage polarization in obese adipose tissue. Other immune cells are also increased in obese adipose tissue which contribute to insulin resistance including mast cells, B cells, and immunoglobulins (Igs). CD8(+) T cells promote ATM accumulation and proinflammatory gene expression and are also increased as well. Macrophages are not homogenously distributed throughout obese adipose tissue but rather aggregated around dead adipocytes forming crown-like structures (CLS). M1 macrophages are proinflammatory, secreting cytokines such as TNF-α and IL-1β. Macrophages are bone-marrow-derived myeloid cells hence both M1 and M2 macrophages express the myeloid cell surface markers F4/80 and CD11b. However, only the M1 population expresses the marker CD11c.
Figure 2
Figure 2
Various signaling pathways promoting or inhibiting inflammatory signaling (green arrows represent activation, and red arrows represent inhibition). Jun N-terminal kinase-1 (JNK1) and inhibitor of kB kinase (IKKβ) are inflammatory signaling pathways which promote insulin resistance. Activation of either pathway leads to serine phosphorylation of insulin receptor substrate protein-1 (IRS-1), causing attenuation of insulin action. IKKβ also phosphorylates inhibitor of nuclear factor-κB (IκB) which essentially frees nuclear factor-κB (NF-κB), allowing it to translocate to the nucleus, bind to DNA, and activate inflammatory mediators. JNK1 can also stimulate transcription of inflammatory genes in association with transcription factor activator protein 1 (AP1). Toll-like receptor 4 (TLR4) activation, which normally binds lipopolysaccharides (LPS) and saturated fatty acids (FA), results in activation of JNK and IKKβ. Endoplasmic reticulum (ER) stress, stimulated by FA, nutrient excess, and microhypoxia, leads to the unfolded protein response (UPR). UPR comprises three main pathways: inositol-requiring enzyme (IRE)-1, protein kinase-like ER kinase (PERK), and activating transcription factor (ATF)α which all lead to activation of JNK1 and IKKβ. Hypoxia also activates the transcription factor hypoxia-inducible factor-1α (HIF-1α), which induces expression of various target genes. Conversely, insulin sensitivity is promoted by activation of the omega-3 fatty acid receptor (GRP120) which inhibits JNK1 and IKKβ. PPARγ also promotes insulin sensitivity by interfering with the NF-κB and AP1 signaling pathways and subsequent expression of inflammatory genes.

References

    1. Flegal KM, Carroll MD, Ogden CL, Johnson CL. Prevalence and trends in obesity among US adults, 1999-2000. Journal of the American Medical Association. 2002;288(14):1723–1727.
    1. Xu H, Barnes GT, Yang Q, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. Journal of Clinical Investigation. 2003;112(12):1821–1830.
    1. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW. Obesity is associated with macrophage accumulation in adipose tissue. Journal of Clinical Investigation. 2003;112(12):1796–1808.
    1. Altintas MM, Azad A, Nayer B, et al. Mast cells, macrophages, and crown-like structures distinguish subcutaneous from visceral fat in mice. Journal of Lipid Research. 2011;52(3):480–488.
    1. Cancello R, Tordjman J, Poitou C, et al. Increased infiltration of macrophages in omental adipose tissue is associated with marked hepatic lesions in morbid human obesity. Diabetes. 2006;55(6):1554–1561.
    1. Kolak M, Westerbacka J, Velagapudi VR, et al. Adipose tissue inflammation and increased ceramide content characterize subjects with high liver fat content independent of obesity. Diabetes. 2007;56(8):1960–1968.
    1. Gastaldelli A, Miyazaki Y, Pettiti M, et al. Metabolic effects of visceral fat accumulation in type 2 diabetes. Journal of Clinical Endocrinology and Metabolism. 2002;87(11):5098–5103.
    1. Kosteli A, Sugaru E, Haemmerle G, et al. Weight loss and lipolysis promote a dynamic immune response in murine adipose tissue. Journal of Clinical Investigation. 2010;120(10):3466–3479.
    1. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annual Review of Physiology. 2009;72:219–246.
    1. Wang P, Mariman E, Renes J, Keijer J. The secretory function of adipocytes in the physiology of white adipose tissue. Journal of Cellular Physiology. 2008;216(1):3–13.
    1. Weisberg SP, Hunter D, Huber R, et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. Journal of Clinical Investigation. 2006;116(1):115–124.
    1. Oh DY, Morinaga H, Talukdar S, et al. Increased macrophage migration into adipose tissue in obese mice. Diabetes. 2012;61(2):346–354.
    1. Kanda H, Tateya S, Tamori Y, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. Journal of Clinical Investigation. 2006;116(6):1494–1505.
    1. Féral CC, Neels JG, Kummer C, Slepak M, Olefsky JM, Ginsberg MH. Blockade of α4 integrin signaling ameliorates the metabolic consequences of high-fat diet-induced obesity. Diabetes. 2008;57(7):1842–1851.
    1. Chang L, Chiang SH, Saltiel AR. Insulin signaling and the regulation of glucose transport. Molecular Medicine. 2004;10(7–12):65–71.
    1. Lesniewski LA, Hosch SE, Neels JG, et al. Bone marrow-specific Cap gene deletion protects against high-fat diet-induced insulin resistance. Nature Medicine. 2007;13(4):455–462.
    1. Chakrabarti SK, Wen Y, Dobrian AD, et al. Evidence for activation of inflammatory lipoxygenase pathways in visceral adipose tissue of obese Zucker rats. American Journal of Physiology. 2011;300(1):E175–E187.
    1. Spite M, Hellmann J, Tang Y, et al. Deficiency of the leukotriene B4 receptor, BLT-1 protects against systemic insulin resistance in diet-induced obesity. The Journal of Immunology. 2011;187(4):1942–1949.
    1. Kitade H, Sawamoto K, Nagashimada M, et al. CCR5 plays a critical role in obesity-induced adipose tissue inflammation and insulin resistance by regulating both macrophage recruitment and M1/M2 status. Diabetes. 2012;61(7):1680–1690.
    1. Goerdt S, Politz O, Schledzewski K, et al. Alternative versus classical activation of macrophages. Pathobiology. 2000;67(5-6):222–226.
    1. Gordon S. Alternative activation of macrophages. Nature Reviews Immunology. 2003;3(1):23–35.
    1. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. Journal of Clinical Investigation. 2007;117(1):175–184.
    1. Lumeng CN, Delproposto JB, Westcott DJ, Saltiel AR. Phenotypic switching of adipose tissue macrophages with obesity is generated by spatiotemporal differences in macrophage subtypes. Diabetes. 2008;57(12):3239–3246.
    1. Lumeng CN, DeYoung SM, Bodzin JL, Saltiel AR. Increased inflammatory properties of adipose tissue macrophages recruited during diet-induced obesity. Diabetes. 2007;56(1):16–23.
    1. Nguyen MTA, Favelyukis S, Nguyen AK, 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. Journal of Biological Chemistry. 2007;282(48):35279–35292.
    1. Patsouris D, Li PP, Thapar D, Chapman J, Olefsky JM, Neels JG. Ablation of CD11c-Positive Cells Normalizes Insulin Sensitivity in Obese Insulin Resistant Animals. Cell Metabolism. 2008;8(4):301–309.
    1. Li P, Lu M, Nguyen MT, et al. Functional heterogeneity of CD11c-positive adipose tissue macrophages in diet-induced obese mice. The Journal of Biological Chemistry. 2010;285(20):15333–15345.
    1. Fujisaka S, Usui I, Bukhari A, et al. Regulatory mechanisms for adipose tissue M1 and M2 macrophages in diet-induced obese mice. Diabetes. 2009;58(11):2574–2582.
    1. Shaul ME, Bennett G, Strissel KJ, Greenberg AS, Obin MS. Dynamic, M2-like remodeling phenotypes of CD11c+ adipose tissue macrophages during high-fat diet—induced obesity in mice. Diabetes. 2010;59(5):1171–1181.
    1. Strissel KJ, Stancheva Z, Miyoshi H, et al. Adipocyte death, adipose tissue remodeling, and obesity complications. Diabetes. 2007;56(12):2910–2918.
    1. Kabon B, Nagele A, Reddy D, et al. Obesity decreases perioperative tissue oxygenation. Anesthesiology. 2004;100(2):274–280.
    1. Jantsch J, Chakravortty D, Turza N, et al. Hypoxia and hypoxia-inducible factor-1α modulate lipopolysaccharide-induced dendritic cell activation and function. Journal of Immunology. 2008;180(7):4697–4705.
    1. Jiang C, Qu A, Matsubara T, et al. Disruption of hypoxia-inducible factor 1 in adipocytes improves insulin sensitivity and decreases adiposity in high-fat diet-fed mice. Diabetes. 2011;60(10):2484–2495.
    1. Greenberg AS, Egan JJ, Wek SA, Garty NB, Blanchette-Mackie EJ, Londos C. Perilipin, a major hormonally regulated adipocyte-specific phosphoprotein associated with the periphery of lipid storage droplets. Journal of Biological Chemistry. 1991;266(17):11341–11346.
    1. Cinti S, Mitchell G, Barbatelli G, et al. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. Journal of Lipid Research. 2005;46(11):2347–2355.
    1. Murano I, Barbatelli G, Parisani V, et al. Dead adipocytes, detected as crown-like structures, are prevalent in visceral fat depots of genetically obese mice. Journal of Lipid Research. 2008;49(7):1562–1568.
    1. Apovian CM, Bigornia S, Mott M, et al. Adipose macrophage infiltration is associated with insulin resistance and vascular endothelial dysfunction in obese subjects. Arteriosclerosis, Thrombosis, and Vascular Biology. 2008;28(9):1654–1659.
    1. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-α in human obesity and insulin resistance. Journal of Clinical Investigation. 1995;95(5):2409–2415.
    1. Gao Z, Hwang J, Bataille F, et al. Serine phosphorylation of insulin receptor substrate 1 by inhibitor κB kinase complex. The Journal of Biological Chemistry. 2002;277(50):48115–48121.
    1. Hirosumi J, Tuncman G, Chang L, et al. A central role for JNK in obesity and insulin resistance. Nature. 2002;420(6913):333–336.
    1. Vallabhapurapu S, Karin M. Regulation and function of NF-κB transcription factors in the immune system. Annual Review of Immunology. 2009;27:693–733.
    1. Arkan MC, Hevener AL, Greten FR, et al. IKK-β links inflammation to obesity-induced insulin resistance. Nature Medicine. 2005;11(2):191–198.
    1. Solinas G, Karin M. JNK1 and IKKβ: molecular links between obesity and metabolic dysfunction. FASEB Journal. 2010;24(8):2596–2611.
    1. Solinas G, Vilcu C, Neels JG, et al. JNK1 in hematopoietically derived cells contributes to diet-induced inflammation and insulin resistance without affecting obesity. Cell Metabolism. 2007;6(5):386–397.
    1. Lee JY, Sohn KH, Rhee SH, Hwang D. Saturated fatty acids, but not unsaturated fatty acids, induce the expression of cyclooxygenase-2 mediated through toll-like receptor 4. Journal of Biological Chemistry. 2001;276(20):16683–16689.
    1. Tsukumo DML, Carvalho-Filho MA, Carvalheira JBC, et al. Loss-of-function mutation in toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes. 2007;56(8):1986–1998.
    1. Koliwad SK, Streeper RS, Monetti M, et al. DGAT1-dependent triacylglycerol storage by macrophages protects mice from diet-induced insulin resistance and inflammation. Journal of Clinical Investigation. 2010;120(3):756–767.
    1. Vandanmagsar B, Youm YH, Ravussin A, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nature Medicine. 2011;17(2):179–189.
    1. Saltiel AR. Fishing out a sensor for anti-inflammatory oils. Cell. 2010;142(5):672–674.
    1. Hummasti S, Hotamisligil GS. Endoplasmic reticulum stress and inflammation in obesity and diabetes. Circulation Research. 2010;107(5):579–591.
    1. Bocher V, Chinetti G, Fruchart JC, Staels B. Role of the peroxisome proliferator-activated receptors (PPARS) in the regulation of lipids and inflammation control. Journal de la Societe de Biologie. 2002;196(1):47–52.
    1. Hevener AL, Olefsky JM, Reichart D, et al. Macrophage PPARγ is required for normal skeletal muscle and hepatic insulin sensitivity and full antidiabetic effects of thiazolidinediones. Journal of Clinical Investigation. 2007;117(6):1658–1669.
    1. Kang K, Reilly SM, Karabacak V, et al. Adipocyte-derived Th2 cytokines and myeloid PPARδ regulate macrophage polarization and insulin sensitivity. Cell Metabolism. 2008;7(6):485–495.
    1. 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. Nature Medicine. 2009;15(8):930–939.
    1. Nishimura S, Manabe I, Nagasaki M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nature Medicine. 2009;15(8):914–920.
    1. Winer S, Chan Y, Paltser G, et al. Normalization of obesity-associated insulin resistance through immunotherapy. Nature Medicine. 2009;15(8):921–929.
    1. Winer DA, Winer S, Shen L, et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nature Medicine. 2011;17(5):610–617.
    1. Liu J, Divoux A, Sun J, et al. Genetic deficiency and pharmacological stabilization of mast cells reduce diet-induced obesity and diabetes in mice. Nature Medicine. 2009;15(8):940–945.
    1. Wu D, Molofsky AB, Liang HE, et al. Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis. Science. 2011;332(6026):243–247.

Source: PubMed

3
Sottoscrivi