T Cells in Adipose Tissue: Critical Players in Immunometabolism

Qun Wang, Huaizhu Wu, Qun Wang, Huaizhu Wu

Abstract

Adipose tissue performs immunoregulatory functions in addition to fat storage. Various T cells in different fat depots either help maintain metabolic homeostasis under healthy conditions or contribute to metabolic disorders in pathological conditions such as obesity, diabetes, cardiovascular diseases, or even cancer. These T cells play critical roles in immunometabolism, which refers to the intersection of immunity and metabolism. Numerous studies have examined the presence and changes of different T cell subsets, including helper T cells, regulatory T cells, cytotoxic T cells, and natural killer T cells, in adipose depots in health and diseases. In this review, we will discuss the adipose tissue niches that influence the patterns and functions of T cell subsets and in turn the impact of these T cells on cell- or body-based immunometabolism accounting for health and obesity.

Keywords: T cell; adipose tissue; immunometabolism; insulin resistance; obesity.

Figures

Figure 1
Figure 1
T cell subsets in different adipose niches in lean and obese conditions.
Figure 2
Figure 2
Changes of T cell subsets in adipose tissue in obesity.

References

    1. Man K, Kutyavin VI, Chawla A. Tissue immunometabolism: development, physiology, and pathobiology. Cell Metab. (2017) 25:11–26. 10.1016/j.cmet.2016.08.016
    1. O'Neill LA, Kishton RJ, Rathmell J. A guide to immunometabolism for immunologists. Nat Rev Immunol. (2016) 16:553–65. 10.1038/nri.2016.70
    1. Mathis D, Shoelson SE. Immunometabolism: an emerging frontier. Nat Rev Immunol. (2011) 11:81. 10.1038/nri2922
    1. Kohlgruber AC, LaMarche NM, Lynch L. Adipose tissue at the nexus of systemic and cellular immunometabolism. Semin Immunol. (2016) 28:431–40. 10.1016/j.smim.2016.09.005
    1. Wensveen FM, Valentic S, Sestan M, Wensveen TT, Polic B. Interactions between adipose tissue and the immune system in health and malnutrition. Semin Immunol. (2015) 27:322–33. 10.1016/j.smim.2015.10.006
    1. Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. J Clin Endocrinol Metab. (2004) 89:2548–56. 10.1210/jc.2004-0395
    1. Ahima RS, Flier JS. Adipose tissue as an endocrine organ. Trends Endocrinol Metab. (2000) 11:327–32. 10.1016/S1043-2760(00)00301-5
    1. Scherer PE. Adipose tissue: from lipid storage compartment to endocrine organ. Diabetes (2006) 55:1537–45. 10.2337/db06-0263
    1. Han SJ, Glatman Zaretsky A, Andrade-Oliveira V, Collins N, Dzutsev A, Shaik J, et al. . White adipose tissue is a reservoir for memory t cells and promotes protective memory responses to infection. Immunity (2017) 47:1154–68.e6. 10.1016/j.immuni.2017.11.009
    1. Eberl G, Colonna M, Di Santo JP, McKenzie AN. Innate lymphoid cells. Innate lymphoid cells: a new paradigm in immunology. Science (2015) 348:aaa6566. 10.1126/science.aaa6566
    1. Wu H, Ballantyne CM. Skeletal muscle inflammation and insulin resistance in obesity. J Clin Invest. (2017) 127:43–54. 10.1172/JCI88880
    1. Gesta S, Tseng YH, Kahn CR. Developmental origin of fat: tracking obesity to its source. Cell (20072) 131:242–56. 10.1016/j.cell.2007.10.004
    1. Peirce V, Carobbio S, Vidal-Puig A. The different shades of fat. Nature (2014) 510:76–83. 10.1038/nature13477
    1. Tchkonia T, Thomou T, Zhu Y, Karagiannides I, Pothoulakis C, Jensen MD, et al. . Mechanisms and metabolic implications of regional differences among fat depots. Cell Metab. (2013) 17:644–56. 10.1016/j.cmet.2013.03.008
    1. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science (1993) 259:87–91. 10.1126/science.7678183
    1. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest. (1995) 95:2409–15. 10.1172/JCI117936
    1. Rotter V, Nagaev I, Smith U. Interleukin-6 (IL-6) induces insulin resistance in 3T3-L1 adipocytes and is, like IL-8 and tumor necrosis factor-alpha, overexpressed in human fat cells from insulin-resistant subjects. J Biol Chem. (2003) 278:45777–84. 10.1074/jbc.M301977200
    1. Sabio G, Das M, Mora A, Zhang Z, Jun JY, Ko HJ, et al. . A stress signaling pathway in adipose tissue regulates hepatic insulin resistance. Science (2008) 322:1539–43. 10.1126/science.1160794
    1. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW, Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. (2003) 112:1796–808. 10.1172/JCI19246
    1. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, et al. . Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. (2003) 112:1821–30. 10.1172/JCI200319451
    1. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. (2007) 117:175–84. 10.1172/JCI29881
    1. Wu H, Perrard XD, Wang Q, Perrard JL, Polsani VR, Jones PH, et al. . CD11c expression in adipose tissue and blood and its role in diet-induced obesity. Arterioscler Thromb Vasc Biol. (2010) 30:186–92. 10.1161/ATVBAHA.109.198044
    1. Sartipy P, Loskutoff DJ. Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc Natl Acad Sci USA. (2003) 100:7265–70. 10.1073/pnas.1133870100
    1. Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, et al. . MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. (2006) 116:1494–505. 10.1172/JCI26498
    1. Weisberg SP, Hunter D, Huber R, Lemieux J, Slaymaker S, Vaddi K, et al. . CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest. (2006) 116:115–24. 10.1172/JCI24335
    1. Wu H, Ghosh S, Perrard XD, Feng L, Garcia GE, Perrard JL, et al. . T-cell accumulation and regulated on activation, normal T cell expressed and secreted upregulation in adipose tissue in obesity. Circulation (2007) 115:1029–38. 10.1161/CIRCULATIONAHA.106.638379
    1. Kaminski DA, Randall TD. Adaptive immunity and adipose tissue biology. Trends Immunol. (2010) 31:384–90. 10.1016/j.it.2010.08.001
    1. Jiang E, Perrard XD, Yang D, Khan IM, Perrard JL, Smith CW, et al. . Essential role of CD11a in CD8+ T-cell accumulation and activation in adipose tissue. Arterioscler Thromb Vasc Biol. (2014) 34:34–43. 10.1161/ATVBAHA.113.302077
    1. Khan IM, Dai Perrard XY, Perrard JL, Mansoori A, Smith CW, Wu H, et al. . Attenuated adipose tissue and skeletal muscle inflammation in obese mice with combined CD4+ and CD8+ T cell deficiency. Atherosclerosis (2014) 233:419–28. 10.1016/j.atherosclerosis.2014.01.011
    1. Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, et al. . CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. (2009) 15:914–20. 10.1038/nm.1964
    1. Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med. (2009) 15:930–9. 10.1038/nm.2002
    1. Schipper HS, Prakken B, Kalkhoven E, Boes M. Adipose tissue-resident immune cells: key players in immunometabolism. Trends Endocrinol Metab. (2012) 23:407–15. 10.1016/j.tem.2012.05.011
    1. Nishimura S, Manabe I, Takaki S, Nagasaki M, Otsu M, Yamashita H, et al. Adipose natural regulatory B cells negatively control adipose tissue inflammation. Cell Metab. (2013) 18:759–66. 10.1016/j.cmet.2013.09.017
    1. Molofsky AB, Nussbaum JC, Liang HE, Van Dyken SJ, Cheng LE, Mohapatra A, et al. . Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages. J Exp Med. (2013) 210:535–49. 10.1084/jem.20121964
    1. Wu D, Molofsky AB, Liang HE, Ricardo-Gonzalez RR, Jouihan HA, Bando JK, et al. . Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis. Science (2011) 332:243–7. 10.1126/science.1201475
    1. Abbas AL, Pillai AS. Cellular and Molecular Immunology. 8th ed. Philadelphia, PA: Elsevier Saunders; (2014).
    1. Rocha VZ, Libby P. Obesity, inflammation, and atherosclerosis. Nat Rev Cardiol. (2009) 6:399–409. 10.1038/nrcardio.2009.55
    1. Huber J, Kiefer FW, Zeyda M, Ludvik B, Silberhumer GR, Prager G, et al. . CC chemokine and CC chemokine receptor profiles in visceral and subcutaneous adipose tissue are altered in human obesity. J Clin Endocrinol Metab. (2008) 93:3215–21. 10.1210/jc.2007-2630
    1. Kintscher U, Hartge M, Hess K, Foryst-Ludwig A, Clemenz M, Wabitsch 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) 28:1304–10. 10.1161/ATVBAHA.108.165100
    1. Deng T, Lyon CJ, Minze LJ, Lin J, Zou J, Liu JZ, et al. . Class II major histocompatibility complex plays an essential role in obesity-induced adipose inflammation. Cell Metab. (2013) 17:411–22. 10.1016/j.cmet.2013.02.009
    1. Panduro M, Benoist C, Mathis D. Tissue tregs. Annu Rev Immunol. (2016) 34:609–33. 10.1146/annurev-immunol-032712-095948
    1. Cipolletta D, Feuerer M, Li A, Kamei N, Lee J, Shoelson SE, et al. . PPAR-gamma is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature (2012) 486:549–53. 10.1038/nature11132
    1. Cipolletta D. Adipose tissue-resident regulatory T cells: phenotypic specialization, functions and therapeutic potential. Immunology (2014) 142:517–25. 10.1111/imm.12262
    1. Cipolletta D, Cohen P, Spiegelman BM, Benoist C, Mathis D. Appearance and disappearance of the mRNA signature characteristic of Treg cells in visceral adipose tissue: age, diet, and PPARgamma effects. Proc Natl Acad Sci USA. (2015) 112:482–7. 10.1073/pnas.1423486112
    1. Vasanthakumar A, Moro K, Xin A, Liao Y, Gloury R, Kawamoto S, et al. The transcriptional regulators IRF4, BATF and IL-33 orchestrate development and maintenance of adipose tissue-resident regulatory T cells. Nat Immunol. (2015) 16:276–85. 10.1038/ni.3085
    1. Becker M, Levings MK, Daniel C. Adipose-tissue regulatory T cells: critical players in adipose-immune crosstalk. Eur J Immunol. (2017) 47:1867–74. 10.1002/eji.201646739
    1. Han JM, Wu D, Denroche HC, Yao Y, Verchere CB, Levings MK. IL-33 Reverses an obesity-induced deficit in visceral adipose tissue ST2+ T regulatory cells and ameliorates adipose tissue inflammation and insulin resistance. J Immunol. (2015) 194:4777–83. 10.4049/jimmunol.1500020
    1. Wood IS, Wang B, Trayhurn P. IL-33, a recently identified interleukin-1 gene family member, is expressed in human adipocytes. Biochem Biophys Res Commun. (2009) 384:105–9. 10.1016/j.bbrc.2009.04.081
    1. Kohlgruber AC, Gal-Oz ST, LaMarche NM, Shimazaki M, Duquette D, Nguyen HN, et al. . Gammadelta T cells producing interleukin-17A regulate adipose regulatory T cell homeostasis and thermogenesis. Nat Immunol. (2018) 19:464–74. 10.1038/s41590-018-0094-2
    1. Lynch L, Michelet X, Zhang S, Brennan PJ, Moseman A, Lester C, et al. Regulatory iNKT cells lack expression of the transcription factor PLZF and control the homeostasis of Treg cells and macrophages in adipose tissue. Nat Immunol. (2015) 16:85–95. 10.1038/ni.3047
    1. Molofsky AB, Van Gool F, Liang HE, Van Dyken SJ, Nussbaum JC, Lee J, et al. . Interleukin-33 and interferon-gamma counter-regulate group 2 innate lymphoid cell activation during immune perturbation. Immunity (2015) 43:161–74. 10.1016/j.immuni.2015.05.019
    1. Li C, DiSpirito JR, Zemmour D, Spallanzani RG, Kuswanto W, Benoist C, et al. . TCR Transgenic mice reveal stepwise, multi-site acquisition of the distinctive fat-treg phenotype. Cell (2018) 174:285–99.e12. 10.1016/j.cell.2018.05.004
    1. Matarese G, Procaccini C, De Rosa V, Horvath TL, La Cava A. Regulatory T cells in obesity: the leptin connection. Trends Mol Med. (2010) 16:247–56. 10.1016/j.molmed.2010.04.002
    1. Lee HY, Kim J, Quan W, Lee JC, Kim MS, Kim SH, et al. . Autophagy deficiency in myeloid cells increases susceptibility to obesity-induced diabetes and experimental colitis. Autophagy (2016) 12:1390–403. 10.1080/15548627.2016.1184799
    1. Fabrizi M, Marchetti V, Mavilio M, Marino A, Casagrande V, Cavalera M, et al. . IL-21 is a major negative regulator of IRF4-dependent lipolysis affecting Tregs in adipose tissue and systemic insulin sensitivity. Diabetes (2014) 63:2086–96. 10.2337/db13-0939
    1. Cho KW, Morris DL, DelProposto JL, Geletka L, Zamarron B, Martinez-Santibanez G, et al. . An MHC II-dependent activation loop between adipose tissue macrophages and CD4+ T cells controls obesity-induced inflammation. Cell Rep. (2014) 9:605–17. 10.1016/j.celrep.2014.09.004
    1. Morris DL, Cho KW, Delproposto JL, Oatmen KE, Geletka LM, Martinez-Santibanez G, et al. . Adipose tissue macrophages function as antigen-presenting cells and regulate adipose tissue CD4+ T cells in mice. Diabetes (2013) 62:2762–72. 10.2337/db12-1404
    1. Khan IM, Perrard XY, Brunner G, Lui H, Sparks LM, Smith SR, et al. . Intermuscular and perimuscular fat expansion in obesity correlates with skeletal muscle T cell and macrophage infiltration and insulin resistance. Int J Obes. (2015) 39:1607–18. 10.1038/ijo.2015.104
    1. Rocha VZ, Folco EJ, Sukhova G, Shimizu K, Gotsman I, Vernon AH, et al. . Interferon-gamma, a Th1 cytokine, regulates fat inflammation: a role for adaptive immunity in obesity. Circ Res. (2008) 103:467–76. 10.1161/CIRCRESAHA.108.177105
    1. McGillicuddy FC, Chiquoine EH, Hinkle CC, Kim RJ, Shah R, Roche HM, et al. . Interferon gamma attenuates insulin signaling, lipid storage, and differentiation in human adipocytes via activation of the JAK/STAT pathway. J Biol Chem. (2009) 284:31936–44. 10.1074/jbc.M109.061655
    1. Cho KW, Zamarron BF, Muir LA, Singer K, Porsche CE, DelProposto JB, et al. . Adipose tissue dendritic cells are independent contributors to obesity–induced inflammation and insulin resistance. J Immunol. (2016) 197:3650–61. 10.4049/jimmunol.1600820
    1. Xiao L, Yang X, Lin Y, Li S, Jiang J, Qian S, et al. . Large adipocytes function as antigen-presenting cells to activate CD4+ T cells via upregulating MHCII in obesity. Int J Obes. (2016) 40:112–20. 10.1038/ijo.2015.145
    1. Deng T, Liu J, Deng Y, Minze L, Xiao X, Wright V, et al. Adipocyte adaptive immunity mediates diet-induced adipose inflammation and insulin resistance by decreasing adipose Treg cells. Nat Commun. (2017) 8:15725 10.1038/ncomms15725
    1. Kim CS, Kim JG, Lee BJ, Choi MS, Choi HS, Kawada T, et al. . Deficiency for costimulatory receptor 4-1BB protects against obesity-induced inflammation and metabolic disorders. Diabetes (2011) 60:3159–68. 10.2337/db10-1805
    1. Poggi M, Morin SO, Bastelica D, Govers R, Canault M, Bernot D, et al. . CD28 deletion improves obesity-induced liver steatosis but increases adiposity in mice. Int J Obes. (2015) 39:977–85. 10.1038/ijo.2015.26
    1. Kim YJ, Kim SH, Mantel P, Kwon BS. Human 4-1BB regulates CD28 co-stimulation to promote Th1 cell responses. Eur J Immunol. (1998) 28:881–90. 10.1002/(SICI)1521-4141(199803)28:03<881::AID-IMMU881>;2-0
    1. Kwon B, Lee HW, Kwon BS. New insights into the role of 4-1BB in immune responses: beyond CD8+ T cells. Trends Immunol. (2002) 23:378–80. 10.1016/S1471-4906(02)02263-9
    1. Vinay DS, Kwon BS. Role of 4-1BB in immune responses. Semin Immunol. (1998) 10:481–9.
    1. Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol. (2013) 13:227–42. 10.1038/nri3405
    1. Simpson TR, Quezada SA, Allison JP. Regulation of CD4 T cell activation and effector function by inducible costimulator (ICOS). Curr Opin Immunol. (2010) 22:326–32. 10.1016/j.coi.2010.01.001
    1. Khayyamian S, Hutloff A, Buchner K, Grafe M, Henn V, Kroczek RA, et al. . ICOS-ligand, expressed on human endothelial cells, costimulates Th1 and Th2 cytokine secretion by memory CD4+ T cells. Proc Natl Acad Sci USA. (2002) 99:6198–203. 10.1073/pnas.092576699
    1. Cameron G, Godfrey DI. Differential surface phenotype and context-dependent reactivity of functionally diverse NKT cells. Immunol Cell Biol. (2018) 79:759–71. 10.1111/imcb.12034
    1. Wu J, Yang J, Yang K, Wang H, Gorentla B, Shin J, et al. . iNKT cells require TSC1 for terminal maturation and effector lineage fate decisions. J Clin Invest. (2014) 124:1685–98. 10.1172/JCI69780
    1. Thery C, Duban L, Segura E, Veron P, Lantz O, Amigorena S. Indirect activation of naive CD4+ T cells by dendritic cell-derived exosomes. Nat Immunol. (2002) 3:1156–62. 10.1038/ni854
    1. Thery C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol. (2009) 9:581–93. 10.1038/nri2567
    1. Nakayama M. Antigen presentation by MHC-dressed cells. Front Immunol. (2014) 5:672. 10.3389/fimmu.2014.00672
    1. Winer S, Paltser G, Chan Y, Tsui H, Engleman E, Winer D, et al. . Obesity predisposes to Th17 bias. Eur J Immunol. (2009) 39:2629–35. 10.1002/eji.200838893
    1. Ahmed M, Gaffen SL. IL-17 in obesity and adipogenesis. Cytokine Growth Factor Rev. (2010) 21:449–53. 10.1016/j.cytogfr.2010.10.005
    1. Sumarac-Dumanovic M, Stevanovic D, Ljubic A, Jorga J, Simic M, Stamenkovic-Pejkovic D, et al. . Increased activity of interleukin-23/interleukin-17 proinflammatory axis in obese women. Int J Obes. (2009) 33:151–6. 10.1038/ijo.2008.216
    1. Bertola A, Ciucci T, Rousseau D, Bourlier V, Duffaut C, Bonnafous S, et al. . Identification of adipose tissue dendritic cells correlated with obesity-associated insulin-resistance and inducing Th17 responses in mice and patients. Diabetes (2012) 61:2238–47. 10.2337/db11-1274
    1. Fabbrini E, Cella M, McCartney SA, Fuchs A, Abumrad NA, Pietka TA, et al. . Association between specific adipose tissue CD4+ T-cell populations and insulin resistance in obese individuals. Gastroenterology (2013) 145:366–74.e1-3. 10.1053/j.gastro.2013.04.010
    1. Chehimi M, Vidal H, Eljaafari A. Pathogenic role of IL-17-producing immune cells in obesity, and related inflammatory diseases. J Clin Med. (2017) 6:E68. 10.3390/jcm6070068
    1. McLaughlin T, Liu LF, Lamendola C, Shen L, Morton J, Rivas H, et al. . T-cell profile in adipose tissue is associated with insulin resistance and systemic inflammation in humans. Arterioscler Thromb Vasc Biol. (2014) 34:2637–43. 10.1161/ATVBAHA.114.304636
    1. Pandolfi JB, Ferraro AA, Sananez I, Gancedo MC, Baz P, Billordo LA, et al. . ATP-induced inflammation drives tissue-resident Th17 cells in metabolically unhealthy obesity. J Immunol. (2016) 196:3287–96. 10.4049/jimmunol.1502506
    1. Mehta P, Nuotio-Antar AM, Smith CW. Gammadelta T cells promote inflammation and insulin resistance during high fat diet-induced obesity in mice. J Leukoc Biol. (2015) 97:121–34. 10.1189/jlb.3A0414-211RR
    1. Winer S, Chan Y, Paltser G, Truong D, Tsui H, Bahrami J, et al. . Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med. (2009) 15:921–9. 10.1038/nm.2001
    1. Rausch ME, Weisberg S, Vardhana P, Tortoriello DV. Obesity in C57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T-cell infiltration. Int J Obes. (2008) 32:451–63. 10.1038/sj.ijo.0803744
    1. Duffaut C, Zakaroff-Girard A, Bourlier V, Decaunes P, Maumus M, Chiotasso P, et al. . Interplay between human adipocytes and T lymphocytes in obesity: CCL20 as an adipochemokine and T lymphocytes as lipogenic modulators. Arterioscler Thromb Vasc Biol. (2009) 29:1608–14. 10.1161/ATVBAHA.109.192583
    1. Yang H, Youm YH, Vandanmagsar B, Ravussin A, Gimble JM, Greenway F, et al. . Obesity increases the production of proinflammatory mediators from adipose tissue T cells and compromises TCR repertoire diversity: implications for systemic inflammation and insulin resistance. J Immunol. (2010) 185:1836–45. 10.4049/jimmunol.1000021
    1. Wensveen FM, Valentic S, Sestan M, Turk Wensveen T, Polic B. The “Big Bang” in obese fat: events initiating obesity-induced adipose tissue inflammation. Eur J Immunol. (2015) 45:2446–56. 10.1002/eji.201545502
    1. Terrell CE, Jordan MB. Perforin deficiency impairs a critical immunoregulatory loop involving murine CD8+ T cells and dendritic cells. Blood (2013) 121:5184–91. 10.1182/blood-2013-04-495309
    1. Lykens JE, Terrell CE, Zoller EE, Risma K, Jordan MB. Perforin is a critical physiologic regulator of T-cell activation. Blood (2011) 118:618–26. 10.1182/blood-2010-12-324533
    1. Revelo XS, Tsai S, Lei H, Luck H, Ghazarian M, Tsui H, et al. . Perforin is a novel immune regulator of obesity-related insulin resistance. Diabetes (2015) 64:90–103. 10.2337/db13-1524
    1. Gapin L. Development of invariant natural killer T cells. Curr Opin Immunol. (2016) 39:68–74. 10.1016/j.coi.2016.01.001
    1. Godfrey DI, MacDonald HR, Kronenberg M, Smyth MJ, Van Kaer L. NKT cells: what's in a name? Nat Rev Immunol. (2004) 4:231–7. 10.1038/nri1309
    1. Kumar A, Suryadevara N, Hill TM, Bezbradica JS, Van Kaer L, Joyce S. Natural killer T cells: an ecological evolutionary developmental biology perspective. Front Immunol. (2017) 8:1858. 10.3389/fimmu.2017.01858
    1. Lynch L, O'Shea D, Winter DC, Geoghegan J, Doherty DG, O'Farrelly C. Invariant NKT cells and CD1d+ cells amass in human omentum and are depleted in patients with cancer and obesity. Eur J Immunol. (2009) 39:1893–901. 10.1002/eji.200939349
    1. Lynch L, Nowak M, Varghese B, Clark J, Hogan AE, Toxavidis V, et al. . Adipose tissue invariant NKT cells protect against diet-induced obesity and metabolic disorder through regulatory cytokine production. Immunity (2012) 37:574–87. 10.1016/j.immuni.2012.06.016
    1. Huh JY, Park J, Kim JI, Park YJ, Lee YK, Kim JB. Deletion of CD1d in adipocytes aggravates adipose tissue inflammation and insulin resistance in obesity. Diabetes (2017) 66:835–47. 10.2337/db16-1122
    1. Huh JY, Park YJ, Kim JB. Adipocyte CD1d determines adipose inflammation and insulin resistance in obesity. Adipocyte (2018) 7:129–36. 10.1080/21623945.2018.1440928
    1. Huh JY, Kim JI, Park YJ, Hwang IJ, Lee YS, Sohn JH, et al. . A novel function of adipocytes in lipid antigen presentation to iNKT cells. Mol Cell Biol. (2013) 33:328–39. 10.1128/MCB.00552-12
    1. Schipper HS, Rakhshandehroo M, van de Graaf SF, Venken K, Koppen A, Stienstra R, et al. . Natural killer T cells in adipose tissue prevent insulin resistance. J Clin Invest. (2012) 122:3343–54. 10.1172/JCI62739
    1. Lynch L, Hogan AE, Duquette D, Lester C, Banks A, LeClair K, et al. . iNKT cells induce FGF21 for thermogenesis and are required for maximal weight loss in GLP1 therapy. Cell Metab. (2016) 24:510–9. 10.1016/j.cmet.2016.08.003
    1. Zhao H, Shang Q, Pan Z, Bai Y, Li Z, Zhang H, et al. . Exosomes from adipose-derived stem cells attenuate adipose inflammation and obesity through polarizing M2 macrophages and beiging in white adipose tissue. Diabetes (2018) 67:235–47. 10.2337/db17-0356
    1. Shang Q, Bai Y, Wang G, Song Q, Guo C, Zhang L, et al. . Delivery of adipose-derived stem cells attenuates adipose tissue inflammation and insulin resistance in obese mice through remodeling macrophage phenotypes. Stem Cells Dev. (2015) 24:2052–64. 10.1089/scd.2014.0557
    1. Eljaafari A, Robert M, Chehimi M, Chanon S, Durand C, Vial G, et al. . Adipose tissue-derived stem cells from obese subjects contribute to inflammation and reduced insulin response in adipocytes through differential regulation of the Th1/Th17 balance and monocyte activation. Diabetes (2015) 64:2477–88. 10.2337/db15-0162

Source: PubMed

3
订阅