The impact of adiposity on adipose tissue-resident lymphocyte activation in humans

R L Travers, A C Motta, J A Betts, A Bouloumié, D Thompson, R L Travers, A C Motta, J A Betts, A Bouloumié, D Thompson

Abstract

Background/objectives: The presence of T lymphocytes in human adipose tissue has only recently been demonstrated and relatively little is known of their potential relevance in the development of obesity-related diseases. We aimed to further characterise these cells and in particular to investigate how they interact with modestly increased levels of adiposity typical of common overweight and obesity.

Subjects/methods: Subcutaneous adipose tissue and fasting blood samples were obtained from healthy males aged 35-55 years with waist circumferences in lean (<94 cm), overweight (94-102 cm) and obese (>102 cm) categories. Adipose tissue-resident CD4+ and CD8+ T lymphocytes together with macrophages were identified by gene expression and flow cytometry. T lymphocytes were further characterised by their expression of activation markers CD25 and CD69. Adipose tissue inflammation was investigated using gene expression analysis and tissue culture.

Results: Participants reflected a range of adiposity from lean to class I obesity. Expression of CD4 (T-helper cells) and CD68 (macrophage), as well as FOXP3 RNA transcripts, was elevated in subcutaneous adipose tissue with increased levels of adiposity (P<0.001, P<0.001 and P=0.018, respectively). Flow cytometry revealed significant correlations between waist circumference and levels of CD25 and CD69 expression per cell on activated adipose tissue-resident CD4+ and CD8+ T lymphocytes (P-values ranging from 0.053 to <0.001). No such relationships were found with blood T lymphocytes. This increased T lymphocyte activation was related to increased expression and secretion of various pro- and anti-inflammatory cytokines from subcutaneous whole adipose tissue explants.

Conclusions: This is the first study to demonstrate that even modest levels of overweight/obesity elicit modifications in adipose tissue immune function. Our results underscore the importance of T lymphocytes during adipose tissue expansion, and the presence of potential compensatory mechanisms that may work to counteract adipose tissue inflammation, possibly through an increased number of T-regulatory cells.

Figures

Figure 1
Figure 1
Lymphocyte phenotype and activation in adipose tissue according to levels of central adiposity. (a) Relative gene expression of cluster differentiation markers to identify T lymphocytes (n=30), (b) CD4+ T lymphocyte subsets present within the adipose tissue (n=30) and (c) proportions of activated T lymphocytes in adipose tissue SVF as a percentage of CD4+ and CD8+ cells measured by flow cytometry (n=17). Gene expression data presented as mean 2−▵▵Ct±s.e.m. Effects of adiposity were analysed by one-way analysis of variance (ANOVA), *P<0.05 and **P<0.001. (d) (i–iv) Correlations between waist circumference and activation status of CD4+ and CD8+ T lymphocytes in the adipose tissue (n=17) were analysed using Pearson's (R) correlation; coefficients shown along with significance values. MFI, mean fluorescence intensity.
Figure 2
Figure 2
Macrophage numbers and activation in adipose tissue with varying levels of adiposity. (a) Relative gene expression of CD68 used to identify macrophages (n=30). (b) Proportions of macrophages in adipose tissue SVF as a percentage of the total cells (n=17). Data are presented as mean 2−▵▵Ct±s.e.m. Effects of adiposity were analysed by one-way analysis of variance (ANOVA), *P<0.05 and **P<0.001. (c) Correlations between waist circumference and proportion of macrophages present in the adipose tissue SVF (n=17) were analysed using Pearson's (R) correlation. MFI, mean fluorescence intensity.
Figure 3
Figure 3
Relative gene expression of parameters related to metabolism, appetite/adiposity and inflammatory cytokines in whole adipose tissue samples with varying levels of adiposity. Data are presented as mean 2−▵▵Ct±s.e.m. with participants classified equally based on waist circumference (n=30). Effects of adiposity were analysed by one-way analysis of variance (ANOVA), *P<0.05 and **P<0.005. Note that IL-10 was expressed in three lean, eight overweight and seven obese individuals. IL-6, IL-8 and IL-1β were detected in all overweight and obese individuals but only in eight, six and nine lean individuals, respectively.
Figure 4
Figure 4
Cytokine secretion by whole adipose tissue explants cultured for 3h with varying levels of adiposity. (a) Adipokine secretion normalised per 100 mg ml−1 adipose tissue cultured. (b) Adipokine secretion multiplied by L1–L4 fat mass to predict total central adipose tissue adipokine secretion. Mean and s.e.m. values were shown for groups based on waist circumference. Effects of adiposity were analysed by one-way analysis of variance (ANOVA), *P<0.05, **P<0.005 and $P=0.06 (lean, n=8; overweight, n=6; obese, n=10).

References

    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;112:1796–1808.
    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;112:1821–1830.
    1. Curat CA, Miranville A, Sengenes C, Diehl M, Tonus C, Busse R, et al. From blood monocytes to adipose tissue-resident macrophages: induction of diapedesis by human mature adipocytes. Diabetes. 2004;53:1285–1292.
    1. Zeyda M, Farmer D, Todoric J, Aszmann O, Speiser M, Gyori G, et al. Human adipose tissue macrophages are of an anti-inflammatory phenotype but capable of excessive pro-inflammatory mediator production. Int J Obes (Lond) 2007;31:1420–1428.
    1. Bourlier V, Zakaroff-Girard A, Miranville A, De Barros S, Maumus M, Sengenes C, et al. Remodeling phenotype of human subcutaneous adipose tissue macrophages. Circulation. 2008;117:806–815.
    1. Witztum JL, Lichtman AH. The influence of innate and adaptive immune responses on atherosclerosis. Annu Rev Pathol. 2014;9:73–102.
    1. van Oosterhout AJ, Bloksma N. Regulatory T-lymphocytes in asthma. Eur Resp J. 2005;26:918–932.
    1. Cope AP, Schulze-Koops H, Aringer M. The central role of T cells in rheumatoid arthritis. Clin Exp Rheumatol. 2007;25:S4–11.
    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;115:1029–1038.
    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;29:1608–1614.
    1. Zeyda M, Huber J, Prager G, Stulnig TM. Inflammation correlates with markers of T-cell subsets including regulatory T cells in adipose tissue from obese patients. Obesity (Silver Spring, MD) 2011;19:743–748.
    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;28:1304–1310.
    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;15:930–939.
    1. Goossens GH, Blaak EE, Theunissen R, Duijvestijn AM, Clement K, Tervaert JW, et al. Expression of NLRP3 inflammasome and T cell population markers in adipose tissue are associated with insulin resistance and impaired glucose metabolism in humans. Mol Immunol. 2012;50:142–149.
    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;15:914–920.
    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;61:2238–2247.
    1. Health and Social Care Information Centre Health Survey for England 2012 Trend Tables: Adult Trend TablesHealth and Social Care Information, , . 18 December 2013.
    1. World Health Organisation . Waist Circumference and Waist-Hip Ratio: Report of a WHO Expert Consultation. Geneva, Switzerland; WHO; 2008.
    1. Paradisi G, Smith L, Burtner C, Leaming R, Garvey WT, Hook G, et al. Dual energy X-ray absorptiometry assessment of fat mass distribution and its association with the insulin resistance syndrome. Diabetes Care. 1999;22:1310–1317.
    1. Kelly TL, Wilson KE, Heymsfield SB. Dual energy X-ray absorptiometry body composition reference values from NHANES. PLoS One. 2009;4:e7038.
    1. Walhin JP, Richardson JD, Betts JA, Thompson D. Exercise counteracts the effects of short-term overfeeding and reduced physical activity independent of energy imbalance in healthy young men. J Physiol. 2013;591:6231–6243.
    1. Fain JN, Madan AK, Hiler ML, Cheema P, Bahouth SW. Comparison of the release of adipokines by adipose tissue, adipose tissue matrix, and adipocytes from visceral and subcutaneous abdominal adipose tissues of obese humans. Endocrinology. 2004;145:2273–2282.
    1. Neville MJ, Collins JM, Gloyn AL, McCarthy MI, Karpe F. Comprehensive human adipose tissue mRNA and microRNA endogenous control selection for quantitative real-time-PCR normalization. Obesity (Silver Spring, MD) 2011;19:888–892.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 2001;25:402–408.
    1. Maxwell SE, Delaney HD. Designing Experiments and Analysing Data: A Model Comparison Perspective. Belmont, CA, USA; Wadsworth; 1990.
    1. Schmetterer KG, Neunkirchner A, Pickl WF. Naturally occurring regulatory T cells: markers, mechanisms, and manipulation. FASEB J. 2012;26:2253–2276.
    1. Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol. 2012;30:531–564.
    1. Lord GM, Matarese G, Howard JK, Baker RJ, Bloom SR, Lechler RI. Leptin modulates the T-cell immune response and reverses starvation-induced immunosuppression. Nature. 1998;394:897–901.
    1. Sanchez-Margalet V, Martin-Romero C, Santos-Alvarez J, Goberna R, Najib S, Gonzalez-Yanes C. Role of leptin as an immunomodulator of blood mononuclear cells: mechanisms of action. Clin Exp Immunol. 2003;133:11–19.
    1. Martin-Romero C, Santos-Alvarez J, Goberna R, Sanchez-Margalet V. Human leptin enhances activation and proliferation of human circulating T lymphocytes. Cell Immunol. 2000;199:15–24.
    1. Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011;17:179–188.
    1. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117:175–184.
    1. Fjeldborg K, Pedersen SB, Moller HJ, Christiansen T, Bennetzen M, Richelsen B. Human adipose tissue macrophages are enhanced but changed to an anti-inflammatory profile in obesity. J Immunol Res. 2014;2014:309548.
    1. McQuaid SE, Hodson L, Neville MJ, Dennis AL, Cheeseman J, Humphreys SM, et al. Downregulation of adipose tissue fatty acid trafficking in obesity: a driver for ectopic fat deposition. Diabetes. 2011;60:47–55.
    1. Galli SJ, Borregaard N, Wynn TA. Phenotypic and functional plasticity of cells of innate immunity: macrophages, mast cells and neutrophils. Nat Immunol. 2011;12:1035–1044.
    1. Mora JR, von Andrian UH. T-cell homing specificity and plasticity: new concepts and future challenges. Trends Immunol. 2006;27:235–243.

Source: PubMed

3
Iratkozz fel