CD4+ T cells memorize obesity and promote weight regain

Jianghuan Zou, Beibei Lai, Mingzhu Zheng, Qin Chen, Shujun Jiang, Anying Song, Zan Huang, Peiliang Shi, Xin Tu, Di Wang, Linrong Lu, Zhaoyu Lin, Xiang Gao, Jianghuan Zou, Beibei Lai, Mingzhu Zheng, Qin Chen, Shujun Jiang, Anying Song, Zan Huang, Peiliang Shi, Xin Tu, Di Wang, Linrong Lu, Zhaoyu Lin, Xiang Gao

Abstract

Body weight regain often causes failure of obesity therapies while the underlying mechanism remains largely unknown. In this study, we report that immune cells, especially CD4+ T cells, mediate the 'memory' of previous obese status. In a weight gain-loss-regain model, we found that C57BL/6J mice with an obesity history showed a much faster rate of body weight regain. This obesity memory could last for at least 2 months after previously obese mice were kept at the same body weight as non-obese mice. Surprisingly, such obesity memory was abrogated by dexamethasone treatment, whereas immunodeficient Rag1-/- and H2A-/- mice failed to establish such memory. Rag1-/- mice repossessed the obesity memory when immune cells or CD4+ T cells isolated from previously obese mice were transferred. Furthermore, depletion of CD4+ T cells led to obesity memory ablation. Taken together, we conclude that CD4+ T cells mediate obesity memory and promote weight regain.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Weight gain-loss-regain model and long-term obesity memory. The DIO model in C57BL/6J mice was established as per the simulative graph and timeline (a). Body weight curves (b–d) and body weight increment ratios during the weight regain period (e–g) were recorded to display obesity memory. Weight regain was induced with a HFD (b, e and d, g) and a RD (d, f). The data are shown as means±s.d. *P<0.05 and **P<0.01, unpaired Student’s t-test. §§§P<0.001, two-way ANOVA followed by the Bonferroni post hoc test. CTR, control group; DIO, diet-induced-obesity; HFD, high fat diet; MOH, obesity history group; RD, regular diet.
Figure 2
Figure 2
Metabolic parameters of C57BL/6J mice before weight regain. Body weight (a), body length (b), overall and local bone mineral density (c), bone mineral content, lean mass and fat mass (d) and body fat percentage (e) values were measured to determine body compositions, especially fat content, of the MOH and CTR mice. The fasting and postprandial blood glucose levels (f) and insulin sensitivity (g) were detected to determine glucose metabolism in both cohorts. O2 consumption (h), CO2 production (i), RER (j), ambulatory activity (k), heat production (l) and food intake (m) were detected to examine energy expenditure. Data are shown as means±s.d. No difference was detected in this figure. CTR, control group; MOH, obesity history group; RER, respiratory exchange ratio.
Figure 3
Figure 3
Immunodeficiency leads to failure in establishing obesity memory. Immunodeficient Rag1−/− mice and C57BL/6J mice that were treated with dexamethasone (dexa) were subjected to a weight gain-loss-regain cycle. Body weight curve (a, c) and body weight increment ratio values during the weight regain period (b, d) were recorded to examine obesity memory in Rag1−/− mice (a, b) and dexa-treated C57BL/6J mice (c, d), respectively. Adoptive transfer of splenocytes was performed to determine the role of immune cells in memorizing obesity. The body weight curves of donor C57BL/6J mice (e, h) and recipient Rag1−/− mice (f, i) and the body weight increment ratios of recipients during HFD feeding (g, j) were recorded after weight loss (eg) and after 1 month of weight maintenance (hj), respectively. The cell collection time is indicated by a black arrow. Data are shown as means±s.d. ***P<0.001, unpaired Student’s t-test. §§§P<0.001, two-way ANOVA followed by the Bonferroni post hoc test. HFD, high fat diet; Ns, no significance.
Figure 4
Figure 4
Role of CD4+ T cells in establishing and storing obesity memory. CD4+T cell-deficient H2A−/− mice were subjected to the weight gain-loss-regain cycle. Body weight curve (a) and body weight increment ratio values during the weight regain period (b) were recorded to examine obesity memory in H2A−/− mice. MOH C57BL/6J mice were injected with an anti-CD4 antibody (dMOH) or corresponding isotype IgG (sMOH) to determine the role of CD4+ T cells in obesity memory. CTR C57BL/6J mice were injected with saline. The injection time is indicated with a black arrow. Body weight curve (c) and body weight increment ratio values during the weight regain period (d) were recorded to examine obesity memory. Adoptive transfer of CD4+ T cells was conducted to confirm their role in memorizing obesity. The body weight curves of donor C57BL/6J (e, h) and recipient Rag1−/− mice (f, i) and the body weight increment ratios of recipients during the HFD feeding period (g, j) were recorded after weight loss (eg) and after 1 month of weight maintenance (hj), respectively. The cell collection time is indicated by a black arrow. Data are shown as means±s.d. *P<0.05, **P<0.01 and ***P<0.001, unpaired Student’s t-test. §§§P<0.001, two-way ANOVA followed by the Bonferroni post hoc test. CTR, control group; dMOH, CD4+ T cell-deficient MOH group; HFD, high fat diet; mCD4T, CD4+ T cells carrying obesity memory group; MOH, obesity history group; NC, negative control group; Ns, no significance; PC, positive control group; sMOH, CD4+ T cell-sufficient MOH group.

References

    1. Yoon KH, Lee JH, Kim JW, Cho JH, Choi YH, Ko SH, et al. Epidemic obesity and type 2 diabetes in Asia. Lancet. 2006;368:1681–1688. doi: 10.1016/S0140-6736(06)69703-1.
    1. Apovian CM. Obesity: definition, comorbidities, causes, and burden. Am J Manag Care. 2016;22:s176–s185.
    1. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006;444:840–846. doi: 10.1038/nature05482.
    1. Woo Baidal JA, Lavine JE. The intersection of nonalcoholic fatty liver disease and obesity. Sci Transl Med. 2016;8:323rv321. doi: 10.1126/scitranslmed.aad8390.
    1. Kraschnewski JL, Boan J, Esposito J, Sherwood NE, Lehman EB, Kephart DK, et al. Long-term weight loss maintenance in the United States. Int J Obes. 2010;34:1644–1654. doi: 10.1038/ijo.2010.94.
    1. Svetkey LP, Stevens VJ, Brantley PJ, Appel LJ, Hollis JF, Loria CM, et al. Comparison of strategies for sustaining weight loss: the weight loss maintenance randomized controlled trial. JAMA. 2008;299:1139–1148. doi: 10.1001/jama.299.10.1139.
    1. Wadden TA, Neiberg RH, Wing RR, Clark JM, Delahanty LM, Hill JO, et al. Four-year weight losses in the Look AHEAD study: factors associated with long-term success. Obesity. 2011;19:1987–1998. doi: 10.1038/oby.2011.230.
    1. Look ARG. Eight-year weight losses with an intensive lifestyle intervention: the look AHEAD study. Obesity. 2014;22:5–13. doi: 10.1002/oby.20662.
    1. Langeveld M, DeVries JH. The long-term effect of energy restricted diets for treating obesity. Obesity. 2015;23:1529–1538. doi: 10.1002/oby.21146.
    1. Gursoy A, Erdogan MF, Cin MO, Cesur M, Baskal N. Comparison of orlistat and sibutramine in an obesity management program: efficacy, compliance, and weight regain after noncompliance. Eat Weight Disord. 2006;11:e127–e132. doi: 10.1007/BF03327578.
    1. Odom J, Zalesin KC, Washington TL, Miller WW, Hakmeh B, Zaremba DL, et al. Behavioral predictors of weight regain after bariatric surgery. Obes Surg. 2010;20:349–356. doi: 10.1007/s11695-009-9895-6.
    1. Smith SR, Weissman NJ, Anderson CM, Sanchez M, Chuang E, Stubbe S, et al. Multicenter, placebo-controlled trial of lorcaserin for weight management. N Engl J Med. 2010;363:245–256. doi: 10.1056/NEJMoa0909809.
    1. Sjostrom L, Lindroos AK, Peltonen M, Torgerson J, Bouchard C, Carlsson B, et al. Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med. 2004;351:2683–2693. doi: 10.1056/NEJMoa035622.
    1. Dietz WH, Goodwin NJ, Hill JO, Pi-Sunyer FX, Rolls B, Stern J, et al. Long-term pharmacotherapy in the management of obesity. National Task Force on the prevention and treatment of obesity. JAMA. 1996;276:1907–1915. doi: 10.1001/jama.1996.03540230057036.
    1. Kliewer KL, Ke JY, Lee HY, Stout MB, Cole RM, Samuel VT, et al. Short-term food restriction followed by controlled refeeding promotes gorging behavior, enhances fat deposition, and diminishes insulin sensitivity in mice. J Nutr Biochem. 2015;26:721–728. doi: 10.1016/j.jnutbio.2015.01.010.
    1. Cummings DE, Weigle DS, Frayo RS, Breen PA, Ma MK, Dellinger EP, et al. Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N Engl J Med. 2002;346:1623–1630. doi: 10.1056/NEJMoa012908.
    1. le Roux CW, Welbourn R, Werling M, Osborne A, Kokkinos A, Laurenius A, et al. Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Ann Surg. 2007;246:780–785. doi: 10.1097/SLA.0b013e3180caa3e3.
    1. de Luis DA, Gonzalez Sagrado M, Conde R, Aller R, Izaola O. Decreased basal levels of glucagon-like peptide-1 after weight loss in obese subjects. Ann Nutr Metab. 2007;51:134–138. doi: 10.1159/000103273.
    1. Sumithran P, Prendergast LA, Delbridge E, Purcell K, Shulkes A, Kriketos A, et al. Long-term persistence of hormonal adaptations to weight loss. N Engl J Med. 2011;365:1597–1604. doi: 10.1056/NEJMoa1105816.
    1. Ebbeling CB, Swain JF, Feldman HA, Wong WW, Hachey DL, Garcia-Lago E, et al. Effects of dietary composition on energy expenditure during weight-loss maintenance. JAMA. 2012;307:2627–2634. doi: 10.1001/jama.2012.6607.
    1. Crujeiras AB, Goyenechea E, Abete I, Lage M, Carreira MC, Martinez JA, et al. Weight regain after a diet-induced loss is predicted by higher baseline leptin and lower ghrelin plasma levels. J Clin Endocrinol Metab. 2010;95:5037–5044. doi: 10.1210/jc.2009-2566.
    1. Chaput JP, Pelletier C, Despres JP, Lemieux S, Tremblay A. Metabolic and behavioral vulnerability related to weight regain in reduced-obese men might be prevented by an adequate diet-exercise intervention. Appetite. 2007;49:691–695. doi: 10.1016/j.appet.2007.06.006.
    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;34:2637–2643. doi: 10.1161/ATVBAHA.114.304636.
    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. doi: 10.1161/ATVBAHA.108.165100.
    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;16:276–285. doi: 10.1038/ni.3085.
    1. Hu ZQ, Zhao WH. The IL-33/ST2 axis is specifically required for development of adipose tissue-resident regulatory T cells. Cell Mol Immunol. 2015;12:521–524. doi: 10.1038/cmi.2015.49.
    1. Kong LC, Wuillemin PH, Bastard JP, Sokolovska N, Gougis S, Fellahi S, et al. Insulin resistance and inflammation predict kinetic body weight changes in response to dietary weight loss and maintenance in overweight and obese subjects by using a Bayesian network approach. Am J Clin Nutr. 2013;98:1385–1394. doi: 10.3945/ajcn.113.058099.
    1. Anderson EK, Gutierrez DA, Kennedy A, Hasty AH. Weight cycling increases T-cell accumulation in adipose tissue and impairs systemic glucose tolerance. Diabetes. 2013;62:3180–3188. doi: 10.2337/db12-1076.
    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;15:921–929. doi: 10.1038/nm.2001.
    1. Schmitz J, Evers N, Awazawa M, Nicholls HT, Bronneke HS, Dietrich A, et al. Obesogenic memory can confer long-term increases in adipose tissue but not liver inflammation and insulin resistance after weight loss. Mol Metab. 2016;5:328–339. doi: 10.1016/j.molmet.2015.12.001.
    1. Ambeba EJ, Styn MA, Kuller LH, Brooks MM, Evans RW, Burke LE. Longitudinal effects of weight loss and regain on cytokine concentration of obese adults. Metabolism. 2013;62:1218–1222. doi: 10.1016/j.metabol.2013.04.004.
    1. Maclean PS, Bergouignan A, Cornier MA, Jackman MR. Biology’s response to dieting: the impetus for weight regain. Am J Physiol Regul Integr Comp Physiol. 2011;301:R581–R600. doi: 10.1152/ajpregu.00755.2010.
    1. Mombaerts P, Iacomini J, Johnson RS, Herrup K, Tonegawa S, Papaioannou VE. RAG-1-deficient mice have no mature B and T lymphocytes. Cell. 1992;68:869–877. doi: 10.1016/0092-8674(92)90030-G.
    1. Diehl R, Ferrara F, Muller C, Dreyer AY, McLeod DD, Fricke S, et al. Immunosuppression for in vivo research: state-of-the-art protocols and experimental approaches. Cell Mol Immunol. 2017;14:146–179. doi: 10.1038/cmi.2016.39.
    1. Mombaerts P, Clarke AR, Rudnicki MA, Iacomini J, Itohara S, Lafaille JJ, et al. Mutations in T-cell antigen receptor genes alpha and beta block thymocyte development at different stages. Nature. 1992;360:225–231. doi: 10.1038/360225a0.
    1. Popkin BM, Gordon-Larsen P. The nutrition transition: worldwide obesity dynamics and their determinants. Int J Obes Relat Metab Disord. 2004;28((Suppl 3)):S2–S9. doi: 10.1038/sj.ijo.0802804.
    1. Leibel RL, Rosenbaum M, Hirsch J. Changes in energy expenditure resulting from altered body weight. N Engl J Med. 1995;332:621–628. doi: 10.1056/NEJM199503093321001.
    1. Greenway FL. Physiological adaptations to weight loss and factors favouring weight regain. Int J Obes. 2015;39:1188–1196. doi: 10.1038/ijo.2015.59.
    1. Blomain ES, Dirhan DA, Valentino MA, Kim GW, Waldman SA. Mechanisms of weight regain following weight loss. ISRN Obes. 2013;2013:210524.
    1. Thaiss CA, Itav S, Rothschild D, Meijer M, Levy M, Moresi C, et al. Persistent microbiome alterations modulate the rate of post-dieting weight regain. Nature. 2016;540:544–551. doi: 10.1038/nature20796.
    1. Barbosa-da-Silva S, Fraulob-Aquino JC, Lopes JR, Mandarim-de-Lacerda CA, Aguila MB. Weight cycling enhances adipose tissue inflammatory responses in male mice. PLoS ONE. 2012;7:e39837. doi: 10.1371/journal.pone.0039837.
    1. Strohacker K, McFarlin BK. Influence of obesity, physical inactivity, and weight cycling on chronic inflammation. Front Biosci (Elite Ed) 2010;2:98–104.
    1. Mathis D. Immunological goings-on in visceral adipose tissue. Cell Metab. 2013;17:851–859. doi: 10.1016/j.cmet.2013.05.008.
    1. Vadacca M, Margiotta DP, Navarini L, Afeltra A. Leptin in immuno-rheumatological diseases. Cell Mol Immunol. 2011;8:203–212. doi: 10.1038/cmi.2010.75.
    1. Mauro C, Smith J, Cucchi D, Coe D, Fu H, Bonacina F, et al. Obesity-induced metabolic stress leads to biased effector memory CD4+ T cell differentiation via PI3K p110delta-Akt-mediated signals. Cell Metab. 2017;25:593–609. doi: 10.1016/j.cmet.2017.01.008.
    1. 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. 2010;18:1918–1925. doi: 10.1038/oby.2010.1.
    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;103:467–476. doi: 10.1161/CIRCRESAHA.108.177105.
    1. Zhang H, Xue R, Zhu S, Fu S, Chen Z, Zhou R, et al. M2-specific reduction of CD1d switches NKT cell-mediated immune responses and triggers metaflammation in adipose tissue. Cell Mol Immunol. 2017;14:1–12. doi: 10.1038/cmi.2016.45.
    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;196:3287–3296. doi: 10.4049/jimmunol.1502506.
    1. Ip B, Cilfone NA, Belkina AC, DeFuria J, Jagannathan-Bogdan M, Zhu M, et al. Th17 cytokines differentiate obesity from obesity-associated type 2 diabetes and promote TNFalpha production. Obesity. 2016;24:102–112. doi: 10.1002/oby.21243.
    1. Zhou X, Tang J, Cao H, Fan H, Li B. Tissue resident regulatory T cells: novel therapeutic targets for human disease. Cell Mol Immunol. 2015;12:543–552. doi: 10.1038/cmi.2015.23.
    1. Chen X, Wu Y, Wang L. Fat-resident Tregs: an emerging guard protecting from obesity-associated metabolic disorders. Obes Rev. 2013;14:568–578. doi: 10.1111/obr.12033.
    1. Eller K, Kirsch A, Wolf AM, Sopper S, Tagwerker A, Stanzl U, et al. Potential role of regulatory T cells in reversing obesity-linked insulin resistance and diabetic nephropathy. Diabetes. 2011;60:2954–2962. doi: 10.2337/db11-0358.
    1. Feng J, Li L, Ou Z, Li Q, Gong B, Zhao Z et al. IL-25 stimulates M2 macrophage polarization and thereby promotes mitochondrial respiratory capacity and lipolysis in adipose tissues against obesity. Cell Mol Immunol 2017.
    1. Donnelly JE, Blair SN, Jakicic JM, Manore MM, Rankin JW, Smith BK, et al. Appropriate physical activity intervention strategies for weight loss and prevention of weight regain for adults. Med Sci Sports Exerc. 2009;41:459–471. doi: 10.1249/MSS.0b013e3181949333.
    1. Snel M, van Diepen JA, Stijnen T, Pijl H, Romijn JA, Meinders AE, et al. Immediate and long-term effects of addition of exercise to a 16-week very low calorie diet on low-grade inflammation in obese, insulin-dependent type 2 diabetic patients. Food Chem Toxicol. 2011;49:3104–3111. doi: 10.1016/j.fct.2011.09.032.
    1. You T, Berman DM, Ryan AS, Nicklas BJ. Effects of hypocaloric diet and exercise training on inflammation and adipocyte lipolysis in obese postmenopausal women. J Clin Endocrinol Metab. 2004;89:1739–1746. doi: 10.1210/jc.2003-031310.

Source: PubMed

3
구독하다