Role of T cells in malnutrition and obesity

Valerie A Gerriets, Nancie J MacIver, Valerie A Gerriets, Nancie J MacIver

Abstract

Nutritional status is critically important for immune cell function. While obesity is characterized by inflammation that promotes metabolic syndrome including cardiovascular disease and insulin resistance, malnutrition can result in immune cell defects and increased risk of mortality from infectious diseases. T cells play an important role in the immune adaptation to both obesity and malnutrition. T cells in obesity have been shown to have an early and critical role in inducing inflammation, accompanying the accumulation of inflammatory macrophages in obese adipose tissue, which are known to promote insulin resistance. How T cells are recruited to adipose tissue and activated in obesity is a topic of considerable interest. Conversely, T cell number is decreased in malnourished individuals, and T cells in the setting of malnutrition have decreased effector function and proliferative capacity. The adipokine leptin, which is secreted in proportion to adipocyte mass, may have a key role in mediating adipocyte-T cell interactions in both obesity and malnutrition, and has been shown to promote effector T cell function and metabolism while inhibiting regulatory T cell proliferation. Additionally, key molecular signals are involved in T cell metabolic adaptation during nutrient stress; among them, the metabolic regulator AMP kinase and the mammalian target of rapamycin have critical roles in regulating T cell number, function, and metabolism. In summary, understanding how T cell number and function are altered in obesity and malnutrition will lead to better understanding of and treatment for diseases where nutritional status determines clinical outcome.

Keywords: T cells; inflammation; leptin; malnutrition; obesity.

Figures

Figure 1
Figure 1
Adipose tissue is remodeled following high-fat diet-induced obesity. Multiple immune cells reside in adipose tissue. High-fat diet-induced obesity results in decreased regulatory T cells (Treg) and alternatively activated M2 macrophages. At the same time, there is an increase in adipose-resident inflammatory macrophages (classically activated M1 macrophages), CD4+ Th1 cells, CD8+ T cells, and B cells. The role of CD4+ Th2 and Th17 cells in adipose tissue in obesity is less certain.
Figure 2
Figure 2
Malnutrition leads to changes in adipocyte-secreted hormones and T cell number and function. Malnutrition leads to decreased adipocyte mass, which results in decreased circulating leptin. Concurrently, malnourished individuals show changes in CD4+ and CD8+ T cell number and function, leading to increased susceptibility to infection and protection against certain forms of autoimmunity. Leptin may mediate a subset of these malnutrition-related changes to T cell number and function, as leptin is known to alter CD4+ T cell number and cytokine production.
Figure 3
Figure 3
Nutritional status is critical for normal immune function. Malnutrition reduces immunity and increases markedly the risks and mortality from severe infections, whereas obesity heightens immune reactivity and predisposes to systemic inflammation that fosters the development of inflammatory disorders and insulin resistance leading to type 2 diabetes mellitus.

References

    1. Hedley AA, Ogden CL, Johnson CL, Carroll MD, Curtin LR, Flegal KM. Prevalence of overweight and obesity among US children, adolescents, and adults, 1999–2002. JAMA (2004) 291:2847–5010.1001/jama.291.23.2847
    1. Li Z, Bowerman S, Heber D. Health ramifications of the obesity epidemic. Surg Clin North Am (2005) 85:681–70110.1016/j.suc.2005.04.006
    1. Mokdad AH, Bowman BA, Ford ES, Vinicor F, Marks JS, Koplan JP. The continuing epidemics of obesity and diabetes in the United States. JAMA (2001) 286:1195–20010.1001/jama.286.10.1195
    1. Boyle JP, Honeycutt AA, Narayan KM, Hoerger TJ, Geiss LS, Chen H, et al. Projection of diabetes burden through 2050: impact of changing demography and disease prevalence in the U.S. Diabetes Care (2001) 24:1936–4010.2337/diacare.24.11.1936
    1. Harpsoe MC, Basit S, Andersson M, Nielsen NM, Frisch M, Wohlfahrt J, et al. Body mass index and risk of autoimmune diseases: a study within the Danish National Birth Cohort. Int J Epidemiol (2014) 43:843–5510.1093/ije/dyu045
    1. Duntas LH, Biondi B. The interconnections between obesity, thyroid function, and autoimmunity: the multifold role of leptin. Thyroid (2013) 23:646–5310.1089/thy.2011.0499
    1. Lukens JR, Dixit VD, Kanneganti TD. Inflammasome activation in obesity-related inflammatory diseases and autoimmunity. Discov Med (2011) 12:65–74
    1. Schottenfeld D, Beebe-Dimmer JL, Buffler PA, Omenn GS. Current perspective on the global and United States cancer burden attributable to lifestyle and environmental risk factors. Annu Rev Public Health (2013) 34:97–11710.1146/annurev-publhealth-031912-114350
    1. Wolin KY, Carson K, Colditz GA. Obesity and cancer. Oncologist (2010) 15:556–6510.1634/theoncologist.2009-0285
    1. Falagas ME, Kompoti M. Obesity and infection. Lancet Infect Dis (2006) 6:438–4610.1016/S1473-3099(06)70523-0
    1. Milner JJ, Beck MA. The impact of obesity on the immune response to infection. Proc Nutr Soc (2012) 71:298–30610.1017/S0029665112000158
    1. Shu CJ, Benoist C, Mathis D. The immune system’s involvement in obesity-driven type 2 diabetes. Semin Immunol (2012) 24:436–4210.1016/j.smim.2012.12.001
    1. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol (2011) 29:415–4510.1146/annurev-immunol-031210-101322
    1. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science (1993) 259:87–9110.1126/science.7678183
    1. Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS. Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function. Nature (1997) 389:610–410.1038/39335
    1. Ventre J, Doebber T, Wu M, MacNaul K, Stevens K, Pasparakis M, et al. Targeted disruption of the tumor necrosis factor-alpha gene: metabolic consequences in obese and nonobese mice. Diabetes (1997) 46:1526–3110.2337/diabetes.46.9.1526
    1. Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA (2001) 286:327–3410.1001/jama.286.3.327
    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) 112:1821–3010.1172/JCI19451
    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) 112:1796–80810.1172/JCI19246
    1. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest (2007) 117:175–8410.1172/JCI29881
    1. Lumeng CN, Deyoung SM, Bodzin JL, Saltiel AR. Increased inflammatory properties of adipose tissue macrophages recruited during diet-induced obesity. Diabetes (2007) 56:16–2310.2337/db06-1076
    1. Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, Long JM, et al. IKK-beta links inflammation to obesity-induced insulin resistance. Nat Med (2005) 11:191–810.1038/nm1185
    1. Hirosumi J, Tuncman G, Chang L, Gorgun CZ, Uysal KT, Maeda K, et al. A central role for JNK in obesity and insulin resistance. Nature (2002) 420:333–610.1038/nature01137
    1. Osborn O, Olefsky JM. The cellular and signaling networks linking the immune system and metabolism in disease. Nat Med (2012) 18:363–7410.1038/nm.2627
    1. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol (2010) 72:219–4610.1146/annurev-physiol-021909-135846
    1. Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Invest (2005) 115:1111–910.1172/JCI25102
    1. Christiansen T, Richelsen B, Bruun JM. Monocyte chemoattractant protein-1 is produced in isolated adipocytes, associated with adiposity and reduced after weight loss in morbid obese subjects. Int J Obes (Lond) (2005) 29:146–5010.1038/sj.ijo.0802839
    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 Metab (2008) 8:301–910.1016/j.cmet.2008.08.015
    1. Huh JY, Park YJ, Ham M, Kim JB. Crosstalk between adipocytes and immune cells in adipose tissue inflammation and metabolic dysregulation in obesity. Mol Cells (2014) 37:365–7110.14348/molcells.2014.0074
    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) 15:914–2010.1038/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) 15:930–910.1038/nm.2002
    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) 15:921–910.1038/nm.2001
    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) 185:1836–4510.4049/jimmunol.1000021
    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) 103:467–7610.1161/CIRCRESAHA.108.177105
    1. O’Rourke RW, White AE, Metcalf MD, Winters BR, Diggs BS, Zhu X, et al. Systemic inflammation and insulin sensitivity in obese IFN-gamma knockout mice. Metabolism (2012) 61:1152–6110.1016/j.metabol.2012.01.018
    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) 233:419–2810.1016/j.atherosclerosis.2014.01.011
    1. Stolarczyk E, Vong CT, Perucha E, Jackson I, Cawthorne MA, Wargent ET, et al. Improved insulin sensitivity despite increased visceral adiposity in mice deficient for the immune cell transcription factor T-bet. Cell Metab (2013) 17:520–3310.1016/j.cmet.2013.02.019
    1. Kim KY, Jeong HJ, Kim HM. The role of T-bet in obesity: lack of T-bet causes obesity in male mice. J Nutr Biochem (2013) 24:240–710.1016/j.jnutbio.2012.05.010
    1. Stolarczyk E, Lord GM, Howard JK. The immune cell transcription factor T-bet: a novel metabolic regulator. Adipocyte (2014) 3:58–6210.4161/adip.26220
    1. Winer S, Paltser G, Chan Y, Tsui H, Engleman E, Winer D, et al. Obesity predisposes to Th17 bias. Eur J Immunol (2009) 39:2629–3510.1002/eji.200838893
    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 (Lond) (2009) 33:151–610.1038/ijo.2008.216
    1. Jagannathan-Bogdan M, McDonnell ME, Shin H, Rehman Q, Hasturk H, Apovian CM, et al. Elevated proinflammatory cytokine production by a skewed T cell compartment requires monocytes and promotes inflammation in type 2 diabetes. J Immunol (2011) 186:1162–7210.4049/jimmunol.1002615
    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):e1–310.1053/j.gastro.2013.04.010
    1. Ahmed M, Gaffen SL. IL-17 in obesity and adipogenesis. Cytokine Growth Factor Rev (2010) 21:449–5310.1016/j.cytogfr.2010.10.005
    1. Zuniga LA, Shen WJ, Joyce-Shaikh B, Pyatnova EA, Richards AG, Thom C, et al. IL-17 regulates adipogenesis, glucose homeostasis, and obesity. J Immunol (2010) 185:6947–5910.4049/jimmunol.1001269
    1. Pini M, Fantuzzi G. Enhanced production of IL-17A during zymosan- induced peritonitis in obese mice. J Leukoc Biol (2010) 87:51–810.1189/jlb.0309188
    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 (Lond) (2008) 32:451–6310.1038/sj.ijo.0803744
    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) 34:34–4310.1161/ATVBAHA.113.302077
    1. Winer DA, Winer S, Chng MH, Shen L, Engleman EG. B Lymphocytes in obesity-related adipose tissue inflammation and insulin resistance. Cell Mol Life Sci (2014) 71:1033–4310.1007/s00018-013-1486-y
    1. Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med (2011) 17:610–710.1038/nm.2353
    1. DeFuria J, Belkina AC, Jagannathan-Bogdan M, Snyder-Cappione J, Carr JD, Nersesova YR, et al. B cells promote inflammation in obesity and type 2 diabetes through regulation of T-cell function and an inflammatory cytokine profile. Proc Natl Acad Sci U S A (2013) 110:5133–810.1073/pnas.1215840110
    1. Wagner NM, Brandhorst G, Czepluch F, Lankeit M, Eberle C, Herzberg S, et al. Circulating regulatory T cells are reduced in obesity and may identify subjects at increased metabolic and cardiovascular risk. Obesity (Silver Spring) (2013) 21:461–810.1002/oby.20087
    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) 486:549–5310.1038/nature11132
    1. Han JM, Patterson SJ, Speck M, Ehses JA, Levings MK. Insulin inhibits IL-10-mediated regulatory T cell function: implications for obesity. J Immunol (2014) 192:623–910.4049/jimmunol.1302181
    1. Rocha VZ, Folco EJ, Ozdemir C, Sheikine Y, Christen T, Sukhova GK, et al. CXCR3 controls T-cell accumulation in fat inflammation. Arterioscler Thromb Vasc Biol (2014) 34:1374–8110.1161/ATVBAHA.113.303133
    1. Deiuliis JA, Oghumu S, Duggineni D, Zhong J, Rutsky J, Banerjee A, et al. CXCR3 modulates obesity-induced visceral adipose inflammation and systemic insulin resistance. Obesity (Silver Spring) (2014) 22:1264–7410.1002/oby.20642
    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) 115:1029–3810.1161/CIRCULATIONAHA.106.638379
    1. Kitade H, Sawamoto K, Nagashimada M, Inoue H, Yamamoto Y, Sai Y, 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) 61:1680–9010.2337/db11-1506
    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) 62:2762–7210.2337/db12-1404
    1. Ioan-Facsinay A, Kwekkeboom JC, Westhoff S, Giera M, Rombouts Y, van Harmelen V, et al. Adipocyte-derived lipids modulate CD4+ T-cell function. Eur J Immunol (2013) 43:1578–8710.1002/eji.201243096
    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) 17:411–2210.1016/j.cmet.2013.02.009
    1. Samaras K, Viardot A, Botelho NK, Jenkins A, Lord RV. Immune cell-mediated inflammation and the early improvements in glucose metabolism after gastric banding surgery. Diabetologia (2013) 56:2564–7210.1007/s00125-013-3033-7
    1. Kawanishi N, Mizokami T, Yano H, Suzuki K. Exercise attenuates M1 macrophages and CD8+ T cells in the adipose tissue of obese mice. Med Sci Sports Exerc (2013) 45:1684–9310.1249/MSS.0b013e31828ff9c6
    1. Lasselin J, Magne E, Beau C, Ledaguenel P, Dexpert S, Aubert A, et al. Adipose inflammation in obesity: relationship with circulating levels of inflammatory markers and association with surgery-induced weight loss. J Clin Endocrinol Metab (2014) 99:E53–6110.1210/jc.2013-2673
    1. Dominguez H, Storgaard H, Rask-Madsen C, Steffen Hermann T, Ihlemann N, Baunbjerg Nielsen D, et al. Metabolic and vascular effects of tumor necrosis factor-alpha blockade with etanercept in obese patients with type 2 diabetes. J Vasc Res (2005) 42:517–2510.1159/000088261
    1. Gonzalez-Gay MA, De Matias JM, Gonzalez-Juanatey C, Garcia-Porrua C, Sanchez-Andrade A, Martin J, et al. Anti-tumor necrosis factor-alpha blockade improves insulin resistance in patients with rheumatoid arthritis. Clin Exp Rheumatol (2006) 24:83–6
    1. Stanley TL, Zanni MV, Johnsen S, Rasheed S, Makimura H, Lee H, et al. TNF-alpha antagonism with etanercept decreases glucose and increases the proportion of high molecular weight adiponectin in obese subjects with features of the metabolic syndrome. J Clin Endocrinol Metab (2011) 96:E146–5010.1210/jc.2010-1170
    1. Yuan M, Konstantopoulos N, Lee J, Hansen L, Li ZW, Karin M, et al. Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of IKKbeta. Science (2001) 293:1673–710.1126/science.1061620
    1. Kim JK, Kim YJ, Fillmore JJ, Chen Y, Moore I, Lee J, et al. Prevention of fat-induced insulin resistance by salicylate. J Clin Invest (2001) 108:437–4610.1172/JCI200111559
    1. Kim MS, Yamamoto Y, Kim K, Kamei N, Shimada T, Liu L, et al. Regulation of diet-induced adipose tissue and systemic inflammation by salicylates and pioglitazone. PLoS One (2013) 8:e82847.10.1371/journal.pone.0082847
    1. Goldfine AB, Conlin PR, Halperin F, Koska J, Permana P, Schwenke D, et al. A randomised trial of salsalate for insulin resistance and cardiovascular risk factors in persons with abnormal glucose tolerance. Diabetologia (2013) 56:714–2310.1007/s00125-012-2819-3
    1. Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, et al. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med (2007) 356:1517–2610.1056/NEJMoa065213
    1. Montes VN, Turner MS, Subramanian S, Ding Y, Hayden-Ledbetter M, Slater S, et al. T cell activation inhibitors reduce CD8+ T cell and pro-inflammatory macrophage accumulation in adipose tissue of obese mice. PLoS One (2013) 8:e67709.10.1371/journal.pone.0067709
    1. Black RE, Victora CG, Walker SP, Bhutta ZA, Christian P, de Onis M, et al. Maternal and child undernutrition and overweight in low-income and middle-income countries. Lancet (2013) 382:427–5110.1016/S0140-6736(13)60937-X
    1. Neumann CG, Gewa C, Bwibo NO. Child nutrition in developing countries. Pediatr Ann (2004) 33:658–7410.3928/0090-4481-20041001-09
    1. Hickman D, Jones MK, Zhu S, Kirkpatrick E, Ostrov DA, Wang X, et al. The effect of malnutrition on norovirus infection. MBio (2014) 5:e01032–1310.1128/mBio.01032-13
    1. Caulfield LE, de Onis M, Blossner M, Black RE. Undernutrition as an underlying cause of child deaths associated with diarrhea, pneumonia, malaria, and measles. Am J Clin Nutr (2004) 80:193–8
    1. Laus MF, Vales LD, Costa TM, Almeida SS. Early postnatal protein-calorie malnutrition and cognition: a review of human and animal studies. Int J Environ Res Public Health (2011) 8:590–61210.3390/ijerph8020590
    1. Kar BR, Rao SL, Chandramouli BA. Cognitive development in children with chronic protein energy malnutrition. Behav Brain Funct (2008) 4:31.10.1186/1744-9081-4-31
    1. Antwi A. Assessment and management of severe malnutrition in children. West Afr J Med (2011) 30:11–8
    1. Schlaudecker EP, Steinhoff MC, Moore SR. Interactions of diarrhea, pneumonia, and malnutrition in childhood: recent evidence from developing countries. Curr Opin Infect Dis (2011) 24:496–50210.1097/QCO.0b013e328349287d
    1. Dewys WD, Begg C, Lavin PT, Band PR, Bennett JM, Bertino JR, et al. Prognostic effect of weight loss prior to chemotherapy in cancer patients. Eastern Cooperative Oncology Group. Am J Med (1980) 69:491–7
    1. von Haehling S, Anker SD. Cachexia as major underestimated unmet medical need: facts and numbers. Int J Cardiol (2012) 161:121–310.1016/j.ijcard.2012.09.213
    1. Howard JK, Lord GM, Matarese G, Vendetti S, Ghatei MA, Ritter MA, et al. Leptin protects mice from starvation-induced lymphoid atrophy and increases thymic cellularity in ob/ob mice. J Clin Invest (1999) 104:1051–910.1172/JCI6762
    1. Procaccini C, De Rosa V, Galgani M, Carbone F, Cassano S, Greco D, et al. Leptin-induced mTOR activation defines a specific molecular and transcriptional signature controlling CD4+ effector T cell responses. J Immunol (2012) 189:2941–5310.4049/jimmunol.1200935
    1. Saucillo DC, Gerriets VA, Sheng J, Rathmell JC, Maciver NJ. Leptin metabolically licenses T cells for activation to link nutrition and immunity. J Immunol (2014) 192:136–4410.4049/jimmunol.1301158
    1. Pena-Cruz V, Reiss CS, McIntosh K. Sendai virus infection of mice with protein malnutrition. J Virol (1989) 63:3541–4
    1. Taylor AK, Cao W, Vora KP, De La Cruz J, Shieh WJ, Zaki SR, et al. Protein energy malnutrition decreases immunity and increases susceptibility to influenza infection in mice. J Infect Dis (2013) 207:501–1010.1093/infdis/jis527
    1. Najera O, Gonzalez C, Toledo G, Lopez L, Ortiz R. Flow cytometry study of lymphocyte subsets in malnourished and well-nourished children with bacterial infections. Clin Diagn Lab Immunol (2004) 11:577–80
    1. Mengheri E, Nobili F, Crocchioni G, Lewis JA. Protein starvation impairs the ability of activated lymphocytes to produce interferon-gamma. J Interferon Res (1992) 12:17–2110.1089/jir.1992.12.17
    1. Gonzalez-Torres C, Gonzalez-Martinez H, Miliar A, Najera O, Graniel J, Firo V, et al. Effect of malnutrition on the expression of cytokines involved in Th1 cell differentiation. Nutrients (2013) 5:579–9310.3390/nu5020579
    1. Gonzalez-Martinez H, Rodriguez L, Najera O, Cruz D, Miliar A, Dominguez A, et al. Expression of cytokine mRNA in lymphocytes of malnourished children. J Clin Immunol (2008) 28:593–910.1007/s10875-008-9204-5
    1. Jacobs SR, Herman CE, Maciver NJ, Wofford JA, Wieman HL, Hammen JJ, et al. Glucose uptake is limiting in T cell activation and requires CD28-mediated AKT-dependent and independent pathways. J Immunol (2008) 180:4476–8610.4049/jimmunol.180.7.4476
    1. Wang R, Dillon CP, Shi LZ, Milasta S, Carter R, Finkelstein D, et al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity (2011) 35:871–8210.1016/j.immuni.2011.09.021
    1. Cham CM, Gajewski TF. Glucose availability regulates IFN-gamma production and p70S6 kinase activation in CD8+ effector T cells. J Immunol (2005) 174:4670–710.4049/jimmunol.174.8.4670
    1. Xia CQ, Chernatynskaya AV, Looney B, Wan S, Clare-Salzler MJ. Anti-CD3 antibody treatment induces hypoglycemia and super tolerance to glucose challenge in mice through enhancing glucose consumption by activated lymphocytes. J Immunol Res (2014) 2014:326708.10.1155/2014/326708
    1. Savy M, Edmond K, Fine PE, Hall A, Hennig BJ, Moore SE, et al. Landscape analysis of interactions between nutrition and vaccine responses in children. J Nutr (2009) 139:2154S–218S10.3945/jn.109.105312
    1. Iyer SS, Chatraw JH, Tan WG, Wherry EJ, Becker TC, Ahmed R, et al. Protein energy malnutrition impairs homeostatic proliferation of memory CD8 T cells. J Immunol (2012) 188:77–8410.4049/jimmunol.1004027
    1. Chatraw JH, Wherry EJ, Ahmed R, Kapasi ZF. Diminished primary CD8 T cell response to viral infection during protein energy malnutrition in mice is due to changes in microenvironment and low numbers of viral-specific CD8 T cell precursors. J Nutr (2008) 138:806–12
    1. Chan J, Tian Y, Tanaka KE, Tsang MS, Yu K, Salgame P, et al. Effects of protein calorie malnutrition on tuberculosis in mice. Proc Natl Acad Sci U S A (1996) 93:14857–6110.1073/pnas.93.25.14857
    1. Najera O, Gonzalez C, Toledo G, Lopez L, Cortes E, Betancourt M, et al. CD45RA and CD45RO isoforms in infected malnourished and infected well-nourished children. Clin Exp Immunol (2001) 126:461–510.1046/j.1365-2249.2001.01694.x
    1. Murphy ED, Roths JB. A Y chromosome associated factor in strain BXSB producing accelerated autoimmunity and lymphoproliferation. Arthritis Rheum (1979) 22:1188–9410.1002/art.1780221105
    1. Kubo C, Gajar A, Johnson BC, Good RA. The effects of dietary restriction on immune function and development of autoimmune disease in BXSB mice. Proc Natl Acad Sci U S A (1992) 89:3145–910.1073/pnas.89.7.3145
    1. Piccio L, Stark JL, Cross AH. Chronic calorie restriction attenuates experimental autoimmune encephalomyelitis. J Leukoc Biol (2008) 84:940–810.1189/jlb.0208133
    1. Sanna V, Di Giacomo A, La Cava A, Lechler RI, Fontana S, Zappacosta S, et al. Leptin surge precedes onset of autoimmune encephalomyelitis and correlates with development of pathogenic T cell responses. J Clin Invest (2003) 111:241–5010.1172/JCI200316721
    1. Esquifino AI, Cano P, Jimenez V, Cutrera RA, Cardinali DP. Experimental allergic encephalomyelitis in male Lewis rats subjected to calorie restriction. J Physiol Biochem (2004) 60:245–5210.1007/BF03167069
    1. Kafami L, Raza M, Razavi A, Mirshafiey A, Movahedian M, Khorramizadeh MR. Intermittent feeding attenuates clinical course of experimental autoimmune encephalomyelitis in C57BL/6 mice. Avicenna J Med Biotechnol (2010) 2:47–52
    1. Procaccini C, Jirillo E, Matarese G. Leptin as an immunomodulator. Mol Aspects Med (2012) 33:35–4510.1016/j.mam.2011.10.012
    1. Ahima RS, Prabakaran D, Mantzoros C, Qu D, Lowell B, Maratos-Flier E, et al. Role of leptin in the neuroendocrine response to fasting. Nature (1996) 382:250–210.1038/382250a0
    1. Boden G, Chen X, Mozzoli M, Ryan I. Effect of fasting on serum leptin in normal human subjects. J Clin Endocrinol Metab (1996) 81:3419–2310.1210/jc.81.9.3419
    1. Bartz S, Mody A, Hornik C, Bain J, Muehlbauer M, Kiyimba T, et al. Severe acute malnutrition in childhood: hormonal and metabolic status at presentation, response to treatment, and predictors of mortality. J Clin Endocrinol Metab (2014) 99(6):2128–3710.1210/jc.2013-4018
    1. Harris RB, Zhou J, Redmann SM, Jr., Smagin GN, Smith SR, Rodgers E, et al. A leptin dose-response study in obese (ob/ob) and lean (+/?) mice. Endocrinology (1998) 139:8–1910.1210/endo.139.1.5675
    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) 394:897–90110.1038/29795
    1. White SJ, Taylor MJ, Hurt RT, Jensen MD, Poland GA. Leptin-based adjuvants: an innovative approach to improve vaccine response. Vaccine (2013) 31:1666–7210.1016/j.vaccine.2013.01.032
    1. Rodriguez L, Graniel J, Ortiz R. Effect of leptin on activation and cytokine synthesis in peripheral blood lymphocytes of malnourished infected children. Clin Exp Immunol (2007) 148:478–8510.1111/j.1365-2249.2007.03361.x
    1. Cauchard S, Bermudez-Humaran LG, Blugeon S, Laugier C, Langella P, Cauchard J. Mucosal co-immunization of mice with recombinant lactococci secreting VapA antigen and leptin elicits a protective immune response against Rhodococcus equi infection. Vaccine (2011) 30:95–10210.1016/j.vaccine.2011.10.026
    1. Wehrens A, Aebischer T, Meyer TF, Walduck AK. Leptin receptor signaling is required for vaccine-induced protection against Helicobacter pylori. Helicobacter (2008) 13:94–10210.1111/j.1523-5378.2008.00591.x
    1. Wang B, Chandrasekera PC, Pippin JJ. Leptin- and leptin receptor-deficient rodent models: relevance for human type 2 diabetes. Curr Diabetes Rev (2014) 10:131–4510.2174/1573399810666140508121012
    1. Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, DePinho RA, et al. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci U S A (2004) 101:3329–3510.1073/pnas.0308061100
    1. Tamas P, Hawley SA, Clarke RG, Mustard KJ, Green K, Hardie DG, et al. Regulation of the energy sensor AMP-activated protein kinase by antigen receptor and Ca2+ in T lymphocytes. J Exp Med (2006) 203:1665–7010.1084/jem.20052469
    1. MacIver NJ, Blagih J, Saucillo DC, Tonelli L, Griss T, Rathmell JC, et al. The liver kinase B1 is a central regulator of T cell development, activation, and metabolism. J Immunol (2011) 187:4187–9810.4049/jimmunol.1100367
    1. Tamas P, Macintyre A, Finlay D, Clarke R, Feijoo-Carnero C, Ashworth A, et al. LKB1 is essential for the proliferation of T-cell progenitors and mature peripheral T cells. Eur J Immunol (2010) 40:242–5310.1002/eji.200939677
    1. Cao Y, Li H, Liu H, Zheng C, Ji H, Liu X. The serine/threonine kinase LKB1 controls thymocyte survival through regulation of AMPK activation and Bcl-XL expression. Cell Res (2010) 20:99–10810.1038/cr.2009.141
    1. Delgoffe GM, Kole TP, Zheng Y, Zarek PE, Matthews KL, Xiao B, et al. The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity (2009) 30:832–4410.1016/j.immuni.2009.04.014
    1. Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, et al. The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nat Immunol (2011) 12:295–30310.1038/ni.2005
    1. Zeng H, Yang K, Cloer C, Neale G, Vogel P, Chi H. mTORC1 couples immune signals and metabolic programming to establish T(reg)-cell function. Nature (2013) 499:485–9010.1038/nature12297
    1. Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, et al. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol (2011) 186:3299–30310.4049/jimmunol.1003613

Source: PubMed

3
Subskrybuj