Adipose tissue in obesity-related inflammation and insulin resistance: cells, cytokines, and chemokines

Kassem Makki, Philippe Froguel, Isabelle Wolowczuk, Kassem Makki, Philippe Froguel, Isabelle Wolowczuk

Abstract

Adipose tissue is a complex organ that comprises a wide range of cell types with diverse energy storage, metabolic regulation, and neuroendocrine and immune functions. Because it contains various immune cells, either adaptive (B and T lymphocytes; such as regulatory T cells) or innate (mostly macrophages and, more recently identified, myeloid-derived suppressor cells), the adipose tissue is now considered as a bona fide immune organ, at the cross-road between metabolism and immunity. Adipose tissue disorders, such as those encountered in obesity and lipodystrophy, cause alterations to adipose tissue distribution and function with broad effects on cytokine, chemokine, and hormone expression, on lipid storage, and on the composition of adipose-resident immune cell populations. The resulting changes appear to induce profound consequences for basal systemic inflammation and insulin sensitivity. The purpose of this review is to synthesize the current literature on adipose cell composition remodeling in obesity, which shows how adipose-resident immune cells regulate inflammation and insulin resistance-notably through cytokine and chemokine secretion-and highlights major research questions in the field.

Figures

Figure 1
Figure 1
Adipose tissue-resident cells, cytokines, and hormones: role in insulin sensitivity (adapted and updated from [7]).

References

    1. Hotamisligil GS, Erbay E. Nutrient sensing and inflammation in metabolic diseases. Nature Reviews Immunology. 2008;8(12):923–934.
    1. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annual Review of Immunology. 2011;29:415–445.
    1. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860–867.
    1. Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nature Reviews Immunology. 2011;11(2):85–97.
    1. Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance. The Journal of Clinical Investigation. 2006;116(7):1793–1801.
    1. Odegaard JI, Chawla A. Pleiotropic actions of insulin resistance and inflammation in metabolic homeostasis. Science. 2013;339:172–177.
    1. Han JM, Levings MK. Immune regulation in obesity-associated adipose inflammation. The Journal of Immunology. 2013;191:527–532.
    1. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-α: direct role in obesity-linked insulin resistance. Science. 1993;259(5091):87–91.
    1. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372:425–432.
    1. Waki H, Tontonoz P. Endocrine functions of adipose tissue. Annual Review of Pathology. 2007;2:31–56.
    1. Tilg H, Moschen AR. Inflammatory mechanisms in the regulation of insulin resistance. Molecular Medicine. 2008;14(3-4):222–231.
    1. Havel PJ. Update on adipocyte hormones: regulation of energy balance and carbohydrate/lipid metabolism. Diabetes. 2004;53(supplement 1):S143–S151.
    1. Lucas S, Verwaerde C, Wolowczuk I. Is the adipose tissue the key road to inflammation? Immunology and Immunogenetics Insights. 2009;1:3–14.
    1. Chen G, Goeddel DV. TNF-R1 signaling: a beautiful pathway. Science. 2002;296(5573):1634–1635.
    1. Hotamisligil GS, Spiegelman BM. Tumor necrosis factor α: a key component of the obesity-diabetes link. Diabetes. 1994;43(11):1271–1278.
    1. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW., Jr. Obesity is associated with macrophage accumulation in adipose tissue. The Journal of Clinical Investigation. 2003;112(12):1796–1808.
    1. Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS. Protection from obesity-induced insulin resistance in mice lacking TNF-α function. Nature. 1997;389(6651):610–614.
    1. Kern PA, Saghizadeh M, Ong JM, Bosch RJ, Deem R, Simsolo RB. The expression of tumor necrosis factor in human adipose tissue. Regulation by obesity, weight loss, and relationship to lipoprotein lipase. The Journal of Clinical Investigation. 1995;95(5):2111–2119.
    1. Hivert M-F, Sullivan LM, Fox CS, et al. Associations of adiponectin, resistin, and tumor necrosis factor-α with insulin resistance. Journal of Clinical Endocrinology and Metabolism. 2008;93(8):3165–3172.
    1. Dominguez H, Storgaard H, Rask-Madsen C, et al. Metabolic and vascular effects of tumor necrosis factor-α blockade with etanercept in obese patients with type 2 diabetes. Journal of Vascular Research. 2005;42(6):517–525.
    1. Stanley TL, Zanni MV, Johnsen S, et al. TNF-α antagonism with etanercept decreases glucose and increases the proportion of high molecular weight adiponectin in obese subjects with features of the metabolic syndrome. Journal of Clinical Endocrinology and Metabolism. 2011;96(1):E146–E150.
    1. Kanety H, Feinstein R, Papa MZ, Hemi R, Karasik A. Tumor necrosis factor α-induced phosphorylation of insulin receptor substrate-1 (IRS-1). Possible mechanism for suppression of insulin-stimulated tyrosine phosphorylation of IRS-1. The Journal of Biological Chemistry. 1995;270(40):23780–23784.
    1. Fève B, Bastard J-P. The role of interleukins in insulin resistance and type 2 diabetes mellitus. Nature Reviews Endocrinology. 2009;5(6):305–311.
    1. Xu H, Sethi JK, Hotamisligil GS. Transmembrane tumor necrosis factor (TNF)-α inhibits adipocyte differentiation by selectively activating TNF receptor 1. The Journal of Biological Chemistry. 1999;274(37):26287–26295.
    1. Ruan H, Miles PDG, Ladd CM, et al. Profiling gene transcription in vivo reveals adipose tissue as an immediate target of tumor necrosis factor-α: implications for insulin resistance. Diabetes. 2002;51(11):3176–3188.
    1. Hector J, Schwarzloh B, Goehring J, et al. TNF-α alters visfatin and adiponectin levels in human fat. Hormone and Metabolic Research. 2007;39(4):250–255.
    1. Whitehead JP, Richards AA, Hickman IJ, Macdonald GA, Prins JB. Adiponectin—a key adipokine in the metabolic syndrome. Diabetes, Obesity and Metabolism. 2006;8(3):264–280.
    1. Eder K, Baffy N, Falus A, Fulop AK. The major inflammatory mediator interleukin-6 and obesity. Inflammation Research. 2009;58(11):727–736.
    1. Wieckowska A, Papouchado BG, Li Z, Lopez R, Zein NN, Feldstein AE. Increased hepatic and circulating interleukin-6 levels in human nonalcoholic steatohepatitis. American Journal of Gastroenterology. 2008;103(6):1372–1379.
    1. Vozarova B, Weyer C, Hanson K, Tataranni PA, Bogardus C, Pratley RE. Circulating interleukin-6 in relation to adiposity, insulin action, and insulin secretion. Obesity Research. 2001;9(7):414–417.
    1. Bastard J-P, Jardel C, Bruckert E, et al. Elevated levels of interleukin 6 are reduced in serum and subcutaneous adipose tissue of obese women after weight loss. Journal of Clinical Endocrinology and Metabolism. 2000;85(9):3338–3342.
    1. Fried SK, Bunkin DA, Greenberg AS. Omental and subcutaneous adipose tissues of obese subjects release interleukin-6: depot difference and regulation by glucocorticoid. Journal of Clinical Endocrinology and Metabolism. 1998;83(3):847–850.
    1. Hansen D, Dendale P, Beelen M, et al. Plasma adipokine and inflammatory marker concentrations are altered in obese, as opposed to non-obese, type 2 diabetes patients. European Journal of Applied Physiology. 2010;109(3):397–404.
    1. Kern PA, Ranganathan S, Li C, Wood L, Ranganathan G. Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. American Journal of Physiology—Endocrinology and Metabolism. 2001;280(5):E745–E751.
    1. Starkie R, Ostrowski SR, Jauffred S, Febbraio M, Pedersen BK. Exercise and IL-6 infusion inhibit endotoxin-induced TNF-alpha production in humans. The FASEB Journal. 2003;17(8):884–886.
    1. Senn JJ, Klover PJ, Nowak IA, et al. Suppressor of cytokine signaling-3 (SOCS-3), a potential mediator of interleukin-6-dependent insulin resistance in hepatocytes. The Journal of Biological Chemistry. 2003;278(16):13740–13746.
    1. Pricola KL, Kuhn NZ, Haleem-Smith H, Song Y, Tuan RS. Interleukin-6 maintains bone marrow-derived mesenchymal stem cell stemness by an ERK1/2-dependent mechanism. Journal of Cellular Biochemistry. 2009;108(3):577–588.
    1. Ellingsgaard H, Hauselmann I, Schuler B, et al. Interleukin-6 enhances insulin secretion by increasing glucagon-like peptide-1 secretion from L cells and alpha cells. Nature Medicine. 2011;17(11):1481–1489.
    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. The Journal of Biological Chemistry. 2003;278(46):45777–45784.
    1. van Hall G, Steensberg A, Sacchetti M, et al. Interleukin-6 stimulates lipolysis and fat oxidation in humans. Journal of Clinical Endocrinology and Metabolism. 2003;88(7):3005–3010.
    1. Sopasakis VR, Sandqvist M, Gustafson B, et al. High local concentrations and effects on differentiation implicate interleukin-6 as a paracrine regulator. Obesity Research. 2004;12(3):454–460.
    1. Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature. 1998;395(6704):763–770.
    1. Campfield LA, Smith FJ, Guisez Y, Devos R, Burn P. Recombinant mouse OB protein: evidence for a peripheral signal linking adiposity and central neural networks. Science. 1995;269(5223):546–549.
    1. van den Hoek AM, Teusink B, Voshol PJ, Havekes LM, Romijn JA, Pijl H. Leptin deficiency per se dictates body composition and insulin action in ob/ob mice. Journal of Neuroendocrinology. 2008;20(1):120–127.
    1. Zhang H, Xie H, Zhao Q, et al. Relationships between serum adiponectin, apelin, leptin, resistin, visfatin levels and bone mineral density, and bone biochemical markers in post-menopausal Chinese women. Journal of Endocrinological Investigation. 2010;33(10):707–711.
    1. Mori H, Hanada R, Hanada T, et al. Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity. Nature Medicine. 2004;10(7):739–743.
    1. Bence KK, Delibegovic M, Xue B, et al. Neuronal PTP1B regulates body weight, adiposity and leptin action. Nature Medicine. 2006;12(8):917–924.
    1. Zabolotny JM, Bence-Hanulec KK, Stricker-Krongrad A, et al. PTP1B regulates leptin signal transduction in vivo. Developmental Cell. 2002;2(4):489–495.
    1. Macdougald OA, Hwang C-S, Fan H, Lane MD. Regulated expression of the obese gene product (leptin) in white adipose tissue and 3T3-L1 adipocytes. Proceedings of the National Academy of Sciences of the United States of America. 1995;92(20):9034–9037.
    1. Segal KR, Landt M, Klein S. Relationship between insulin sensitivity and plasma leptin concentration in lean and obese men. Diabetes. 1996;45(3):988–991.
    1. Vantyghem MC, Balavoine AS, Douillard C, et al. How to diagnose a lipodystrophy syndrome. Annales d'Endocrinologie . 2012;73:170–189.
    1. Moon HS, Dalamaga M, Kim SY, et al. Leptin's role in lipodystrophic and nonlipodystrophic insulin-resistant and diabetic individuals. Endocrine Reviews. 2013;34:377–412.
    1. Oral EA, Simha V, Ruiz E, et al. Leptin-replacement therapy for lipodystrophy. The New England Journal of Medicine. 2002;346(8):570–578.
    1. Matarese G, Moschos S, Mantzoros CS. Leptin in immunology. The Journal of Immunology. 2005;174(6):3137–3142.
    1. Loffreda S, Yang SQ, Lin HZ, et al. Leptin regulates proinflammatory immune responses. The FASEB Journal. 1998;12(1):57–65.
    1. Landman RE, Puder JJ, Xiao E, Freda PU, Ferin M, Wardlaw SL. Endotoxin stimulates leptin in the human and nonhuman primate. Journal of Clinical Endocrinology and Metabolism. 2003;88(3):1285–1291.
    1. Sachot C, Poole S, Luheshi GN. Circulating leptin mediates lipopolysaccharide-induced anorexia and fever in rats. Journal of Physiology. 2004;561(1):263–272.
    1. Dardenne M, Savino W, Gastinel LN, Nabarra B, Bach JF. Thymic dysfunction in the mutant diabetic (db/db) mouse. The Journal of Immunology. 1983;130(3):1195–1199.
    1. Macia L, Belacre M, Abboud G, et al. Impairment of dendritic cell functionality and steady-state number in obese mice. The Journal of Immunology. 2006;177(9):5997–6006.
    1. Verwaerde C, Delanoye A, Macia L, Tailleux A, Wolowczuk I. Influence of high-fat feeding on both naive and antigen-experienced T-cell immune response in DO10.11 mice. Scandinavian Journal of Immunology. 2006;64(5):457–466.
    1. Agrawal S, Gollapudi S, Su H, Gupta S. Leptin activates human B cells to secrete TNF-α, IL-6, and IL-10 via JAK2/STAT3 and p38MAPK/ERK1/2 signaling pathway. Journal of Clinical Immunology. 2011;31(3):472–478.
    1. Zarkesh-Esfahani H, Pockley G, Metcalfe RA, et al. High-dose leptin activates human leukocytes via receptor expression on monocytes. The Journal of Immunology. 2001;167(8):4593–4599.
    1. Arita Y, Kihara S, Ouchi N, et al. Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochemical and Biophysical Research Communications. 1999;257(1):79–83.
    1. Combs TP, Pajvani UB, Berg AH, et al. A transgenic mouse with a deletion in the collagenous domain of adiponectin displays elevated circulating adiponectin and improved insulin sensitivity. Endocrinology. 2004;145(1):367–383.
    1. Berg AH, Combs TP, Scherer PE. ACRP30/adiponectin: an adipokine regulating glucose and lipid metabolism. Trends in Endocrinology and Metabolism. 2002;13(2):84–89.
    1. Almeda-Valdes P, Cuevas-Ramos D, Mehta R, et al. Total and high molecular weight adiponectin have similar utility for the identification of insulin resistance. Cardiovascular Diabetology. 2010;9, article 26
    1. Tsang JYS, Li D, Ho D, et al. Novel immunomodulatory effects of adiponectin on dendritic cell functions. International Immunopharmacology. 2011;11(5):604–609.
    1. Park P-H, Huang H, McMullen MR, Mandal P, Sun L, Nagy LE. Suppression of lipopolysaccharide-stimulated tumor necrosis factor-α production by adiponectin is mediated by transcriptional and post-transcriptional mechanisms. The Journal of Biological Chemistry. 2008;283(40):26850–26858.
    1. Schwartz DR, Lazar MA. Human resistin: found in translation from mouse to man. Trends in Endocrinology and Metabolism. 2011;22(7):259–265.
    1. Steppan CM, Bailey ST, Bhat S, et al. The hormone resistin links obesity to diabetes. Nature. 2001;409(6818):307–312.
    1. Rajala MW, Qi Y, Patel HR, et al. Regulation of resistin expression and circulating levels in obesity, diabetes, and fasting. Diabetes. 2004;53(7):1671–1679.
    1. Kim K-H, Lee K, Moon YS, Sul HS. A cysteine-rich adipose tissue-specific secretory factor inhibits adipocyte differentiation. The Journal of Biological Chemistry. 2001;276(14):11252–11256.
    1. Palanivel R, Maida A, Liu Y, Sweeney G. Regulation of insulin signalling, glucose uptake and metabolism in rat skeletal muscle cells upon prolonged exposure to resistin. Diabetologia. 2006;49(1):183–190.
    1. Fisher JS. Potential role of the AMP-activated protein kinase in regulation of insulin action. Cell Science. 2006;2:68–81.
    1. Cho YM, Youn B-S, Chung SS, et al. Common genetic polymorphisms in the promoter of resistin gene are major determinants of plasma resistin concentrations in humans. Diabetologia. 2004;47(3):559–565.
    1. Chen XY, Zhang JH, Liu F, Liu HM, Song YY, Liu YL. Association of serum resistin levels with metabolic syndrome and early atherosclerosis in obese Chinese children. Journal of Pediatric Endocrinology and Metabolism. 2013;17:1–6.
    1. Bokarewa M, Nagaev I, Dahlberg L, Smith U, Tarkowski A. Resistin, an adipokine with potent proinflammatory properties. The Journal of Immunology. 2005;174(9):5789–5795.
    1. Kang J, Coles M. IL-7: the global builder of the innate lymphoid network and beyond, one niche at a time. Seminars in Immunology. 2012;24:190–197.
    1. Tian B, Nowak DE, Jamaluddin M, Wang S, Brasier AR. Identification of direct genomic targets downstream of the nuclear factor-κB transcription factor mediating tumor necrosis factor signaling. The Journal of Biological Chemistry. 2005;280(17):17435–17448.
    1. Pellegrini M, Calzascia T, Toe JG, et al. IL-7 engages multiple mechanisms to overcome chronic viral infection and limit organ pathology. Cell. 2011;144(4):601–613.
    1. Rathmell JC, Farkash EA, Gao W, Thompson CB. IL-7 enhances the survival and maintains the size of naive T cells. The Journal of Immunology. 2001;167(12):6869–6876.
    1. Lucas S, Taront S, Magnan C, et al. Interleukin-7 regulates adipose tissue mass and insulin sensitivity in high-fat diet-fed mice through lymphocyte-dependent and independent mechanisms. PLoS One. 2012;7e40351
    1. Maury E, Ehala-Aleksejev K, Guiot Y, Detry R, Vandenhooft A, Brichard SM. Adipokines oversecreted by omental adipose tissue in human obesity. American Journal of Physiology—Endocrinology and Metabolism. 2007;293(3):E656–E665.
    1. Macia L, Viltart O, Delacre M, et al. Interleukin-7, a new cytokine targeting the mouse hypothalamic Arcuate nucleus: role in body weight and food intake regulation. PLoS ONE. 2010;5(4)e9953
    1. Piya MK, McTernan PG, Kumar S. Adipokine inflammation and insulin resistance: the role of glucose, lipids and endotoxin. Journal of Endocrinology. 2013;216:T1–T15.
    1. Bruun JM, Lihn AS, Pedersen SB, Richelsen B. Monocyte chemoattractant protein-1 release is higher in visceral than subcutaneous human adipose tissue (AT): implication of macrophages resident in the AT. Journal of Clinical Endocrinology and Metabolism. 2005;90(4):2282–2289.
    1. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annual Review of Physiology. 2010;72:219–246.
    1. Lim LP, Lau NC, Garrett-Engele P, et al. Microarray analysis shows that some microRNAs downregulate large numbers of-target mRNAs. Nature. 2005;433(7027):769–773.
    1. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–233.
    1. Gauthier BR, Wollheim CB. MicroRNAs: “Ribo-regulators” of glucose homeostasis. Nature Medicine. 2006;12(1):36–38.
    1. Xie H, Lim B, Lodish HF. MicroRNAs induced during adipogenesis that accelerate fat cell development are downregulated in obesity. Diabetes. 2009;58(5):1050–1057.
    1. Lodish HF, Zhou B, Liu G, Chen C-Z. Micromanagement of the immune system by microRNAs. Nature Reviews Immunology. 2008;8(2):120–130.
    1. Zhuang G, Meng C, Guo X, et al. A novel regulator of macrophage activation: miR-223 in obesity-associated adipose tissue inflammation. Circulation. 2012;125:2892–2903.
    1. Kim C-S, Park H-S, Kawada T, et al. Circulating levels of MCP-1 and IL-8 are elevated in human obese subjects and associated with obesity-related parameters. International Journal of Obesity. 2006;30(9):1347–1355.
    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. The Journal of Clinical Investigation. 2006;116(6):1494–1505.
    1. Weisberg SP, Hunter D, Huber R, et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. The Journal of Clinical Investigation. 2006;116(1):115–124.
    1. Inouye KE, Shi H, Howard JK, et al. Absence of CC chemokine ligand 2 does not limit obesity-associated infiltration of macrophages into adipose tissue. Diabetes. 2007;56(9):2242–2250.
    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;61:1680–1690.
    1. Huber J, Kiefer FW, Zeyda M, et al. CC chemokine and CC chemokine receptor profiles in visceral and subcutaneous adipose tissue are altered in human obesity. Journal of Clinical Endocrinology and Metabolism. 2008;93(8):3215–3221.
    1. Lumeng CN, Saltiel AR. Inflammatory links between obesity and metabolic disease. The Journal of Clinical Investigation. 2011;121(6):2111–2117.
    1. Deiuliis J, Shah Z, Shah N, et al. Visceral adipose inflammation in obesity is associated with critical alterations in tregulatory cell numbers. PLoS ONE. 2011;6(1)e16376
    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. Talukdar S, Oh da Y, Bandyopadhyay G, et al. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nature Medicine. 2012;18:1407–1412.
    1. Maier T, Holda JH, Claman HN. Natural suppressor cells. Progress in Clinical and Biological Research. 1989;288:235–244.
    1. Strober S. Natural suppressor (NS) cells, neonatal tolerance, and total lymphoid irradiation: exploring obscure relationships. Annual Review of Immunology. 1984;2:219–237.
    1. Gabrilovich DI, Bronte V, Chen S-H, et al. The terminology issue for myeloid-derived suppressor cells. Cancer Research. 2007;67(1):p. 425.
    1. Nagaraj S, Gabrilovich DI. Myeloid-derived suppressor cells in human cancer. Cancer Journal. 2010;16(4):348–353.
    1. Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. The Journal of Immunology. 2001;166(9):5398–5406.
    1. Zhou Z, French DL, Ma G, et al. Development and function of myeloid-derived suppressor cells generated from mouse embryonic and hematopoietic stem cells. Stem Cells. 2010;28(3):620–632.
    1. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nature Reviews Immunology. 2009;9(3):162–174.
    1. Khaled YS, Ammori BJ, Elkord E. Myeloid-derived suppressor cells in cancer: recent progress and prospects. Immunology and Cell Biology. 2013;91:493–502.
    1. Wu L, Yan C, Czader M, et al. Inhibition of PPARγ in myeloid-lineage cells induces systemic inflammation, immunosuppression, and tumorigenesis. Blood. 2012;119(1):115–126.
    1. Shojaei F, Ferrara N. Role of the microenvironment in tumor growth and in refractoriness/resistance to anti-angiogenic therapies. Drug Resistance Updates. 2008;11(6):219–230.
    1. Hock BD, MacKenzie KA, Cross NB, et al. Renal transplant recipients have elevated frequencies of circulating myeloid-derived suppressor cells. Nephrology Dialysis Transplantation. 2012;27(1):402–410.
    1. Xia S, Sha H, Yang L, Ji Y, Ostrand-Rosenberg S, Qi L. Gr-1+ CD11b+ myeloid-derived suppressor cells suppress inflammation and promote insulin sensitivity in obesity. The Journal of Biological Chemistry. 2011;286(26):23591–23599.
    1. Yin B, Ma G, Yen C-Y, et al. Myeloid-derived suppressor cells prevent type 1 diabetes in murine models. The Journal of Immunology. 2010;185(10):5828–5834.
    1. Martelli AM, Chiarini F, Evangelisti C, et al. The phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin signaling network and the control of normal myelopoiesis. Histology and Histopathology. 2010;25(5):669–680.
    1. Lu H, Huang D, Saederup N, Charo IF, Ransohoff RM, Zhou L. Macrophages recruited via CCR2 produce insulin-like growth factor-1 to repair acute skeletal muscle injury. The FASEB Journal. 2011;25(1):358–369.

Source: PubMed

3
Sottoscrivi