Abstract

HomeArteriosclerosis, Thrombosis, and Vascular BiologyVol. 28, No. 7Adipose Tissue Lymphocytes and Macrophages in Obesity and Insulin Resistance Free AccessEditorialPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessEditorialPDF/EPUBAdipose Tissue Lymphocytes and Macrophages in Obesity and Insulin ResistanceMakers or Markers, and Which Comes First? A. Bouloumié, L. Casteilla and M. Lafontan A. BouloumiéA. Bouloumié From the Institut National de la Santé et de la Recherche Médicale (INSERM) (A.B., M.L.), Unité 858 & Université Toulouse III Paul Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, IFR31, BP84225, 31432 Toulouse, France; and UMR 5241 CNRS UPS IFR31 (L.C.), IFR109 Institut Louis Bugnard, BP 84225, 31432 Toulouse Cedex 4, France. Search for more papers by this author , L. CasteillaL. Casteilla From the Institut National de la Santé et de la Recherche Médicale (INSERM) (A.B., M.L.), Unité 858 & Université Toulouse III Paul Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, IFR31, BP84225, 31432 Toulouse, France; and UMR 5241 CNRS UPS IFR31 (L.C.), IFR109 Institut Louis Bugnard, BP 84225, 31432 Toulouse Cedex 4, France. Search for more papers by this author and M. LafontanM. Lafontan From the Institut National de la Santé et de la Recherche Médicale (INSERM) (A.B., M.L.), Unité 858 & Université Toulouse III Paul Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, IFR31, BP84225, 31432 Toulouse, France; and UMR 5241 CNRS UPS IFR31 (L.C.), IFR109 Institut Louis Bugnard, BP 84225, 31432 Toulouse Cedex 4, France. Search for more papers by this author Originally published1 Jul 2008https://doi.org/10.1161/ATVBAHA.108.168229Arteriosclerosis, Thrombosis, and Vascular Biology. 2008;28:1211–1213Obesity, and more specifically accumulation of adipose tissue in the visceral and subcutaneous abdominal locations, is a major risk factor for the development of cardiovascular pathologies including hypertension and atherosclerosis, as well as metabolic disorders such as type 2 diabetes. During recent years, “metaflammation” or metabolically-triggered inflammation1 has emerged as a key process involved in the clustering of those conditions. Although several metabolically active organs such as the liver, muscle, and, recently, the intestine2 certainly play major roles, the white adipose tissue appears as a central and primary player as both a source and site of inflammation. Accumulation of adipose tissue macrophages (ATMs) has been well-described in obese conditions in mice and humans.3–5 Moreover, the ATM proinflammatory phenotype has been linked to the development of insulin resistance in mice,4 although the exact nature of the proinflammatory myeloid cells, ie, macrophages or dentritic cells, remains to be determined.6 Nevertheless, the causal link between inflammation and insulin resistance was further strengthened by the specific knock-out of the inflammation coordinator IkappaB kinase beta of myeloid cells, which gave protection against insulin resistance.7 The study of Kintscher and al in this issue8 extends those original observations to cells of adaptative immunity. The authors suggest that the accumulation of T-lymphocytes, assessed mainly through gene expression analyses and immunohistochemistry, occurs in the perigonadal adipose tissue of mice on a high-fat diet before the accumulation of macrophages. Moreover, the increased expression of T-lymphocyte markers was concomitant with the initiation of insulin resistance characterized by a reduction in systemic glucose tolerance and insulin sensitivity, at least compared with counterpart animals that were 5 weeks younger. Given these new findings in rodents, the authors suggest that early lymphocyte infiltration of the adipose tissue might be considered as a primary event that orchestrates the adipose tissue inflammation (Figure). This provocative and attractive idea poses a number of questions and requires further clinical investigations to validate its relevance in humans. Download figureDownload PowerPointFigure. Putative processes involved the accumulation of immune cells within adipose tissue and their pathogenic roles in obesity and associated pathologies. Blood circulating lymphocytes might be attracted through the production of the SDF-1 produced by endothelial cells. Extravasation of T-lymphocytes and their accumulation within the fat mass might be the primary event involved in the further accumulation of macrophages through the INFγ-stimulated production of MCP-1 by the stromal preadipocytes. In italics are the open questions that remain to be answered.See accompanying article on page 1304One of the most important tasks is to define the process underlying the accumulation of immune cells and the nature of the ATLs within the adipose tissue. Cells of adaptive immunity are able to home to healthy extralymphoid peripheral tissues, including adipose tissue, where they are thought to carry out peripheral immune monitoring and to reside as long-lived resting memory T cells. Alternatively, effector T cells efficiently home into sites of inflammation and are destined to die. Chemokines and their receptors represent a critical element in immune cell trafficking. Adipose tissue produces a wide range of chemokines, the production of which is modulated by the degree of adiposity.9 The potential contribution of MCP1/CCL2 in ATM accumulation, through its interaction with the monocyte receptor CCR2, has given conflicting results.10 In the current work, the authors suggest that resident adipose tissue cells, particularly stromal and endothelial cells (in agreement with a previous study11), could release the chemoattractant SDF1/CXCL12 and so behave as lymphocyte recruiter cells. In turn, lymphocyte-derived interferon gamma (IFNγ) would stimulate preadipocyte-derived MCP1 release, which would attract circulating monocytes. This simplistic view has to be reconciled with the fact that the main receptor of CXCL12, CXCR4, is widely expressed on all leukocytes including monocytes,12 making it unlikely to participate alone in the selective lymphocyte trafficking into adipose tissue. In-depth analysis of the adhesion molecules and chemokine receptors expressed on ATLs might reveal the specific repertoire determinant for adipose tissue homing. Moreover, as the authors point out, the expression of CD3, as well as CD4 and INFγ, is not restricted to Th1 CD4+ effectors but is found in other T cell subsets such as memory and regulatory T cells as well as NKT cells that are present in adipose tissue, at least in mice.13,14Another interesting point will be to determine clearly whether changes in the degree of adiposity are associated with modulation in the number and phenotype of the ATLs. Indeed, dynamic changes in ATM numbers and phenotype have been demonstrated during the development of obesity on high-fat diets in mice. The ATM shifted from a reparative resident phenotype in lean animals to inflammatory newly recruited ATM in obese mice.15 In humans, ATMs appear less polarized16 and possess a proliferative capacity associated with a remodeling proangiogenic phenotype,17 the hallmark of cells involved in chronic inflammation. By correlative analyses and using gene expression analyses only, the present study suggests a recruitment of Th1 CD4+ cells with obesity. Therefore, the detailed characterization of ATL subsets in both human and mice adipose tissue is necessary to determine, whereas the growth of adipose tissue impacts on the phenotype, function and numbers of resident or infiltrated T cells.Finally, the pathogenic role of ATLs in obesity remains to be established. Although correlative data analyses show a clear association between lymphocyte gene markers and waist circumference in patients with type 2 diabetes, similar approaches with nondiabetic patients will determine whether ATLs are markers or players in adipose tissue growth and obesity-associated pathologies. A recent study has suggested that perivascular fat may serves as reservoir for activated effector T cells which in turn promote vascular dysfunction and hypertension.18 Moreover, local intercellular cross-talk is probably more complex than suspected. Although one might expect local interactions between ATLs and ATMs, adipocyte-derived products, including the main adipokines, leptin and adiponectin, and free fatty acids, which have been shown to modulate T-lymphocyte function and proinflammatory responses, are probably major variables to be taken into account. In this field and for long time, the work of C.M. Pond has stressed the close interactions between lymphoid cells in lymph nodes and the surrounding adipose tissue.19 In addition to this local cross-talk, interactions with adjacent tissues might contribute to the pathogenicity of ATLs. Moreover, although increasing lines of evidence point to the links between the innate and adaptative immune systems and metabolism in rodents, we must keep in mind that the jump to patient-related problems is not easy and requires a number of additional clinical studies. Indeed, there are major differences between young mice submitted to an acute nutritional stress promoted by a high-fat diet and patients who could be at different ages and stages of obesity. ATLs and ATMs, makers or markers? The question is undoubtedly there, but its definite answer and its relevance to humans remains open and clearly further complementary investigations are needed.DisclosuresNone.FootnotesCorrespondence to A. Bouloumié, Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 858 & Université Toulouse III Paul Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, IFR31, BP84225, 31432 Toulouse, France. E-mail anne. [email protected] References 1 Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006; 444: 860–867.CrossrefMedlineGoogle Scholar2 Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, Fava F, Tuohy KM, Chabo C, Waget A, Delmee E, Cousin B, Sulpice T, Chamontin B, Ferrieres J, Tanti JF, Gibson GR, Casteilla L, Delzenne NM, Alessi MC, Burcelin R. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007; 56: 1761–1772.CrossrefMedlineGoogle Scholar3 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.CrossrefMedlineGoogle Scholar4 Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, Chen H. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003; 112: 1821–1830.CrossrefMedlineGoogle Scholar5 Curat CA, Miranville A, Sengenes C, Diehl M, Tonus C, Busse R, Bouloumie A. From blood monocytes to adipose tissue-resident macrophages: induction of diapedesis by human mature adipocytes. Diabetes. 2004; 53: 1285–1292.CrossrefMedlineGoogle Scholar6 Nguyen MT, Favelyukis S, Nguyen AK, Reichart D, Scott PA, Jenn A, Liu-Bryan R, Glass CK, Neels JG, Olefsky JM. A subpopulation of macrophages infiltrates hypertrophic adipose tissue and is activated by free fatty acids via Toll-like receptors 2 and 4 and JNK-dependent pathways. J Biol Chem. 2007; 282: 35279–35292.CrossrefMedlineGoogle Scholar7 Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, Long JM, Wynshaw-Boris A, Poli G, Olefsky J, Karin M. IKK-beta links inflammation to obesity-induced insulin resistance. Nat Med. 2005; 11: 191–198.CrossrefMedlineGoogle Scholar8 Kintscher U, Hartge M, Hess K, Foryst-Ludwig A, Clemenz M, Wabitsch M, Fischer-Posovszky P, Barth TF, Dragun D, Skurk T, Hauner H, Bluher M, Unger T, Wolf AM, Knippschild U, Hombach V, Marx N. 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.LinkGoogle Scholar9 Bouloumie A, Curat CA, Sengenes C, Lolmede K, Miranville A, Busse R. Role of macrophage tissue infiltration in metabolic diseases. Curr Opin Clin Nutr Metab Care. 2005; 8: 347–354.CrossrefMedlineGoogle Scholar10 Inouye KE, Shi H, Howard JK, Daly CH, Lord GM, Rollins BJ, Flier JS. Absence of CC chemokine ligand 2 does not limit obesity-associated infiltration of macrophages into adipose tissue. Diabetes. 2007; 56: 2242–2250.CrossrefMedlineGoogle Scholar11 Sengenes C, Miranville A, Maumus M, de Barros S, Busse R, Bouloumie A. Chemotaxis and differentiation of human adipose tissue CD34+/CD31− progenitor cells: role of stromal derived factor-1 released by adipose tissue capillary endothelial cells. Stem Cells. 2007; 25: 2269–2276.CrossrefMedlineGoogle Scholar12 Cho CH, Koh YJ, Han J, Sung HK, Jong Lee H, Morisada T, Schwendener RA, Brekken RA, Kang G, Oike Y, Choi TS, Suda T, Yoo OJ, Koh GY. Angiogenic role of LYVE-1-positive macrophages in adipose tissue. Circ Res. 2007; 100: e47–e57.LinkGoogle Scholar13 Caspar-Bauguil S, Cousin B, Galinier A, Segafredo C, Nibbelink M, Andre M, Casteilla L, Penicaud L. Adipose tissues as an ancestral immune organ: site-specific change in obesity. FEBS Lett. 2005; 579: 3487–3492.CrossrefMedlineGoogle Scholar14 Caspar-Bauguil S, Cousin B, Andre M, Nibbelink M, Galinier A, Periquet B, Casteilla L, Penicaud L. Weight-dependent changes of immune system in adipose tissue: importance of leptin. Exp Cell Res. 2006; 312: 2195–2202.CrossrefMedlineGoogle Scholar15 Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007; 117: 175–184.CrossrefMedlineGoogle Scholar16 Zeyda M, Farmer D, Todoric J, Aszmann O, Speiser M, Gyori G, Zlabinger GJ, Stulnig TM. 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.CrossrefMedlineGoogle Scholar17 Bourlier V, Zakaroff-Girard A, Miranville A, De Barros S, Maumus M, Sengenes C, Galitzky J, Lafontan M, Karpe F, Frayn KN, Bouloumie A. Remodeling phenotype of human subcutaneous adipose tissue macrophages. Circulation. 2008; 117: 806–815.LinkGoogle Scholar18 Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S, Goronzy J, Weyand C, Harrison DG. Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J Exp Med. 2007; 204: 2449–2460.CrossrefMedlineGoogle Scholar19 Pond CM. Adipose tissue and the immune system. Prostaglandins Leukot Essent Fatty Acids. 2005; 73: 17–30.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Quinn G, Dhillon P and Susztak K (2020) It Takes Two to Tango: The Role of Dysregulated Metabolism and Inflammation in Kidney Disease Development, Seminars in Nephrology, 10.1016/j.semnephrol.2020.01.010, 40:2, (199-205), Online publication date: 1-Mar-2020. Cubillos-Zapata C, Almendros I, Díaz-García E, Toledano V, Casitas R, Galera R, López-Collazo E, Farre R, Gozal D and García-Rio F (2019) Differential effect of intermittent hypoxia and sleep fragmentation on PD-1/PD-L1 upregulation, Sleep, 10.1093/sleep/zsz285, 43:5, Online publication date: 12-May-2020. Duran‐Ortiz S, Young J, Jara A, Jensen E, Basu R, List E, Qian Y, Kopchick J and Berryman D (2020) Differential gene signature in adipose tissue depots of growth hormone transgenic mice, Journal of Neuroendocrinology, 10.1111/jne.12893, 32:11, Online publication date: 1-Nov-2020. Sreng N, Champion S, Martin J, Khelaifia S, Christensen J, Padmanabhan R, Azalbert V, Blasco-Baque V, Loubieres P, Pechere L, Landrier J, Burcelin R and Sérée E (2019) Resveratrol-mediated glycemic regulation is blunted by curcumin and is associated to modulation of gut microbiota, The Journal of Nutritional Biochemistry, 10.1016/j.jnutbio.2019.108218, 72, (108218), Online publication date: 1-Oct-2019. Balato A and Megna M (2017) Adipose Tissue and Cutaneous Inflammation Clinical and Basic Immunodermatology, 10.1007/978-3-319-29785-9_14, (219-238), . Burcelin R (2017) When gut fermentation controls satiety: A PYY story, Molecular Metabolism, 10.1016/j.molmet.2016.11.005, 6:1, (10-11), Online publication date: 1-Jan-2017. Echeverri Tirado L and Yassin L (2017) B cells interactions in lipid immune responses: implications in atherosclerotic disease, Lipids in Health and Disease, 10.1186/s12944-016-0390-5, 16:1, Online publication date: 1-Dec-2017. Cinkajzlová A, Mráz M and Haluzík M (2017) Lymphocytes and macrophages in adipose tissue in obesity: markers or makers of subclinical inflammation?, Protoplasma, 10.1007/s00709-017-1082-3, 254:3, (1219-1232), Online publication date: 1-May-2017. Hanefeld M, Pistrosch F, Bornstein S and Birkenfeld A (2016) The metabolic vascular syndrome - guide to an individualized treatment, Reviews in Endocrine and Metabolic Disorders, 10.1007/s11154-016-9345-4, 17:1, (5-17), Online publication date: 1-Mar-2016. Lee S, Norheim F, Langleite T, Noreng H, Storås T, Afman L, Frost G, Bell J, Thomas E, Kolnes K, Tangen D, Stadheim H, Gilfillan G, Gulseth H, Birkeland K, Jensen J, Drevon C and Holen T (2016) Effect of energy restriction and physical exercise intervention on phenotypic flexibility as examined by transcriptomics analyses of mRNA from adipose tissue and whole body magnetic resonance imaging, Physiological Reports, 10.14814/phy2.13019, 4:21, (e13019), Online publication date: 1-Nov-2016. Bandyopadhyay G, Lu M, Avolio E, Siddiqui J, Gayen J, Wollam J, Vu C, Chi N, O’Connor D and Mahata S (2014) Pancreastatin-Dependent Inflammatory Signaling Mediates Obesity-Induced Insulin Resistance, Diabetes, 10.2337/db13-1747, 64:1, (104-116), Online publication date: 1-Jan-2015. Cancello R (2015) Obesity and Inflammation in Pregnancy Metabolic Syndrome and Complications of Pregnancy, 10.1007/978-3-319-16853-1_5, (65-75), . Teixeira T, Alves R, Moreira A and Peluzio M (2015) Main characteristics of metabolically obese normal weight and metabolically healthy obese phenotypes, Nutrition Reviews, 10.1093/nutrit/nuu007, 73:3, (175-190), Online publication date: 1-Mar-2015. Zhang S, Khalyfa A, Wang Y, Carreras A, Hakim F, Neel B, Brady M, Qiao Z, Hirotsu C and Gozal D (2013) Sleep fragmentation promotes NADPH oxidase 2-mediated adipose tissue inflammation leading to insulin resistance in mice, International Journal of Obesity, 10.1038/ijo.2013.139, 38:4, (619-624), Online publication date: 1-Apr-2014. Inzaugarat M, Billordo L, Vodánovich F, Cervini G, Casavalle P, Vedire C and Cherñavsky A (2013) Alterations in innate and adaptive immune leukocytes are involved in paediatric obesity, Pediatric Obesity, 10.1111/j.2047-6310.2013.00179.x, 9:5, (381-390), Online publication date: 1-Oct-2014. Wang Y, Carreras A, Lee S, Hakim F, Zhang S, Nair D, Ye H and Gozal D (2013) Chronic sleep fragmentation promotes obesity in young adult mice, Obesity, 10.1002/oby.20616, 22:3, (758-762), Online publication date: 1-Mar-2014. Nilius B, Szallasi A and Sibley D (2014) Transient Receptor Potential Channels as Drug Targets: From the Science of Basic Research to the Art of Medicine, Pharmacological Reviews, 10.1124/pr.113.008268, 66:3, (676-814), Online publication date: 1-Jul-2014. Blasco-Baque V, Serino M and Burcelin R (2013) La thérapie métabolique à l’interface entre l’homme et le microbiote intestinal, Annales Pharmaceutiques Françaises, 10.1016/j.pharma.2012.08.003, 71:1, (34-41), Online publication date: 1-Jan-2013. Burcelin R, Serino M, Chabo C, Garidou L, Pomié C, Courtney M, Amar J and Bouloumié A (2013) Metagenome and metabolism: the tissue microbiota hypothesis, Diabetes, Obesity and Metabolism, 10.1111/dom.12157, 15:s3, (61-70), Online publication date: 1-Sep-2013. Pistrosch F, Schaper F and Hanefeld M (2013) The Metabolic Syndrome and Cardiovascular Disease The Metabolic Syndrome, 10.1007/978-3-7091-1331-8_4, (43-54), . Burcelin R, Chabo C, Blasco-Baque V, Sérino M and Amar J (2013) Le microbiote intestinal à l’origine de nouvelles perspectives thérapeutiques pour les maladies métaboliques ?, médecine/sciences, 10.1051/medsci/2013298021, 29:8-9, (800-806), Online publication date: 1-Aug-2013. Luche E, Cousin B, Garidou L, Serino M, Waget A, Barreau C, André M, Valet P, Courtney M, Casteilla L and Burcelin R (2013) Metabolic endotoxemia directly increases the proliferation of adipocyte precursors at the onset of metabolic diseases through a CD14-dependent mechanism, Molecular Metabolism, 10.1016/j.molmet.2013.06.005, 2:3, (281-291), Online publication date: 1-Aug-2013. Burcelin R, Garidou L and Pomié C (2012) Immuno-microbiota cross and talk: The new paradigm of metabolic diseases, Seminars in Immunology, 10.1016/j.smim.2011.11.011, 24:1, (67-74), Online publication date: 1-Feb-2012. Pamir N, McMillen T, Edgel K, Kim F and LeBoeuf R (2012) Deficiency of lymphotoxin-α does not exacerbate high-fat diet-induced obesity but does enhance inflammation in mice, American Journal of Physiology-Endocrinology and Metabolism, 10.1152/ajpendo.00447.2011, 302:8, (E961-E971), Online publication date: 15-Apr-2012. Mitterberger M, Lechner S, Mattesich M, Kaiser A, Probst D, Wenger N, Pierer G and Zwerschke W (2012) DLK1(PREF1) is a negative regulator of adipogenesis in CD105+/CD90+/CD34+/CD31−/FABP4− adipose-derived stromal cells from subcutaneous abdominal fat pats of adult women, Stem Cell Research, 10.1016/j.scr.2012.04.001, 9:1, (35-48), Online publication date: 1-Jul-2012. Vantyghem M, Balavoine A, Douillard C, Defrance F, Dieudonne L, Mouton F, Lemaire C, Bertrand-Escouflaire N, Bourdelle-Hego M, Devemy F, Evrard A, Gheerbrand D, Girardot C, Gumuche S, Hober C, Topolinski H, Lamblin B, Mycinski B, Ryndak A, Karrouz W, Duvivier E, Merlen E, Cortet C, Weill J, Lacroix D and Wémeau J (2012) How to diagnose a lipodystrophy syndrome, Annales d'Endocrinologie, 10.1016/j.ando.2012.04.010, 73:3, (170-189), Online publication date: 1-Jun-2012. Burcelin R (2012) Regulation of Metabolism: A Cross Talk Between Gut Microbiota and Its Human Host, Physiology, 10.1152/physiol.00023.2012, 27:5, (300-307), Online publication date: 1-Oct-2012. Dobrian A, Ma Q, Lindsay J, Leone K, Ma K, Coben J, Galkina E and Nadler J (2011) Dipeptidyl peptidase IV inhibitor sitagliptin reduces local inflammation in adipose tissue and in pancreatic islets of obese mice, American Journal of Physiology-Endocrinology and Metabolism, 10.1152/ajpendo.00463.2010, 300:2, (E410-E421), Online publication date: 1-Feb-2011. Sethi J and Vidal-Puig A (2011) Adipose Tissue Development, Structure and Function Metabolic Basis of Obesity, 10.1007/978-1-4419-1607-5_3, (53-68), . Komorowski J, Jankiewicz-Wika J, Kolomecki K, Cywinski J, Piestrzeniewicz K, Swiętoslawski J and Stepien H (2011) Systemic blood osteopontin, endostatin, and E-selectin concentrations after vertical banding surgery in severely obese adults, Cytokine, 10.1016/j.cyto.2011.03.020, 55:1, (56-61), Online publication date: 1-Jul-2011. Zhang H, Potter B, Cao J and Zhang C (2011) Interferon-gamma induced adipose tissue inflammation is linked to endothelial dysfunction in type 2 diabetic mice, Basic Research in Cardiology, 10.1007/s00395-011-0212-x, 106:6, (1135-1145), Online publication date: 1-Nov-2011. Graziani F, Cialdella P, Liuzzo G, Basile E, Brugaletta S, Pedicino D, Leccesi L, Guidone C, Iaconelli A, Mingrone G, Biasucci L and Crea F (2011) Cardiovascular risk in obesity: Different activation of inflammation and immune system between obese and morbidly obese subjects, European Journal of Internal Medicine, 10.1016/j.ejim.2011.04.010, 22:4, (418-423), Online publication date: 1-Aug-2011. Esposito L, Simpson R, Strohacker K, Carpenter K and McFarlin B (2010) Defining a longitudinal survival model to examine forced treadmill running as a countermeasure for diet-induced weight gain, Laboratory Animals, 10.1258/la.2010.010030, 44:4, (305-311), Online publication date: 1-Oct-2010. Poglio S, De Toni-Costes F, Arnaud E, Laharrague P, Espinosa E, Casteilla L and Cousin B (2010) Adipose Tissue as a Dedicated Reservoir of Functional Mast Cell Progenitors, Stem Cells, 10.1002/stem.523, 28:11, (2065-2072), Online publication date: 1-Nov-2010. Burcelin R, Chabo C, Luche É, Serino M and Corthier G (2010) Les lipopolysaccharides bactériens et les maladies métaboliques, Cahiers de Nutrition et de Diététique, 10.1016/j.cnd.2010.03.001, 45:3, (114-121), Online publication date: 1-Jun-2010. Hu Y, Tong G, Xu W, Pan J, Ryan K, Yang R, Shuldiner A, Gong D and Zhu D (2009) Anti-inflammatory effects of simvastatin on adipokines in type 2 diabetic patients with carotid atherosclerosis, Diabetes and Vascular Disease Research, 10.1177/1479164109339966, 6:4, (262-268), Online publication date: 1-Oct-2009. Caspar-Bauguil S, Cousin B, Bour S, Castiella L, Penicaud L and Carpéné C (2009) Adipose tissue lymphocytes: types and roles, Journal of Physiology and Biochemistry, 10.1007/BF03185938, 65:4, (423-436), Online publication date: 1-Dec-2009. Marteau J, Samara A, Dedoussis G, Pfister M and Visvikis-Siest S (2009) Candidate gene microarray analysis in peripheral blood cells for studying hypertension/obesity, Personalized Medicine, 10.2217/pme.09.6, 6:3, (269-291), Online publication date: 1-May-2009. Quaedackers M, Baan C, Weimar W and Hoogduijn M (2009) Cell contact interaction between adipose-derived stromal cells and allo-activated T lymphocytes, European Journal of Immunology, 10.1002/eji.200939584, 39:12, (3436-3446), Online publication date: 1-Dec-2009. Duffaut C, Galitzky J, Lafontan M and Bouloumié A (2009) Unexpected trafficking of immune cells within the adipose tissue during the onset of obesity, Biochemical and Biophysical Research Communications, 10.1016/j.bbrc.2009.05.002, 384:4, (482-485), Online publication date: 1-Jul-2009. Burcelin R and Amar J (2009) Flore intestinale et maladies métaboliques, Médecine des Maladies Métaboliques, 10.1016/S1957-2557(09)71629-3, 3:2, (159-164), Online publication date: 1-Mar-2009. Poitou C, Divoux A, Faty A, Tordjman J, Hugol D, Aissat A, Keophiphath M, Henegar C, Commans S and Clément K (2009) Role of Serum Amyloid A in Adipocyte-Macrophage Cross Talk and Adipocyte Cholesterol Efflux, The Journal of Clinical Endocrinology & Metabolism, 10.1210/jc.2008-2040, 94:5, (1810-1817), Online publication date: 1-May-2009. Chaldakov G, Fiore M, Ghenev P, Beltowski J, Ranćić G, Tunçel N and Aloe L (2014) Triactome: Neuro–Immune–Adipose Interactions. Implication in Vascular Biology, Frontiers in Immunology, 10.3389/fimmu.2014.00130, 5 Kalathookunnel Antony A, Lian Z and Wu H (2018) T Cells in Adipose Tissue in Aging, Frontiers in Immunology, 10.3389/fimmu.2018.02945, 9 July 2008Vol 28, Issue 7 Advertisement Article InformationMetrics https://doi.org/10.1161/ATVBAHA.108.168229PMID: 18565843 Originally publishedJuly 1, 2008 PDF download Advertisement

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call