388
Views
30
CrossRef citations to date
0
Altmetric
Reviews

Inflammatory processes in obesity: focus on endothelial dysfunction and the role of adipokines as inflammatory mediators

We reviewed obesity-induced metabolic and immunological changes at the level of vasculature and emphasize on the importance of adipokines

& ORCID Icon
Pages 157-171 | Received 10 Jun 2019, Accepted 26 Jun 2019, Published online: 09 Jul 2019

References

  • Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol 2011;29:415–445. doi:10.1146/annurev-immunol-031210-101322.
  • Flegal KM, Carroll MD, Kit BK, Ogden CL. Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999-2010. JAMA 2012;307(5):491–497. doi:10.1001/jama.2012.39.
  • Kanneganti TD, Dixit VD. Immunological complications of obesity. Nat Immunol 2012;13(8):707–712. doi:10.1038/ni.2343.
  • Kusminski CM, Bickel PE, Scherer PE. Targeting adipose tissue in the treatment of obesity-associated diabetes. Nat Rev Drug Discov 2016;15(9):639–660. doi:10.1038/nrd.2016.75.
  • Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nat Rev Immunol 2011;11(2):85–97. doi:10.1038/nri2921.
  • Sun S, Ji Y, Kersten S, Qi L. Mechanisms of inflammatory responses in obese adipose tissue. Annu Rev Nutr 2012;32:261–286. doi:10.1146/annurev-nutr-071811-150623.
  • Solinas G, Karin M. JNK1 and IKKbeta: molecular links between obesity and metabolic dysfunction. FASEB J 2010;24(8):2596–2611. doi:10.1096/fj.09-151340.
  • Peraldi P, Hotamisligil GS, Buurman WA, et al. Tumor necrosis factor (TNF)-alpha inhibits insulin signaling through stimulation of the p55 TNF receptor and activation of sphingomyelinase. J Biol Chem 1996;271(22):13018–13022. doi:10.1074/jbc.271.22.13018.
  • Hotamisligil GS, Murray DL, Choy LN, Spiegelman BM. Tumor necrosis factor alpha inhibits signaling from the insulin receptor. Proc Natl Acad Sci U S A 1994;91(11):4854–4858. doi:10.1073/pnas.91.11.4854.
  • Fessler MB, Rudel LL, Brown JM. Toll-like receptor signaling links dietary fatty acids to the metabolic syndrome. Curr Opin Lipidol 2009;20(5):379–385. doi:10.1097/MOL.0b013e32832fa5c4.
  • Shi H, Kokoeva MV, Inouye K, et al. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 2006;116(11):3015–3025. doi:10.1172/JCI28898.
  • Himes RW, Smith CW. Tlr2 is critical for diet-induced metabolic syndrome in a murine model. Faseb J 2010;24(3):731–739. doi:10.1096/fj.09-141929.
  • Chen G, Shaw MH, Kim YG, Nunez G. NOD-like receptors: role in innate immunity and inflammatory disease. Annu Rev Pathol 2009;4:365–398. doi:10.1146/annurev.pathol.4.110807.092239.
  • Schroder K, Zhou R, Tschopp J. The NLRP3 inflammasome: a sensor for metabolic danger?. Science 2010;327(5963):296–300. doi:10.1126/science.1184003.
  • De Nardo D, Latz E. NLRP3 inflammasomes link inflammation and metabolic disease. Trends Immunol 2011;32(8):373–379. doi:10.1016/j.it.2011.05.004.
  • Vandanmagsar B, Youm YH, Ravussin A, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med 2011;17(2):179–188. doi:10.1038/nm.2279.
  • Arkan MC, Hevener AL, Greten FR, et al. IKK-beta links inflammation to obesity-induced insulin resistance. Nat Med 2005;11(2):191–198. doi:10.1038/nm1185.
  • Cai D, Yuan M, Frantz DF, et al. Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB. Nat Med 2005;11(2):183–190. doi:10.1038/nm1166.
  • Tuncman G, Hirosumi J, Solinas G, et al. Functional in vivo interactions between JNK1 and JNK2 isoforms in obesity and insulin resistance. Proc Natl Acad Sci U S A 2006;103(28):10741–10746. doi:10.1073/pnas.0603509103.
  • Belgardt BF, Mauer J, Wunderlich FT, et al. Hypothalamic and pituitary c-Jun N-terminal kinase 1 signaling coordinately regulates glucose metabolism. Proc Natl Acad Sci U S A 2010;107(13):6028–6033. doi:10.1073/pnas.1001796107.
  • Solinas G, Vilcu C, Neels JG, et al. JNK1 in hematopoietically derived cells contributes to diet-induced inflammation and insulin resistance without affecting obesity. Cell Metab 2007;6(5):386–397. doi:10.1016/j.cmet.2007.09.011.
  • Dabo S, Meurs EF. dsRNA-dependent protein kinase PKR and its role in stress, signaling and HCV infection. Viruses 2012;4(11):2598–2635. doi:10.3390/v4112598.
  • Boura-Halfon S, Zick Y. Phosphorylation of IRS proteins, insulin action, and insulin resistance. Am J Physiol Endocrinol Metab 2009;296(4):E581–591. doi:10.1152/ajpendo.90437.2008.
  • Tanti JF, Ceppo F, Jager J, Berthou F. Implication of inflammatory signaling pathways in obesity-induced insulin resistance. Front Endocrinol (Lausanne) 2012;3:181. doi:10.3389/fendo.2012.00181.
  • Kosteli A, Sugaru E, Haemmerle G, et al. Weight loss and lipolysis promote a dynamic immune response in murine adipose tissue. J Clin Invest 2010;120(10):3466–3479. doi:10.1172/JCI42845.
  • Prieur X, Mok CY, Velagapudi VR, et al. Differential lipid partitioning between adipocytes and tissue macrophages modulates macrophage lipotoxicity and M2/M1 polarization in obese mice. Diabetes 2011;60(3):797–809. doi:10.2337/db10-0705.
  • Kern PA, Saghizadeh M, Ong JM, et al. The expression of tumor necrosis factor in human adipose tissue. Regulation by obesity, weight loss, and relationship to lipoprotein lipase. J Clin Invest 1995;95(5):2111–2119. doi:10.1172/JCI117899.
  • Xu H, Barnes GT, Yang Q, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest 2003;112(12):1821–1830. doi:10.1172/JCI200319451.
  • Fontana L, Eagon JC, Trujillo ME, et al. Visceral fat adipokine secretion is associated with systemic inflammation in obese humans. Diabetes 2007;56(4):1010–1013. doi:10.2337/db06-1656.
  • Catalan V, Gomez-Ambrosi J, Ramirez B, et al. Proinflammatory cytokines in obesity: impact of type 2 diabetes mellitus and gastric bypass. Obes Surg 2007;17(11):1464–1474.
  • 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(3):451–463. doi:10.1038/sj.ijo.0803744.
  • Winer S, Chan Y, Paltser G, et al. Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med 2009;15(8):921–929. doi:10.1038/nm.2001.
  • Nishimura S, Manabe I, Nagasaki M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med 2009;15(8):914–920. doi:10.1038/nm.1964.
  • Tiemessen MM, Jagger AL, Evans HG, et al. CD4 + CD25 + Foxp3+ regulatory T cells induce alternative activation of human monocytes/macrophages. Proc Natl Acad Sci U S A 2007;104(49):19446–19451. doi:10.1073/pnas.0706832104.
  • Feuerer M, Herrero L, Cipolletta D, 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(8):930–939. doi:10.1038/nm.2002.
  • Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 2007;117(1):175–184. doi:10.1172/JCI29881.
  • Murano I, Barbatelli G, Parisani V, et al. Dead adipocytes, detected as crown-like structures, are prevalent in visceral fat depots of genetically obese mice. J Lipid Res 2008;49(7):1562–1568. doi:10.1194/jlr.M800019-JLR200.
  • Bluher M. Adipose tissue dysfunction in obesity. Exp Clin Endocrinol Diab 2009;117:241–250.
  • Vestweber D. How leukocytes cross the vascular endothelium. Nat Rev Immunol 2015;15(11):692–704. doi:10.1038/nri3908.
  • Villaret A, Galitzky J, Decaunes P, et al. Adipose tissue endothelial cells from obese human subjects: differences among depots in angiogenic, metabolic, and inflammatory gene expression and cellular senescence. Diabetes 2010;59(11):2755–2763. doi:10.2337/db10-0398.
  • Muller WA. How endothelial cells regulate transmigration of leukocytes in the inflammatory response. Am J Pathol 2014;184(4):886–896. doi:10.1016/j.ajpath.2013.12.033.
  • Pasarica M, Sereda OR, Redman LM, et al. Reduced adipose tissue oxygenation in human obesity: evidence for rarefaction, macrophage chemotaxis, and inflammation without an angiogenic response. Diabetes 2009;58(3):718–725. doi:10.2337/db08-1098.
  • Cao Y. Angiogenesis modulates adipogenesis and obesity. J Clin Invest. 2007;117(9):2362–2368. doi:10.1172/JCI32239.
  • Rupnick MA, Panigrahy D, Zhang CY, et al. Adipose tissue mass can be regulated through the vasculature. Proc Natl Acad Sci U S A 2002;99(16):10730–10735. doi:10.1073/pnas.162349799.
  • Kim DH, Gutierrez-Aguilar R, Kim HJ, et al. Increased adipose tissue hypoxia and capacity for angiogenesis and inflammation in young diet-sensitive C57 mice compared with diet-resistant FVB mice. Int J Obes 2013;37(6):853–860. doi:10.1038/ijo.2012.141.
  • Arroyo AG, Iruela-Arispe ML. Extracellular matrix, inflammation, and the angiogenic response. Cardiovasc Res 2010;86(2):226–235. doi:10.1093/cvr/cvq049.
  • Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011;473(7347):298–307. doi:10.1038/nature10144.
  • Lehr S, Hartwig S, Sell H. Adipokines: a treasure trove for the discovery of biomarkers for metabolic disorders. Proteomics Clin Appl 2012;6(1–2):91–101. doi:10.1002/prca.201100052.
  • Fasshauer M, Bluher M. Adipokines in health and disease. Trends Pharmacol Sci 2015;36(7):461–470. doi:10.1016/j.tips.2015.04.014.
  • Khan M, Joseph F. Adipose tissue and adipokines: the association with and application of adipokines in obesity. Scientifica (Cairo) 2014;2014:328592. doi:10.1155/2014/328592.
  • Zhang Y, Proenca R, Maffei M, et al. Positional cloning of the mouse obese gene and its human homologue. Nature 1994;372(6505):425–432. doi:10.1038/372425a0.
  • Takahashi N, Waelput W, Guisez Y. Leptin is an endogenous protective protein against the toxicity exerted by tumor necrosis factor. J Exp Med 1999;189(1):207–212.
  • Krempler F, Hell E, Winkler C, et al. Plasma leptin levels: interaction of obesity with a common variant of insulin receptor substrate-1. Arterioscler Thromb Vasc Biol 1998;18(11):1686–1690.
  • Falorni A, Bini V, Molinari D, et al. Leptin serum levels in normal weight and obese children and adolescents: relationship with age, sex, pubertal development, body mass index and insulin. Int J Obes Relat Metab Disord 1997;21(10):881–890.
  • Loffreda S, Yang SQ, Lin HZ, et al. Leptin regulates proinflammatory immune responses. FASEB J 1998;12(1):57–65.
  • Santos-Alvarez J, Goberna R, Sanchez-Margalet V. Human leptin stimulates proliferation and activation of human circulating monocytes. Cell Immunol 1999;194(1):6–11. doi:10.1006/cimm.1999.1490.
  • Kiguchi N, Maeda T, Kobayashi Y, et al. Leptin enhances CC-chemokine ligand expression in cultured murine macrophage. Biochem Biophys Res Commun 2009;384(3):311–315. doi:10.1016/j.bbrc.2009.04.121.
  • Grunfeld C, Zhao C, Fuller J, et al. Endotoxin and cytokines induce expression of leptin, the ob gene product, in hamsters. J Clin Invest 1996;97(9):2152–2157. doi:10.1172/JCI118653.
  • Matarese G, Di Giacomo A, Sanna V, et al. Requirement for leptin in the induction and progression of autoimmune encephalomyelitis. J Immunol 2001;166(10):5909–5916. doi:10.4049/jimmunol.166.10.5909.
  • Momin AU, Melikian N, Shah AM, et al. Leptin is an endothelial-independent vasodilator in humans with coronary artery disease: evidence for tissue specificity of leptin resistance. Eur Heart J 2006;27(19):2294–2299. doi:10.1093/eurheartj/ehi831.
  • Bouloumie A, Marumo T, Lafontan M, Busse R. Leptin induces oxidative stress in human endothelial cells. FASEB J 1999;13(10):1231–1238.
  • Singh P, Hoffmann M, Wolk R, et al. Leptin induces C-reactive protein expression in vascular endothelial cells. Arterioscler Thromb Vasc Biol 2007;27(9):e302–307. doi:10.1161/ATVBAHA.107.148353.
  • Aleffi S, Petrai I, Bertolani C, et al. Upregulation of proinflammatory and proangiogenic cytokines by leptin in human hepatic stellate cells. Hepatology 2005;42(6):1339–1348. doi:10.1002/hep.20965.
  • Hida K, Wada J, Eguchi J, et al. Visceral adipose tissue-derived serine protease inhibitor: a unique insulin-sensitizing adipocytokine in obesity. Proc Natl Acad Sci U S A 2005;102(30):10610–10615. doi:10.1073/pnas.0504703102.
  • Teshigawara S, Wada J, Hida K, et al. Serum vaspin concentrations are closely related to insulin resistance, and rs77060950 at SERPINA12 genetically defines distinct group with higher serum levels in Japanese population. J Clin Endocrinol Metab 2012;97(7):E1202–1207. doi:10.1210/jc.2011-3297.
  • Bluher M. Vaspin in obesity and diabetes: pathophysiological and clinical significance. Endocrine 2012;41:176–182.
  • Li H, Peng W, Zhuang J, et al. Vaspin attenuates high glucose-induced vascular smooth muscle cells proliferation and chemokinesis by inhibiting the MAPK, PI3K/Akt, and NF-κB signaling pathways . Atherosclerosis 2013;228(1):61–68. doi:10.1016/j.atherosclerosis.2013.02.013.
  • Youn BS, Kloting N, Kratzsch J, et al. Serum vaspin concentrations in human obesity and type 2 diabetes. Diabetes 2008;57(2):372–377. doi:10.2337/db07-1045.
  • Phalitakul S, Okada M, Hara Y, Yamawaki H. Vaspin prevents TNF-α-induced intracellular adhesion molecule-1 via inhibiting reactive oxygen species-dependent NF-κB and PKCθ activation in cultured rat vascular smooth muscle cells . Pharmacol Res 2011;64(5):493–500. doi:10.1016/j.phrs.2011.06.001.
  • Jung CH, Lee MJ, Kang YM, et al. Vaspin inhibits cytokine-induced nuclear factor-kappa B activation and adhesion molecule expression via AMP-activated protein kinase activation in vascular endothelial cells. Cardiovasc Diabetol 2014;13:41 doi:10.1186/1475-2840-13-41.
  • Samal B, Sun Y, Stearns G, et al. Cloning and characterization of the cDNA encoding a novel human pre-B-cell colony-enhancing factor. Mol Cell Biol. 1994;14(2):1431–1437. doi:10.1128/mcb.14.2.1431.
  • Rongvaux A, Shea RJ, Mulks MH, et al. Pre-B-cell colony-enhancing factor, whose expression is up-regulated in activated lymphocytes, is a nicotinamide phosphoribosyltransferase, a cytosolic enzyme involved in NAD biosynthesis. Eur J Immunol 2002;32(11):3225–3234. doi:10.1002/1521-4141(200211)32:11<:AID-IMMU3225&gt;3.0.CO;2-L.
  • Fukuhara A, Matsuda M, Nishizawa M, et al. Visfatin: a protein secreted by visceral fat that mimics the effects of insulin. Science 2005;307(5708):426–430. doi:10.1126/science.1097243.
  • Friebe D, Neef M, Kratzsch J, et al. Leucocytes are a major source of circulating nicotinamide phosphoribosyltransferase (NAMPT)/pre-B cell colony (PBEF)/visfatin linking obesity and inflammation in humans. Diabetologia 2011;54(5):1200–1211. doi:10.1007/s00125-010-2042-z.
  • Dahl TB, Holm S, Aukrust P, Halvorsen B. Visfatin/NAMPT: a multifaceted molecule with diverse roles in physiology and pathophysiology. Annu Rev Nutr 2012;32:229–243. doi:10.1146/annurev-nutr-071811-150746.
  • Garten A, Schuster S, Penke M, et al. Physiological and pathophysiological roles of NAMPT and NAD metabolism. Nat Rev Endocrinol 2015;11(9):535–546. doi:10.1038/nrendo.2015.117.
  • Zhao B, Zhang M, Han X, et al. Cerebral ischemia is exacerbated by extracellular nicotinamide phosphoribosyltransferase via a non-enzymatic mechanism. PLoS One 2013;8(12):e85403. doi:10.1371/journal.pone.0085403.
  • Jing Z, Xing J, Chen X, et al. Neuronal NAMPT is released after cerebral ischemia and protects against white matter injury. J Cereb Blood Flow Metab 2014;34(10):1613–1621. doi:10.1038/jcbfm.2014.119.
  • Revollo JR, Korner A, Mills KF, et al. Nampt/PBEF/visfatin regulates insulin secretion in beta cells as a systemic NAD biosynthetic enzyme. Cell Metab 2007;6(5):363–375. doi:10.1016/j.cmet.2007.09.003.
  • Lim SY, Davidson SM, Paramanathan AJ, et al. The novel adipocytokine visfatin exerts direct cardioprotective effects. J Cell Mol Med 2008;12(4):1395–1403. doi:10.1111/j.1582-4934.2008.00332.x.
  • Song HK, Lee MH, Kim BK, et al. Visfatin: a new player in mesangial cell physiology and diabetic nephropathy. Am J Physiol Renal Physiol 2008;295(5):F1485–1494. doi:10.1152/ajprenal.90231.2008.
  • Brentano F, Schorr O, Ospelt C, et al. Pre-B cell colony-enhancing factor/visfatin, a new marker of inflammation in rheumatoid arthritis with proinflammatory and matrix-degrading activities. Arthritis Rheum 2007;56(9):2829–2839. doi:10.1002/art.22833.
  • Montecucco F, Bauer I, Braunersreuther V, et al. Inhibition of nicotinamide phosphoribosyltransferase reduces neutrophil-mediated injury in myocardial infarction. Antioxid Redox Signal 2013;18(6):630–641. doi:10.1089/ars.2011.4487.
  • Dahl TB, Yndestad A, Skjelland M, et al. Increased expression of visfatin in macrophages of human unstable carotid and coronary atherosclerosis: possible role in inflammation and plaque destabilization. Circulation 2007;115(8):972–980. doi:10.1161/CIRCULATIONAHA.106.665893.
  • Romacho T, Sanchez-Ferrer CF, Peiro C. Visfatin/Nampt: an adipokine with cardiovascular impact. Mediators Inflamm 2013;2013:946427doi:10.1155/2013/946427.
  • Jurdana MPA, Černelič Bizjak M, Bizjak M, et al. Increased serum visfatin levels in obesity and its association with anthropometric/biochemical parameters, physical inactivity and nutrition. Clin Nutr ESPEN. 2013;8:e59–e67. doi:10.1016/j.clnme.2013.02.001.
  • Taşkesen D, Kirel B, Us T. Serum visfatin levels, adiposity and glucose metabolism in obese adolescents. J Clin Res Pediatr Endocrinol 2012;4(2):76–81. doi:10.4274/jcrpe.547.
  • Sun G, Bishop J, Khalili S, et al. Serum visfatin concentrations are positively correlated with serum triacylglycerols and down-regulated by overfeeding in healthy young men. Am J Clin Nutr. 2007;85(2):399–404. doi:10.1093/ajcn/85.2.399.
  • Moschen AR, Kaser A, Enrich B, et al. Visfatin, an adipocytokine with proinflammatory and immunomodulating properties. J Immunol 2007;178(3):1748–1758. doi:10.4049/jimmunol.178.3.1748.
  • Lee WJ, Wu CS, Lin H, et al. Visfatin-induced expression of inflammatory mediators in human endothelial cells through the NF-kappaB pathway. Int J Obes 2009;33(4):465–472. doi:10.1038/ijo.2009.24.
  • Adya R, Tan BK, Chen J, Randeva HS. Nuclear factor-kappaB induction by visfatin in human vascular endothelial cells: its role in MMP-2/9 production and activation. Diabetes Care 2008;31(4):758–760. doi:10.2337/dc07-1544.
  • Schuster S, Penke M, Gorski T, et al. FK866-induced NAMPT inhibition activates AMPK and downregulates mTOR signaling in hepatocarcinoma cells. Biochem Biophys Res Commun 2015;458(2):334–340. doi:10.1016/j.bbrc.2015.01.111.
  • Busso N, Karababa M, Nobile M, et al. Pharmacological inhibition of nicotinamide phosphoribosyltransferase/visfatin enzymatic activity identifies a new inflammatory pathway linked to NAD. PLoS One 2008;3(5):e2267. doi:10.1371/journal.pone.0002267.
  • Gehrke I, Bouchard ED, Beiggi S, et al. On-target effect of FK866, a nicotinamide phosphoribosyl transferase inhibitor, by apoptosis-mediated death in chronic lymphocytic leukemia cells. Clin Cancer Res 2014;20(18):4861–4872. doi:10.1158/1078-0432.CCR-14-0624.
  • Matsuda A, Yang WL, Jacob A, et al. FK866, a visfatin inhibitor, protects against acute lung injury after intestinal ischemia-reperfusion in mice via NF-κB pathway. Ann Surg 2014;259(5):1007–1017. doi:10.1097/SLA.0000000000000329.
  • Moore Z, Chakrabarti G, Luo X, et al. NAMPT inhibition sensitizes pancreatic adenocarcinoma cells to tumor-selective, PAR-independent metabolic catastrophe and cell death induced by β-lapachone. Cell Death Dis 2015;6:e1599. doi:10.1038/cddis.2014.564.
  • Li Y, Zhang Y, Dorweiler B, et al. Extracellular Nampt promotes macrophage survival via a nonenzymatic interleukin-6/STAT3 signaling mechanism. J Biol Chem 2008;283(50):34833–34843. doi:10.1074/jbc.M805866200.
  • Bi TQ, Che XM. Nampt/PBEF/visfatin and cancer. Cancer Biol Ther 2010;10(2):119–125. doi:10.4161/cbt.10.2.12581.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.