Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter September 25, 2017

The color of fat and its central role in the development and progression of metabolic diseases

  • Melania Gaggini , Fabrizia Carli and Amalia Gastaldelli ORCID logo EMAIL logo

Abstract

Excess caloric intake does not always translate to an expansion of the subcutaneous adipose tissue (SAT) and increase in fat mass. It is now recognized that adipocyte type (white, WAT, or brown, BAT), size (large vs. small) and metabolism are important factors for the development of cardiometabolic diseases. When the subcutaneous adipose tissue is not able to expand in response to increased energy intake the excess substrate is stored as visceral adipose tissue or as ectopic fat in tissues as muscle, liver and pancreas. Moreover, adipocytes become dysfunctional (adiposopathy, or sick fat), adipokines secretion is increased, fat accumulates in ectopic sites like muscle and liver and alters insulin signaling, increasing the demand for insulin secretion. Thus, there are some subjects that despite having normal weight have the metabolic characteristics of the obese (NWMO), while some obese expand their SAT and remain metabolically healthy (MHO). In this paper we have reviewed the recent findings that relate the metabolism of adipose tissue and its composition to metabolic diseases. In particular, we have discussed the possible role of dysfunctional adipocytes and adipose tissue resistance to the antilipolytic effect of insulin on the development of impaired glucose metabolism. Finally we have reviewed the possible role of BAT vs. WAT in the alteration of lipid and glucose metabolism and the recent studies that have tried to stimulate browning in human adipose tissue.

Author Statement

  1. Research funding: AG is a member of the “EPoS” (Elucidating Pathways of Steatohepatitis) consortium, which is funded by the Horizon 2020 Framework Program of the European Union under Grant Agreement 634413 and of the European Training Network “Foie Gras” (on Bioenergetic Remodelling in the Pathophysiology and Treatment of Non-Alcoholic Fatty Liver Disease) which is funded by the Horizon 2020 Framework Program of the European Union under Grant Agreement 722619. AG has received research funding and FC a scholarship by MIUR-CNR “Progetto Premiale” (Environment, life style and cardiovascular diseases: from molecules to man).

  2. Conflict of interest: Authors state no conflict of interest.

  3. Informed consent: Informed consent is not applicable.

  4. Ethical approval: The conducted research is not related to either human or animals use.

References

[1] Abraham TM, Pedley A, Massaro JM, Hoffmann U, Fox CS. Association between visceral and subcutaneous adipose depots and incident cardiovascular disease risk factors. Circulation. 2015;132:1639–47.Search in Google Scholar

[2] Neeland IJ, Turer AT, Ayers CR, Powell-Wiley TM, Vega GL, Farzaneh-Far R, et al. Dysfunctional adiposity and the risk of prediabetes and type 2 diabetes in obese adults. J Am Med Assoc. 2012;308:1150–9.Search in Google Scholar

[3] Saponaro C, Gaggini M, Carli F, Gastaldelli A. The subtle balance between lipolysis and lipogenesis: a critical point in metabolic homeostasis. Nutrients. 2015;7:9453–74.Search in Google Scholar

[4] Gaggini M, Saponaro C, Gastaldelli A. Not all fats are created equal: adipose vs. ectopic fat, implication in cardiometabolic diseases. Horm Mol Biol Clin Investig. 2015;22:7–18.Search in Google Scholar

[5] Bartelt A, Heeren J. Adipose tissue browning and metabolic health. Nat Rev Endocrinol. 2014;10:24–36.Search in Google Scholar

[6] Oelkrug R, Polymeropoulos ET, Jastroch M. Brown adipose tissue: physiological function and evolutionary significance. J Comp Physiol B. 2015;185:587–606.Search in Google Scholar

[7] Gastaldelli A. Visceral adipose tissue and ectopic fat deposition. In: Bray GA, Bouchard C, editors. Handbook of obesity – Volume 1. 1: CRC Press; 2014. p. 237–48.Search in Google Scholar

[8] Ye R, Scherer PE. Adiponectin, driver or passenger on the road to insulin sensitivity? Mol Metab. 2013;2:133–41.Search in Google Scholar

[9] Arner E, Westermark PO, Spalding KL, Britton T, Ryden M, Frisen J, et al. Adipocyte turnover: relevance to human adipose tissue morphology. Diabetes. 2010;59:105–9.Search in Google Scholar

[10] Sainz N, Barrenetxe J, Moreno-Aliaga MJ, Martinez JA. Leptin resistance and diet-induced obesity: central and peripheral actions of leptin. Metabol Clin Exp. 2015;64:35–46.Search in Google Scholar

[11] Zingaretti MC, Crosta F, Vitali A, Guerrieri M, Frontini A, Cannon B, et al. The presence of UCP1 demonstrates that metabolically active adipose tissue in the neck of adult humans truly represents brown adipose tissue. FASEB J. 2009;23:3113–20.Search in Google Scholar

[12] Virtanen KA, Lidell ME, Orava J, Heglind M, Westergren R, Niemi T, et al. Functional brown adipose tissue in healthy adults. N Engl J Med. 2009;360:1518–25.Search in Google Scholar

[13] Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, et al. Identification and importance of brown adipose tissue in adult humans. N Engl J Med. 2009;360:1509–17.Search in Google Scholar

[14] Heeren J, Munzberg H. Novel aspects of brown adipose tissue biology. Endocrinol Metab Clin North Am. 2013;42:89–107.Search in Google Scholar

[15] Young P, Arch JR, Ashwell M. Brown adipose tissue in the parametrial fat pad of the mouse. FEBS Lett. 1984;167:10–4.Search in Google Scholar

[16] Vitali A, Murano I, Zingaretti MC, Frontini A, Ricquier D, Cinti S. The adipose organ of obesity-prone C57BL/6J mice is composed of mixed white and brown adipocytes. J Lipid Res. 2012;53:619–29.Search in Google Scholar

[17] Sidossis L, Kajimura S. Brown and beige fat in humans: thermogenic adipocytes that control energy and glucose homeostasis. J Clin Invest. 2015;125:478–86.Search in Google Scholar

[18] Mueller E. Understanding the variegation of fat: novel regulators of adipocyte differentiation and fat tissue biology. Biochim Biophys Acta. 2014;1842:352–7.Search in Google Scholar

[19] Frontini A, Vitali A, Perugini J, Murano I, Romiti C, Ricquier D, et al. White-to-brown transdifferentiation of omental adipocytes in patients affected by pheochromocytoma. Biochim Biophys Acta. 2013;1831:950–9.Search in Google Scholar

[20] Sacks HS, Fain JN, Bahouth SW, Ojha S, Frontini A, Budge H, et al. Adult epicardial fat exhibits beige features. J Clin Endocrinol Metab. 2013;98:E1448–55.Search in Google Scholar

[21] Gnad T, Scheibler S, von Kugelgen I, Scheele C, Kilic A, Glode A, et al. Adenosine activates brown adipose tissue and recruits beige adipocytes via A2A receptors. Nature. 2014;516:395–9.Search in Google Scholar

[22] Sacks HS, Fain JN, Holman B, Cheema P, Chary A, Parks F, et al. Uncoupling protein-1 and related messenger ribonucleic acids in human epicardial and other adipose tissues: epicardial fat functioning as brown fat. J Clin Endocrinol Metab. 2009;94:3611–5.Search in Google Scholar

[23] Gustafson B, Hammarstedt A, Hedjazifar S, Hoffmann JM, Svensson PA, Grimsby J, et al. BMP4 and BMP antagonists regulate human white and beige adipogenesis. Diabetes. 2015;64:1670–81.Search in Google Scholar

[24] Wu J, Bostrom P, Sparks LM, Ye L, Choi JH, Giang AH, et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell. 2012;150:366–76.Search in Google Scholar

[25] Kajimura S, Spiegelman BM, Seale P. Brown and beige fat: physiological roles beyond heat generation. Cell Metab. 2015;22:546–59.Search in Google Scholar

[26] Walden TB, Hansen IR, Timmons JA, Cannon B, Nedergaard J. Recruited vs. nonrecruited molecular signatures of brown, “brite,” and white adipose tissues. Am J Physiol Endocrinol Metab. 2012;302:E19–31.Search in Google Scholar

[27] Schaffer JE. Lipotoxicity: when tissues overeat. Curr Opin Lipidol. 2003;14:281–7.Search in Google Scholar

[28] Spalding KL, Arner E, Westermark PO, Bernard S, Buchholz BA, Bergmann O, et al. Dynamics of fat cell turnover in humans. Nature. 2008;453:783–7.Search in Google Scholar

[29] Jo J, Gavrilova O, Pack S, Jou W, Mullen S, Sumner AE, et al. Hypertrophy and/or hyperplasia: dynamics of adipose tissue growth. PLoS Comput Biol. 2009;5:e1000324.Search in Google Scholar

[30] Yang J, Eliasson B, Smith U, Cushman SW, Sherman AS. The size of large adipose cells is a predictor of insulin resistance in first-degree relatives of type 2 diabetic patients. Obesity (Silver Spring). 2012;20:932–8.Search in Google Scholar

[31] Johannsen DL, Tchoukalova Y, Tam CS, Covington JD, Xie W, Schwarz JM, et al. Effect of 8 weeks of overfeeding on ectopic fat deposition and insulin sensitivity: testing the “adipose tissue expandability” hypothesis. Diabetes Care. 2014;37:2789–97.Search in Google Scholar

[32] Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 2006;116:1494–505.Search in Google Scholar

[33] Arner P. The adipocyte in insulin resistance: key molecules and the impact of the thiazolidinediones. Trends Endocrinol Metab. 2003;14:137–45.Search in Google Scholar

[34] Bays H. Adiposopathy, “sick fat,” Ockham’s razor, and resolution of the obesity paradox. Curr Atheroscler Rep. 2014;16:409.Search in Google Scholar

[35] Morelli M, Gaggini M, Daniele G, Marraccini P, Sicari R, Gastaldelli A. Ectopic fat: the true culprit linking obesity and cardiovascular disease? Thromb Haemost. 2013;110:651–60.Search in Google Scholar

[36] Alligier M, Gabert L, Meugnier E, Lambert-Porcheron S, Chanseaume E, Pilleul F, et al. Visceral fat accumulation during lipid overfeeding is related to subcutaneous adipose tissue characteristics in healthy men. J Clin Endocrinol Metab. 2013;98:802–10.Search in Google Scholar

[37] Arner P, Arner E, Hammarstedt A, Smith U. Genetic predisposition for Type 2 diabetes, but not for overweight/obesity, is associated with a restricted adipogenesis. PLoS One. 2011;6:e18284.Search in Google Scholar

[38] Samocha-Bonet D, Dixit VD, Kahn CR, Leibel RL, Lin X, Nieuwdorp M, et al. Metabolically healthy and unhealthy obese–the 2013 Stock Conference Report. Obes Rev. 2014;15:697–708.Search in Google Scholar

[39] Itani SI, Ruderman NB, Schmieder F, Boden G. Lipid-induced insulin resistance in human muscle is associated with changes in diacylglycerol, protein kinase C, and IkappaB-alpha. Diabetes. 2002;51:2005–11.Search in Google Scholar

[40] Boden G, Lebed B, Schatz M, Homko C, Lemieux S. Effects of acute changes of plasma free fatty acids on intramyocellular fat content and insulin resistance in healthy subjects. Diabetes. 2001;50:1612–7.Search in Google Scholar

[41] McGarry JD. Banting lecture 2001: dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Diabetes. 2002;51:7–18.Search in Google Scholar

[42] Brehm A, Krssak M, Schmid AI, Nowotny P, Waldhausl W, Roden M. Increased lipid availability impairs insulin-stimulated ATP synthesis in human skeletal muscle. Diabetes. 2006;55:136–40.Search in Google Scholar

[43] Wang XL, Zhang L, Youker K, Zhang MX, Wang J, LeMaire SA, et al. Free fatty acids inhibit insulin signaling-stimulated endothelial nitric oxide synthase activation through upregulating PTEN or inhibiting Akt kinase. Diabetes. 2006;55:2301–10.Search in Google Scholar

[44] Kashyap S, Belfort R, Gastaldelli A, Pratipanawatr T, Berria R, Pratipanawatr W, et al. A sustained increase in plasma free fatty acids impairs insulin secretion in nondiabetic subjects genetically predisposed to develop type 2 diabetes. Diabetes. 2003;52:2461–74.Search in Google Scholar

[45] Lupi R, Dotta F, Marselli L, Del Guerra S, Masini M, Santangelo C, et al. Prolonged exposure to free fatty acids has cytostatic and pro-apoptotic effects on human pancreatic islets: evidence that beta-cell death is caspase mediated, partially dependent on ceramide pathway, and Bcl-2 regulated. Diabetes. 2002;51:1437–42.Search in Google Scholar

[46] Gastaldelli A, Coggan AR, Wolfe RR. Assessment of methods for improving tracer estimation of non-steady-state rate of appearance. J Appl Physiol. 1999;87:1813–22.Search in Google Scholar

[47] Natali A, Gastaldelli A, Galvan AQ, Sironi AM, Ciociaro D, Sanna G, et al. Effects of acute alpha 2-blockade on insulin action and secretion in humans. Am J Physiol. 1998;274(Pt 1):E57–64.Search in Google Scholar

[48] Lefebvre PJ. Glucagon and its family revisited. Diabetes Care. 1995;18:715–30.Search in Google Scholar

[49] Del Prato S, Castellino P, Simonson DC, DeFronzo RA. Hyperglucagonemia and insulin-mediated glucose metabolism. J Clin Invest. 1987;79:547–56.Search in Google Scholar

[50] Gravholt CH, Moller N, Jensen MD, Christiansen JS, Schmitz O. Physiological levels of glucagon do not influence lipolysis in abdominal adipose tissue as assessed by microdialysis. J Clin Endocrinol Metab. 2001;86:2085–9.Search in Google Scholar

[51] Lee MJ, Pramyothin P, Karastergiou K, Fried SK. Deconstructing the roles of glucocorticoids in adipose tissue biology and the development of central obesity. Biochim Biophys Acta. 2014;1842:473–81.Search in Google Scholar

[52] Garabedian MJ, Harris CA, Jeanneteau F. Glucocorticoid receptor action in metabolic and neuronal function. F1000Res. 2017;6:1208.Search in Google Scholar

[53] Woods CP, Hazlehurst JM, Tomlinson JW. Glucocorticoids and non-alcoholic fatty liver disease. J Steroid Biochem Mol Biol. 2015;154:94–103.Search in Google Scholar

[54] Campbell JE, Drucker DJ. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 2013;17:819–37.Search in Google Scholar

[55] Ceperuelo-Mallafre V, Duran X, Pachon G, Roche K, Garrido-Sanchez L, et al. Disruption of GIP/GIPR axis in human adipose tissue is linked to obesity and insulin resistance. J Clin Endocrinol Metab. 2014;99:E908–19.Search in Google Scholar

[56] Muscogiuri G, DeFronzo RA, Gastaldelli A, Holst JJ. Glucagon-like peptide-1 and the central/peripheral nervous system: crosstalk in diabetes. Trends Endocrinol Metab. 2017;28:88–103.Search in Google Scholar

[57] Arner P, Pettersson A, Mitchell PJ, Dunbar JD, Kharitonenkov A, Ryden M. FGF21 attenuates lipolysis in human adipocytes – a possible link to improved insulin sensitivity. FEBS Lett. 2008;582:1725–30.Search in Google Scholar

[58] Hotta Y, Nakamura H, Konishi M, Murata Y, Takagi H, Matsumura S, et al. Fibroblast growth factor 21 regulates lipolysis in white adipose tissue but is not required for ketogenesis and triglyceride clearance in liver. Endocrinology. 2009;150:4625–33.Search in Google Scholar

[59] Groop LC, Bonadonna RC, DelPrato S, Ratheiser K, Zyck K, Ferrannini E, et al. Glucose and free fatty acid metabolism in non-insulin-dependent diabetes mellitus. Evidence for multiple sites of insulin resistance. J Clin Invest. 1989;84:205–13.Search in Google Scholar

[60] Gastaldelli A, Cusi K, Pettiti M, Hardies J, Miyazaki Y, Berria R, et al. Relationship between hepatic/visceral fat and hepatic insulin resistance in nondiabetic and type 2 diabetic subjects. Gastroenterology. 2007;133:496–506.Search in Google Scholar

[61] Bugianesi E, Gastaldelli A, Vanni E, Gambino R, Cassader M, Baldi S, et al. Insulin resistance in non-diabetic patients with non-alcoholic fatty liver disease: sites and mechanisms. Diabetologia. 2005;48:634–42.Search in Google Scholar

[62] Sondergaard E, Espinosa De Ycaza AE, Morgan-Bathke M, Jensen MD. How to measure adipose tissue insulin sensitivity. J Clin Endocrinol Metab. 2017;102:1193–9.Search in Google Scholar

[63] Fabbrini E, deHaseth D, Deivanayagam S, Mohammed BS, Vitola BE, Klein S. Alterations in fatty acid kinetics in obese adolescents with increased intrahepatic triglyceride content. Obesity (Silver Spring). 2009;17:25–9.Search in Google Scholar

[64] Gastaldelli A, Gaggini M, DeFronzo RA. Role of adipose tissue insulin resistance in the natural history of type 2 diabetes: results from the San Antonio metabolism study. Diabetes. 2017;66:815–22.Search in Google Scholar

[65] Gastaldelli A. Role of beta-cell dysfunction, ectopic fat accumulation and insulin resistance in the pathogenesis of type 2 diabetes mellitus. Diabetes Res Clin Pract. 2011;93(Suppl 1):S60–5.Search in Google Scholar

[66] Gaggini M, Morelli M, Buzzigoli E, DeFronzo RA, Bugianesi E, Gastaldelli A. Non-alcoholic fatty liver disease (NAFLD) and its connection with insulin resistance, dyslipidemia, atherosclerosis and coronary heart disease. Nutrients. 2013;5:1544–60.Search in Google Scholar

[67] Lomonaco R, Ortiz-Lopez C, Orsak B, Webb A, Hardies J, Darland C, et al. Effect of adipose tissue insulin resistance on metabolic parameters and liver histology in obese patients with nonalcoholic fatty liver disease. Hepatology. 2012;55:1389–97.Search in Google Scholar

[68] Bell LN, Wang J, Muralidharan S, Chalasani S, Fullenkamp AM, Wilson LA, et al. Relationship between adipose tissue insulin resistance and liver histology in nonalcoholic steatohepatitis: a pioglitazone versus vitamin E versus placebo for the treatment of nondiabetic patients with nonalcoholic steatohepatitis trial follow-up study. Hepatology. 2012;56:1311–8.Search in Google Scholar

[69] Gastaldelli A, Harrison SA, Belfort-Aguilar R, Hardies LJ, Balas B, Schenker S, et al. Importance of changes in adipose tissue insulin resistance to histological response during thiazolidinedione treatment of patients with nonalcoholic steatohepatitis. Hepatology. 2009;50:1087–93.Search in Google Scholar

[70] Gastaldelli A, Gaggini M, Daniele G, Ciociaro D, Cersosimo E, Tripathy D, et al. Exenatide improves both hepatic and adipose tissue insulin resistance: a dynamic positron emission tomography study. Hepatology. 2016;64:2028–37.Search in Google Scholar

[71] DeFronzo RA, Tripathy D, Schwenke DC, Banerji M, Bray GA, Buchanan TA, et al. Prevention of diabetes with pioglitazone in ACT NOW: physiologic correlates. Diabetes. 2013;62:3920–6.Search in Google Scholar

[72] Wildman RP, Muntner P, Reynolds K, McGinn AP, Rajpathak S, Wylie-Rosett J, et al. The obese without cardiometabolic risk factor clustering and the normal weight with cardiometabolic risk factor clustering: prevalence and correlates of 2 phenotypes among the US population (NHANES 1999–2004). Arch Intern Med. 2008;168:1617–24.Search in Google Scholar

[73] Bluher M. Are there still healthy obese patients? Curr Opin Endocrinol Diabetes Obes. 2012;19:341–6.Search in Google Scholar

[74] Pataky Z, Makoundou V, Nilsson P, Gabriel RS, Lalic K, Muscelli E, et al. Metabolic normality in overweight and obese subjects. Which parameters? Which risks? Int J Obes. 2011;35:1208–15.Search in Google Scholar

[75] Eckel N, Meidtner K, Kalle-Uhlmann T, Stefan N, Schulze MB. Metabolically healthy obesity and cardiovascular events: a systematic review and meta-analysis. Eur J Prev Cardiol. 2016;23:956–66.Search in Google Scholar

[76] McLaughlin T, Lamendola C, Coghlan N, Liu TC, Lerner K, Sherman A, et al. Subcutaneous adipose cell size and distribution: relationship to insulin resistance and body fat. Obesity (Silver Spring). 2014;22:673–80.Search in Google Scholar

[77] Scherer SS. The debut of a rational treatment for an inherited neuropathy? J Clin Invest. 2011;121:4624–7.Search in Google Scholar

[78] McLaughlin T, Abbasi F, Lamendola C, Reaven G. Heterogeneity in the prevalence of risk factors for cardiovascular disease and type 2 diabetes mellitus in obese individuals: effect of differences in insulin sensitivity. Arch Intern Med. 2007;167:642–8.Search in Google Scholar

[79] Ruderman NB, Schneider SH, Berchtold P. The “metabolically-obese,” normal-weight individual. Am J Clin Nutr. 1981;34:1617–21.Search in Google Scholar

[80] Pou KM, Massaro JM, Hoffmann U, Lieb K, Vasan RS, O’Donnell CJ, et al. Patterns of abdominal fat distribution: the Framingham Heart Study. Diabetes Care. 2009;32:481–5.Search in Google Scholar

[81] Isokuortti E, Zhou Y, Peltonen M, Bugianesi E, Clement K, Bonnefont-Rousselot D, et al. Use of HOMA-IR to diagnose non-alcoholic fatty liver disease: a population-based and inter-laboratory study. Diabetologia. 2017;60:1873–1882.Search in Google Scholar

[82] Muir LA, Neeley CK, Meyer KA, Baker NA, Brosius AM, Washabaugh AR, et al. Adipose tissue fibrosis, hypertrophy, and hyperplasia: correlations with diabetes in human obesity. Obes (Silver Spring). 2016;24:597–605.Search in Google Scholar

[83] O’Connell J, Lynch L, Cawood TJ, Kwasnik A, Nolan N, Geoghegan J, et al. The relationship of omental and subcutaneous adipocyte size to metabolic disease in severe obesity. PLoS One. 2010;5:e9997.Search in Google Scholar

[84] Sidossis LS, Porter C, Saraf MK, Borsheim E, Radhakrishnan RS, Chao T, et al. Browning of subcutaneous white adipose tissue in humans after severe adrenergic stress. Cell Metab. 2015;22:219–27.Search in Google Scholar

[85] Liu X, Wang S, You Y, Meng M, Zheng Z, Dong M, et al. Brown adipose tissue transplantation reverses obesity in Ob/Ob mice. Endocrinology. 2015;156:2461–9.Search in Google Scholar

[86] Morrison SF, Madden CJ, Tupone D. Central neural regulation of brown adipose tissue thermogenesis and energy expenditure. Cell Metab. 2014;19:741–56.Search in Google Scholar

[87] Chondronikola M, Volpi E, Borsheim E, Porter C, Annamalai P, Enerback S, et al. Brown adipose tissue improves whole-body glucose homeostasis and insulin sensitivity in humans. Diabetes. 2014;63:4089–99.Search in Google Scholar

[88] Ouellet V, Labbe SM, Blondin DP, Phoenix S, Guerin B, Haman F, et al. Brown adipose tissue oxidative metabolism contributes to energy expenditure during acute cold exposure in humans. J Clin Invest. 2012;122:545–52.Search in Google Scholar

[89] van der Lans AA, Hoeks J, Brans B, Vijgen GH, Visser MG, Vosselman MJ, et al. Cold acclimation recruits human brown fat and increases nonshivering thermogenesis. J Clin Invest. 2013;123:3395–403.Search in Google Scholar

[90] Blondin DP, Labbe SM, Tingelstad HC, Noll C, Kunach M, Phoenix S, et al. Increased brown adipose tissue oxidative capacity in cold-acclimated humans. J Clin Endocrinol Metab. 2014;99:E438–46.Search in Google Scholar

[91] Robinson LJ, Law JM, Symonds ME, Budge H. Brown adipose tissue activation as measured by infrared thermography by mild anticipatory psychological stress in lean healthy females. Exp Physiol. 2016;101:549–57.Search in Google Scholar

[92] Ramage LE, Akyol M, Fletcher AM, Forsythe J, Nixon M, Carter RN, et al. Glucocorticoids acutely increase brown adipose tissue activity in humans, revealing species-specific differences in UCP-1 regulation. Cell Metab. 2016;24:130–41.Search in Google Scholar

[93] Hoeke G, Kooijman S, Boon MR, Rensen PC, Berbee JF. Role of brown fat in lipoprotein metabolism and atherosclerosis. Circ Res. 2016;118:173–82.Search in Google Scholar

[94] Kuji I, Imabayashi E, Minagawa A, Matsuda H, Miyauchi T. Brown adipose tissue demonstrating intense FDG uptake in a patient with mediastinal pheochromocytoma. Ann Nucl Med. 2008;22:231–5.Search in Google Scholar

[95] Zuriaga MA, Fuster JJ, Gokce N, Walsh K. Humans and mice display opposing patterns of “browning” gene expression in visceral and subcutaneous white adipose tissue depots. Front Cardiovasc Med. 2017;4:27.Search in Google Scholar

[96] Fisher FM, Kleiner S, Douris N, Fox EC, Mepani RJ, Verdeguer F, et al. FGF21 regulates PGC-1 alpha and browning of white adipose tissues in adaptive thermogenesis. Gene Dev. 2012;26:271–81.Search in Google Scholar

[97] Hondares E, Iglesias R, Giralt A, Gonzalez FJ, Giralt M, Mampel T, et al. Thermogenic activation induces FGF21 expression and release in brown adipose tissue. J Biol Chem. 2011;286:12983–90.Search in Google Scholar

[98] Lee P, Linderman JD, Smith S, Brychta RJ, Wang J, Idelson C, et al. Irisin and FGF21 are cold-induced endocrine activators of brown fat function in humans. Cell Metab. 2014;19:302–9.Search in Google Scholar

[99] Berti L, Irmler M, Zdichavsky M, Meile T, Bohm A, Stefan N, et al. Fibroblast growth factor 21 is elevated in metabolically unhealthy obesity and affects lipid deposition, adipogenesis, and adipokine secretion of human abdominal subcutaneous adipocytes. Mol Metab. 2015;4:519–27.Search in Google Scholar

[100] Giralt M, Gavalda-Navarro A, Villarroya F. Fibroblast growth factor-21, energy balance and obesity. Mol Cell Endocrinol. 2015;418(Pt 1):66–73.Search in Google Scholar

[101] Lockie SH, Heppner KM, Chaudhary N, Chabenne JR, Morgan DA, Veyrat-Durebex C, et al. Direct control of brown adipose tissue thermogenesis by central nervous system glucagon-like peptide-1 receptor signaling. Diabetes. 2012;61:2753–62.Search in Google Scholar

[102] Gastaldelli A, Gaggini M, DeFronzo R. Glucose kinetics: an update and novel insights into its regulation by glucagon and GLP-1. Curr Opin Clin Nutr Metab Care. 2017;20:300–9.Search in Google Scholar

[103] Beiroa D, Imbernon M, Gallego R, Senra A, Herranz D, Villarroya F, et al. GLP-1 agonism stimulates brown adipose tissue thermogenesis and browning through hypothalamic AMPK. Diabetes. 2014;63:3346–58.Search in Google Scholar

[104] Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004;84:277–359.Search in Google Scholar

[105] Hanssen MJ, Wierts R, Hoeks J, Gemmink A, Brans B, Mottaghy FM, et al. Glucose uptake in human brown adipose tissue is impaired upon fasting-induced insulin resistance. Diabetologia. 2015;58:586–595.Search in Google Scholar

[106] Orava J, Nuutila P, Lidell ME, Oikonen V, Noponen T, Viljanen T, et al. Different metabolic responses of human brown adipose tissue to activation by cold and insulin. Cell Metab. 2011;14:272–9.Search in Google Scholar

[107] Blondin DP, Frisch F, Phoenix S, Guerin B, Turcotte EE, Haman F, et al. Inhibition of intracellular triglyceride lipolysis suppresses cold-induced brown adipose tissue metabolism and increases shivering in humans. Cell Metab. 2017;25:438–47.Search in Google Scholar

[108] Guerra C, Navarro P, Valverde AM, Arribas M, Bruning J, Kozak LP, et al. Brown adipose tissue-specific insulin receptor knockout shows diabetic phenotype without insulin resistance. J Clin Invest. 2001;108:1205–13.Search in Google Scholar

[109] Blondin DP, Tingelstad HC, Noll C, Frisch F, Phoenix S, Guerin B, et al. Dietary fatty acid metabolism of brown adipose tissue in cold-acclimated men. Nat Commun. 2017;8:14146.Search in Google Scholar

[110] Chondronikola M, Volpi E, Borsheim E, Porter C, Saraf MK, Annamalai P, et al. Brown adipose tissue activation is linked to distinct systemic effects on lipid metabolism in humans. Cell Metab. 2016;23:1200–6.Search in Google Scholar

[111] Bartelt A, Bruns OT, Reimer R, Hohenberg H, Ittrich H, Peldschus K, et al. Brown adipose tissue activity controls triglyceride clearance. Nat Med. 2011;17:200–5.Search in Google Scholar

[112] Dijk W, Heine M, Vergnes L, Boon MR, Schaart G, Hesselink MK, et al. ANGPTL4 mediates shuttling of lipid fuel to brown adipose tissue during sustained cold exposure. Elife. 2015;4:e08428.Search in Google Scholar

[113] Dong M, Yang X, Lim S, Cao Z, Honek J, Lu H, et al. Cold exposure promotes atherosclerotic plaque growth and instability via UCP1-dependent lipolysis. Cell Metab. 2013;18:118–29.Search in Google Scholar

[114] Matsushita M, Yoneshiro T, Aita S, Kameya T, Sugie H, Saito M. Impact of brown adipose tissue on body fatness and glucose metabolism in healthy humans. Int J Obes. 2014;38:812–7.Search in Google Scholar

[115] De Lorenzo F, Mukherjee M, Kadziola Z, Sherwood R, Kakkar VV. Central cooling effects in patients with hypercholesterolaemia. Clin Sci (Lond). 1998;95:213–7.Search in Google Scholar

Received: 2017-07-28
Accepted: 2017-08-29
Published Online: 2017-09-25

©2017 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 1.6.2024 from https://www.degruyter.com/document/doi/10.1515/hmbci-2017-0060/html
Scroll to top button