Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter July 21, 2017

Hormonal factors in the control of the browning of white adipose tissue

  • Jiamiao Hu and Mark Christian EMAIL logo

Abstract

Adipose tissue has been historically classified into anabolic white adipose tissue (WAT) and catabolic brown adipose tissue (BAT). Recent studies have revealed the plasticity of WAT, where white adipocytes can be induced into ‘brown-like’ heat-producing adipocytes (BRITE or beige adipocytes). Recruiting and activating BRITE adipocytes in WAT (so-called ‘browning’) is believed to provide new avenues for the treatment of obesity-related diseases. A number of hormonal factors have been found to regulate BRITE adipose development and activity through autocrine, paracrine and systemic mechanisms. In this mini-review we will discuss the impact of these factors on the browning process, especially those hormonal factors identified with direct effects on white adipocytes.

Author statement

  1. Research funding: Authors state no funding involved.

  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 animal use.

References

[1] Rosell M, Kaforou M, Frontini A, Okolo A, Chan YW, Nikolopoulou E, et al. Brown and white adipose tissues: intrinsic differences in gene expression and response to cold exposure in mice. Am J Physiol Endocrinol Metab. 2014;306:E945–64.Search in Google Scholar

[2] Chakraborty D, Bhattacharya A, Mittal BR. Patterns of brown fat uptake of 18F-fluorodeoxyglucose in positron emission tomography/computed tomography scan. Indian J Nucl Med. 2015;30:320–2.Search in Google Scholar

[3] Kozak LP. Brown fat and the myth of diet-induced thermogenesis. Cell Metab. 2010;11:263–57.Search in Google Scholar

[4] Himms-Hagen J, Cui J, Danforth E, Taatjes DJ, Lang SS, Waters BL, et al. Effect of CL-316,243, a thermogenic beta 3-agonist, on energy balance and brown and white adipose tissues in rats. Am J Physiol. 1994;266:R1371–82.Search in Google Scholar

[5] Cousin B, Cinti S, Morroni M, Raimbault S, Ricquier D, Penicaud L, et al. Occurrence of brown adipocytes in rat white adipose tissue: molecular and morphological characterization. J Cell Sci. 1992;103:931–42.Search in Google Scholar

[6] Shan T, Liang X, Bi P, Zhang P, Liu W, Kuang S. Distinct populations of adipogenic and myogenic Myf5-lineage progenitors in white adipose tissues. J Lipid Res. 2013;54:2214–24.Search in Google Scholar

[7] 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

[8] Carey AL, Vorlander C, Reddy-Luthmoodoo M, Natoli AK, Formosa MF, Bertovic DA, et al. Reduced UCP-1 content in in vitro differentiated beige/brite adipocytes derived from preadipocytes of human subcutaneous white adipose tissues in obesity. PLoS One. 2014;e919979.Search in Google Scholar

[9] Sharp LZ, Shinoda K, Ohno H, Scheel DW, Tomoda E, Ruiz L, et al. Human BAT possesses molecular signatures that resemble beige/brite cells. PLoS One. 2012;e494527.Search in Google Scholar

[10] Villarroya F, Vidal-Puig A. Beyond the sympathetic tone: the new brown fat activators. Cell Metab. 2013;17:638–43.Search in Google Scholar

[11] Kim SH, Plutzky J. Brown fat and browning for the treatment of obesity and related metabolic disorders. Diabetes Metab J. 2016;40:12–21.Search in Google Scholar

[12] Collins S, Yehuda-Shnaidman E, Wang H. Positive and negative control of Ucp1 gene transcription and the role of beta-adrenergic signaling networks. Int J Obes (Lond). 2010;34:S28–33.Search in Google Scholar

[13] Fedorenko A, Lishko PV, Kirichok Y. Mechanism of fatty-acid-dependent UCP1 uncoupling in brown fat mitochondria. Cell. 2012;151:400–13.Search in Google Scholar

[14] Ricquier D. Uncoupling protein 1 of brown adipocytes, the only uncoupler: a historical perspective. Front Endocrinol (Lausanne). 2011;2:85.Search in Google Scholar

[15] Rim JS, Kozak LP. Regulatory motifs for CREB-binding protein and Nfe2l2 transcription factors in the upstream enhancer of the mitochondrial uncoupling protein 1 gene. J Biol Chem. 2002;277:34589–600.Search in Google Scholar

[16] Villarroya F, Iglesias R, Giralt M. PPARs in the control of uncoupling proteins gene expression. PPAR Res. 2007;2007:74364.Search in Google Scholar

[17] Cannon B, Jacobsson A, Rehnmark S, Nedergaard J. Signal transduction in brown adipose tissue recruitment: noradrenaline and beyond. Int J Obes Relat Metab Disord. 1996;20:S36–42.Search in Google Scholar

[18] Laiglesia LM, Lorente-Cebrian S, Prieto-Hontoria PL, Fernandez-Galilea M, Ribeiro SM, Sainz N, et al. Eicosapentaenoic acid promotes mitochondrial biogenesis and beige-like features in subcutaneous adipocytes from overweight subjects. J Nutr Biochem. 2016;37:76–82.Search in Google Scholar

[19] Cinti S. The adipose organ at a glance. Dis Model Mech. 2012;5:588–94.Search in Google Scholar

[20] Matthias A, Ohlson KB, Fredriksson JM, Jacobsson A, Nedergaard J, Cannon B. Thermogenic responses in brown fat cells are fully UCP1-dependent. UCP2 or UCP3 do not substitute for UCP1 in adrenergically or fatty scid-induced thermogenesis. J Biol Chem. 2000;275:25073–81.Search in Google Scholar

[21] Puigserver P, Pico C, Stock MJ, Palou A. Effect of selective beta-adrenoceptor stimulation on UCP synthesis in primary cultures of brown adipocytes. Mol Cell Endocrinol. 1996;117:7–16.Search in Google Scholar

[22] Wu Q, Kazantzis M, Doege H, Ortegon AM, Tsang B, Falcon A, et al. Fatty acid transport protein 1 is required for nonshivering thermogenesis in brown adipose tissue. Diabetes. 2006;55:3229–37.Search in Google Scholar

[23] Galitzky J, Carpene C, Bousquet-Melou A, Berlan M, Lafontan M. Differential activation of beta 1-, beta 2- and beta 3-adrenoceptors by catecholamines in white and brown adipocytes. Fundam Clin Pharmacol. 1995;9:324–31.Search in Google Scholar

[24] Jimenez M, Barbatelli G, Allevi R, Cinti S, Seydoux J, Giacobino JP, et al. Beta 3-adrenoceptor knockout in C57BL/6J mice depresses the occurrence of brown adipocytes in white fat. Eur J Biochem. 2003;270:699–705.Search in Google Scholar

[25] de Jong Jasper M. A., Wouters René T. F., Boulet Nathalie, Cannon Barbara, Nedergaard Jan, Petrovic Natasa. The β 3 -adrenergic receptor is dispensable for browning of adipose tissues. American Journal of Physiology - Endocrinology And Metabolism. 2017 2 21;312(6):E508–E518. DOI:10.1152/ajpendo.00437.2016.Search in Google Scholar PubMed

[26] 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

[27] Cypess AM, Weiner LS, Roberts-Toler C, Franquet Elia E, Kessler SH, Kahn PA, et al. Activation of human brown adipose tissue by a beta3-adrenergic receptor agonist. Cell Metab. 2015;21:33–8.Search in Google Scholar

[28] Christian M. Nuclear receptor-mediated regulation of lipid droplet-associated protein gene expression in adipose tissue. Horm Mol Biol Clin Investig. 2013;14:87–97.Search in Google Scholar

[29] Weiner J, Kranz M, Klöting N, Kunath A, Steinhoff K, Rijntjes E, et al. Thyroid hormone status defines brown adipose tissue activity and browning of white adipose tissues in mice. Sci Rep. 2016;6:38124.Search in Google Scholar

[30] Lopez M, Varela L, Vazquez MJ, Rodriguez-Cuenca S, Gonzalez CR, Velagapudi VR, et al. Hypothalamic AMPK and fatty acid metabolism mediate thyroid regulation of energy balance. Nat Med. 2010;16:1001–8.Search in Google Scholar

[31] Cassard-Doulcier AM, Larose M, Matamala JC, Champigny O, Bouillaud F, Ricquier D. In vitro interactions between nuclear proteins and uncoupling protein gene promoter reveal several putative transactivating factors including Ets1, retinoid X receptor, thyroid hormone receptor, and a CACCC box-binding protein. J Biol Chem. 1994;269:24335–42.Search in Google Scholar

[32] de Jesus LA, Carvalho SD, Ribeiro MO, Schneider M, Kim SW, Harney JW, et al. The type 2 iodothyronine deiodinase is essential for adaptive thermogenesis in brown adipose tissue. J Clin Invest. 2001;108:1379–85.Search in Google Scholar

[33] Krause K, Kranz M, Weiner J, Klöting N, Rijntjes E, Köhrle J, et al. Influence of thyroid hormones on brown adipose tissue activity and browning of white adipose tissues in mice. Exp Clin Endocrinol Diabetes. 2015;123:LB_10.Search in Google Scholar

[34] Lin JZ, Martagon AJ, Cimini SL, Gonzalez DD, Tinkey DW, Biter A, et al. Pharmacological activation of thyroid hormone receptors elicits a functional conversion of white to brown fat. Cell Rep. 2015;13:1528–37.Search in Google Scholar

[35] Basse AL, Dixen K, Yadav R, Tygesen MP, Qvortrup K, Kristiansen K, et al. Global gene expression profiling of brown to white adipose tissue transformation in sheep reveals novel transcriptional components linked to adipose remodeling. BMC Geno. 2015;16:215.Search in Google Scholar

[36] Miao Y, Wu W, Dai Y, Maneix L, Huang B, Warner M, et al. Liver X receptor beta controls thyroid hormone feedback in the brain and regulates browning of subcutaneous white adipose tissue. Proc Natl Acad Sci USA. 2015;112:14006–11.Search in Google Scholar

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

[38] Bostrom P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, et al. A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature. 2012;481:463–8.Search in Google Scholar

[39] Zhang Y, Li R, Meng Y, Li S, Donelan W, Zhao Y, et al. Irisin stimulates browning of white adipocytes through mitogen-activated protein kinase p38 MAP kinase and ERK MAP kinase signaling. Diabetes. 2014;63:514–25.Search in Google Scholar

[40] Zhang Y, Xie C, Wang H, Foss RM, Clare M, George EV, et al. Irisin exerts dual effects on browning and adipogenesis of human white adipocytes. Am J Physiol Endocrinol Metab. 2016;311:E530–41.Search in Google Scholar

[41] 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

[42] Owen BM, Ding X, Morgan DA, Coate KC, Bookout AL, Rahmouni K, et al. FGF21 acts centrally to induce sympathetic nerve activity, energy expenditure, and weight loss. Cell Metab. 2014;20:670–7.Search in Google Scholar

[43] Douris N, Stevanovic DM, Fisher FM, Cisu TI, Chee MJ, Nguyen NL, et al. Central fibroblast growth factor 21 browns white fat via sympathetic action in male mice. Endocrinology. 2015;156:2470–81.Search in Google Scholar

[44] Mu J, Pinkstaff J, Li Z, Skidmore L, Li N, Myler H, et al. FGF21 analogs of sustained action enabled by orthogonal biosynthesis demonstrate enhanced antidiabetic pharmacology in rodents. Diabetes. 2012;61:505–12.Search in Google Scholar

[45] Hecht R, Li YS, Sun J, Belouski E, Hall M, Hager T, et al. Rationale-based engineering of a potent long-acting FGF21 analog for the treatment of type 2 Diabetes. PLoS One. 2012;e493457.Search in Google Scholar

[46] Huang J, Ishino T, Chen G, Rolzin P, Osothprarop TF, Retting K, et al. Development of a novel long-acting antidiabetic FGF21 mimetic by targeted conjugation to a scaffold antibody. J Pharmacol Exp Ther. 2013;346:270–80.Search in Google Scholar

[47] Veniant MM, Sivits G, Helmering J, Komorowski R, Lee J, Fan W, et al. Pharmacologic effects of FGF21 are independent of the “Browning” of white adipose tissue. Cell Metab. 2015;21:731–8.Search in Google Scholar

[48] Jeanson Y, Ribas F, Galinier A, Arnaud E, Ducos M, Andre M, et al. Lactate induces FGF21 expression in adipocytes through a p38-MAPK pathway. Biochem J. 2016;473:685–92.Search in Google Scholar

[49] Quesada-Lopez T, Cereijo R, Turatsinze JV, Planavila A, Cairo M, Gavalda-Navarro A, et al. The lipid sensor GPR120 promotes brown fat activation and FGF21 release from adipocytes. Nat Commun. 2016;7:13479.Search in Google Scholar

[50] Liang Q, Zhong L, Zhang J, Wang Y, Bornstein SR, Triggle CR, et al. FGF21 maintains glucose homeostasis by mediating the cross talk between liver and brain during prolonged fasting. Diabetes. 2014;63:4064–75.Search in Google Scholar

[51] Ribas F, Villarroya J, Hondares E, Giralt M, Villarroya F. FGF21 expression and release in muscle cells: involvement of MyoD and regulation by mitochondria-driven signalling. Biochem J. 2014;463:191–9.Search in Google Scholar

[52] Vandanmagsar B, Warfel JD, Wicks SE, Ghosh S, Salbaum JM, Burk D, et al. Impaired mitochondrial fat oxidation induces FGF21 in muscle. Cell Rep. 2016;15:1686–99.Search in Google Scholar

[53] Rao RR, Long JZ, White JP, Svensson KJ, Lou J, Lokurkar I, et al. Meteorin-like is a hormone that regulates immune-adipose interactions to increase beige fat thermogenesis. Cell. 2014;157:1279–91.Search in Google Scholar

[54] Mossenbock K, Vegiopoulos A, Rose AJ, Sijmonsma TP, Herzig S, Schafmeier T. Browning of white adipose tissue uncouples glucose uptake from insulin signaling. PLoS One. 2014;e1104289.Search in Google Scholar

[55] Dodd GT, Decherf S, Loh K, Simonds SE, Wiede F, Balland E, et al. Leptin and insulin act on POMC neurons to promote the browning of white fat. Cell. 2015;160:88–104.Search in Google Scholar

[56] Billington CJ, Bartness TJ, Briggs J, Levine AS, Morley JE. Glucagon stimulation of brown adipose tissue growth and thermogenesis. Am J Physiol. 1987;252:R160–5.Search in Google Scholar

[57] Kinoshita K, Ozaki N, Takagi Y, Murata Y, Oshida Y, Hayashi Y. Glucagon is essential for adaptive thermogenesis in brown adipose tissue. Endocrinology. 2014;155:3484–92.Search in Google Scholar

[58] Salem V, Izzi-Engbeaya C, Coello C, Thomas DB, Chambers ES, Comninos AN, et al. Glucagon increases energy expenditure independently of brown adipose tissue activation in humans. Diabetes Obes Metab. 2016;18:72–81.Search in Google Scholar

[59] 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

[60] 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

[61] Fernandes-Santos C, Zhang Z, Morgan DA, Guo DF, Russo AF, Rahmouni K. Amylin acts in the central nervous system to increase sympathetic nerve activity. Endocrinology. 2013;154:2481–8.Search in Google Scholar

[62] Yoshimatsu H, Egawa M, Bray GA. Effects of cholecystokinin on sympathetic activity to interscapular brown adipose tissue. Brain Res. 1992;597:298–303.Search in Google Scholar

[63] Madden CJ. Systemic CCK increases brown adipose tissue sympathetic nerve activity. FASEB J. 2013;27:1120–5.Search in Google Scholar

[64] Skibicka KP, Hansson C, Alvarez-Crespo M, Friberg PA, Dickson SL. Ghrelin directly targets the ventral tegmental area to increase food motivation. Neuroscience. 2011;180:129–37.Search in Google Scholar

[65] Tschop M, Smiley DL, Heiman ML. Ghrelin induces adiposity in rodents. Nature. 2000;407:908–13.Search in Google Scholar

[66] Cowley MA, Smith RG, Diano S, Tschop M, Pronchuk N, Grove KL, et al. The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Neuron. 2003;37:649–61.Search in Google Scholar

[67] Yasuda T, Masaki T, Kakuma T, Yoshimatsu H. Centrally administered ghrelin suppresses sympathetic nerve activity in brown adipose tissue of rats. Neurosci Lett. 2003;349:75–8.Search in Google Scholar

[68] Nagase H, Bray GA, York DA. Effect of galanin and enterostatin on sympathetic nerve activity to interscapular brown adipose tissue. Brain Res. 1996;709:44–50.Search in Google Scholar

[69] Teodoro JS, Zouhar P, Flachs P, Bardova K, Janovska P, Gomes AP, et al. Enhancement of brown fat thermogenesis using chenodeoxycholic acid in mice. Int J Obes (Lond). 2014;38:1027–34.Search in Google Scholar

[70] Zietak M, Kozak LP. Bile acids induce uncoupling protein 1-dependent thermogenesis and stimulate energy expenditure at thermoneutrality in mice. Am J Physiol Endocrinol Metab. 2016;310:E346–54.Search in Google Scholar

[71] Broeders EP, Nascimento EB, Havekes B, Brans B, Roumans KH, Tailleux A, et al. The bile acid chenodeoxycholic acid increases human brown adipose tissue activity. Cell Metab. 2015;22:418–26.Search in Google Scholar

[72] Fang S, Suh JM, Reilly SM, Yu E, Osborn O, Lackey D, et al. Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nat Med. 2015;21:159–65.Search in Google Scholar

[73] Steculorum SM, Ruud J, Karakasilioti I, Backes H, Engstrom Ruud L, Timper K, et al. AgRP neurons control systemic insulin sensitivity via myostatin expression in brown adipose tissue. Cell. 2016;165:125–38.Search in Google Scholar

[74] Zhang W, Bi S. Hypothalamic regulation of brown adipose tissue thermogenesis and energy homeostasis. Front Endocrinol (Lausanne). 2015;6:136.Search in Google Scholar

[75] Ruan HB, Dietrich MO, Liu ZW, Zimmer MR, Li MD, Singh JP, et al. O-GlcNAc transferase enables AgRP neurons to suppress browning of white fat. Cell. 2014;159:306–17.Search in Google Scholar

[76] Turtzo LC, Marx R, Lane MD. Cross-talk between sympathetic neurons and adipocytes in coculture. Proc Natl Acad Sci USA. 2001;98:12385–90.Search in Google Scholar

[77] Bi S. Dorsomedial hypothalamic NPY modulation of adiposity and thermogenesis. Physiol Behav. 2013;121:56–60.Search in Google Scholar

[78] Blevins JE, Ho JM. Role of oxytocin signaling in the regulation of body weight. Rev Endocr Metab Disord. 2013;14:311–29.Search in Google Scholar

[79] Klockars A, Levine AS, Olszewski PK. Central oxytocin and food intake: focus on macronutrient-driven reward. Front Endocrinol. 2015;6:65.Search in Google Scholar

[80] Takayanagi Y, Kasahara Y, Onaka T, Takahashi N, Kawada T, Nishimori K. Oxytocin receptor-deficient mice developed late-onset obesity. Neuroreport. 2008;19:951–5.Search in Google Scholar

[81] Plante E, Menaouar A, Danalache BA, Yip D, Broderick TL, Chiasson JL, et al. Oxytocin treatment prevents the cardiomyopathy observed in obese diabetic male db/db mice. Endocrinology. 2015;156:1416–28.Search in Google Scholar

[82] LeFeuvre RA, Rothwell NJ, Stock MJ. Activation of brown fat thermogenesis in response to central injection of corticotropin releasing hormone in the rat. Neuropharmacology. 1987;26:1217–21.Search in Google Scholar

[83] Seres J, Bornstein SR, Seres P, Willenberg HS, Schulte KM, Scherbaum WA, et al. Corticotropin-releasing hormone system in human adipose tissue. J Clin Endocrinol Metab. 2004;89:965–70.Search in Google Scholar

[84] Lu B, Markovic D, Pessin J, Lehnert H, Grammatopoulos D. Unique roles of CRH-Rs controlling adipocyte biology and transdifferentiation. Endocrine Abstracts. 2012;28:P218.Search in Google Scholar

[85] Lu B, Diz-Chaves Y, Markovic D, Contarino A, Penicaud L, Fanelli F, et al. The corticotrophin-releasing factor/urocortin system regulates white fat browning in mice through paracrine mechanisms. Int J Obes (Lond). 2015;39:408–17.Search in Google Scholar

[86] Wang C, Billington CJ, Levine AS, Kotz CM. Effect of CART in the hypothalamic paraventricular nucleus on feeding and uncoupling protein gene expression. Neuroreport. 2000;11:3251–5.Search in Google Scholar

[87] Masaki T, Yoshimichi G, Chiba S, Yasuda T, Noguchi H, Kakuma T, et al. Corticotropin-releasing hormone-mediated pathway of leptin to regulate feeding, adiposity, and uncoupling protein expression in mice. Endocrinology. 2003;144:3547–54.Search in Google Scholar

[88] Beauregard C, Utz AL, Schaub AE, Nachtigall L, Biller BM, Miller KK, et al. Growth hormone decreases visceral fat and improves cardiovascular risk markers in women with hypopituitarism: a randomized, placebo-controlled study. J Clin Endocrinol Metab. 2008;93:2063–71.Search in Google Scholar

[89] Abrahamsen B, Nielsen TL, Hangaard J, Gregersen G, Vahl N, Korsholm L, et al. Dose-, IGF-I- and sex-dependent changes in lipid profile and body composition during GH replacement therapy in adult onset GH deficiency. Eur J Endocrinol. 2004;150:671–9.Search in Google Scholar

[90] Chihara K, Fujieda K, Shimatsu A, Miki T, Tachibana K. Dose-dependent changes in body composition during growth hormone (GH) treatment in Japanese patients with adult GH deficiency: a randomized, placebo-controlled trial. Growth Horm IGF Res. 2010;20:205–11.Search in Google Scholar

[91] Egecioglu E, Bjursell M, Ljungberg A, Dickson SL, Kopchick JJ, Bergstrom G, et al. Growth hormone receptor deficiency results in blunted ghrelin feeding response, obesity, and hypolipidemia in mice. Am J Physiol Endocrinol Metab. 2006;290:E317–25.Search in Google Scholar

[92] Li Y, Knapp JR, Kopchick JJ. Enlargement of interscapular brown adipose tissue in growth hormone antagonist transgenic and in growth hormone receptor gene-disrupted dwarf mice. Exp Biol Med (Maywood). 2003;228:207–15.Search in Google Scholar

[93] Sun LY. Growth Hormone-Releasing Hormone Disruption Extends Longevity, Regulates Response to Dietary Restriction, Promotes Browning of White Adipose Tissue and Ameliorates High Fat Diet-Induced Insulin Resistance.. Endocrine Society: GH, IGF and Posterior Pituitary 2015:FRI-448 Endocrine Society's 97th Annual Meeting and Expo, March 5–8, 2015 - San Diego.Search in Google Scholar

[94] Woo MN, Jeon SM, Shin YC, Lee MK, Kang MA, Choi MS. Anti-obese property of fucoxanthin is partly mediated by altering lipid-regulating enzymes and uncoupling proteins of visceral adipose tissue in mice. Mol Nutr Food Res. 2009;53:1603–11.Search in Google Scholar

[95] Kim M, Goto T, Yu R, Uchida K, Tominaga M, Kano Y, et al. Fish oil intake induces UCP1 upregulation in brown and white adipose tissue via the sympathetic nervous system. Sci Rep. 2015;5:18013.Search in Google Scholar

[96] Cao L, Choi EY, Liu X, Martin A, Wang C, Xu X, et al. White to brown fat phenotypic switch induced by genetic and environmental activation of a hypothalamic-adipocyte axis. Cell Metab. 2011;14:324–38.Search in Google Scholar

[97] Guerre-Millo M. Adipose tissue hormones. J Endocrinol Invest. 2002;25:855–61.Search in Google Scholar

[98] Commins SP, Watson PM, Padgett MA, Dudley A, Argyropoulos G, Gettys TW. Induction of uncoupling protein expression in brown and white adipose tissue by leptin. Endocrinology. 1999;140:292–300.Search in Google Scholar

[99] Commins SP, Watson PM, Levin N, Beiler RJ, Gettys TW. Central leptin regulates the UCP1 and ob genes in brown and white adipose tissue via different beta-adrenoceptor subtypes. J Biol Chem. 2000;275:33059–67.Search in Google Scholar

[100] Qian SW, Tang Y, Li X, Liu Y, Zhang YY, Huang HY, et al. BMP4-mediated brown fat-like changes in white adipose tissue alter glucose and energy homeostasis. Proc Natl Acad Sci USA. 2013;110:E798–807.Search in Google Scholar

[101] Tseng YH, Kokkotou E, Schulz TJ, Huang TL, Winnay JN, Taniguchi CM, et al. New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature. 2008;454:1000–4.Search in Google Scholar

[102] Whittle AJ, Carobbio S, Martins L, Slawik M, Hondares E, Vazquez MJ, et al. BMP8B increases brown adipose tissue thermogenesis through both central and peripheral actions. Cell. 2012;149:871–85.Search in Google Scholar

[103] 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

[104] Madsen L, Pedersen LM, Lillefosse HH, Fjaere E, Bronstad I, Hao Q, et al. UCP1 induction during recruitment of brown adipocytes in white adipose tissue is dependent on cyclooxygenase activity. PLoS One. 2010;5:e11391.Search in Google Scholar

[105] Vegiopoulos A, Muller-Decker K, Strzoda D, Schmitt I, Chichelnitskiy E, Ostertag A, et al. Cyclooxygenase-2 controls energy homeostasis in mice by de novo recruitment of brown adipocytes. Sci. 2010;328:1158–61.Search in Google Scholar

[106] Harms M, Seale P. Brown and beige fat: development, function and therapeutic potential. Nat Med. 2013;19:1252–63.Search in Google Scholar

[107] Murano I, Barbatelli G, Giordano A, Cinti S. Noradrenergic parenchymal nerve fiber branching after cold acclimatisation correlates with brown adipocyte density in mouse adipose organ. J Anat. 2009;214:171–8.Search in Google Scholar

[108] Asano A, Morimatsu M, Nikami H, Yoshida T, Saito M. Adrenergic activation of vascular endothelial growth factor mRNA expression in rat brown adipose tissue: implication in cold-induced angiogenesis. Biochem J. 1997;328:179–83.Search in Google Scholar

[109] Xue Y, Petrovic N, Cao R, Larsson O, Lim S, Chen S, et al. Hypoxia-independent angiogenesis in adipose tissues during cold acclimation. Cell Metab. 2009;9:99–109.Search in Google Scholar

[110] Elias I, Franckhauser S, Ferre T, Vila L, Tafuro S, Munoz S, et al. Adipose tissue overexpression of vascular endothelial growth factor protects against diet-induced obesity and insulin resistance. Diabetes. 2012;61:1801–13.Search in Google Scholar

[111] During MJ, Liu X, Huang W, Magee D, Slater A, McMurphy T, et al. Adipose VEGF links the white-to-brown fat switch with environmental, genetic, and pharmacological stimuli in male mice. Endocrinology. 2015;156:2059–73.Search in Google Scholar

[112] Ye L, Wu J, Cohen P, Kazak L, Khandekar MJ, Jedrychowski MP, et al. Fat cells directly sense temperature to activate thermogenesis. Proc Natl Acad Sci USA. 2013;110:12480–5.Search in Google Scholar

[113] Nguyen KD, Qiu Y, Cui X, Goh YP, Mwangi J, David T, et al. Alternatively activated macrophages produce catecholamines to sustain adaptive thermogenesis. Nature. 2011;480:104–8.Search in Google Scholar

[114] 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

[115] Iyer RB, Guo CC, Perrier N. Adrenal pheochromocytoma with surrounding brown fat stimulation. AJR Am J Roentgenol. 2009;192:300–1.Search in Google Scholar

[116] Bordicchia M, Liu D, Amri EZ, Ailhaud G, Dessi-Fulgheri P, Zhang C, et al. Cardiac natriuretic peptides act via p38 MAPK to induce the brown fat thermogenic program in mouse and human adipocytes. J Clin Invest. 2012;122:1022–36.Search in Google Scholar

[117] Moreno-Aliaga MJ, Perez-Echarri N, Marcos-Gomez B, Larequi E, Gil-Bea FJ, Viollet B, et al. Cardiotrophin-1 is a key regulator of glucose and lipid metabolism. Cell Metab. 2011;14:242–53.Search in Google Scholar

[118] van Baak MA, Hul GB, Toubro S, Astrup A, Gottesdiener KM, DeSmet M, et al. Acute effect of L-796568, a novel beta 3-adrenergic receptor agonist, on energy expenditure in obese men. Clin Pharmacol Ther. 2002;71:272–9.Search in Google Scholar

[119] Larsen TM, Toubro S, van Baak MA, Gottesdiener KM, Larson P, Saris WH, et al. Effect of a 28-d treatment with L-796568, a novel beta(3)-adrenergic receptor agonist, on energy expenditure and body composition in obese men. Am J Clin Nutr. 2002;76:780–8.Search in Google Scholar

[120] Baskaran P, Krishnan V, Ren J, Thyagarajan B. Capsaicin induces browning of white adipose tissue and counters obesity by activating TRPV1 channel-dependent mechanisms. Br J Pharmacol. 2016;173:2369–89.Search in Google Scholar

[121] Saito M, Yoneshiro T. Capsinoids and related food ingredients activating brown fat thermogenesis and reducing body fat in humans. Curr Opin Lipidol. 2013;24:71–7.Search in Google Scholar

[122] Maeda H, Hosokawa M, Sashima T, Funayama K, Miyashita K. Fucoxanthin from edible seaweed, Undaria pinnatifida, shows antiobesity effect through UCP1 expression in white adipose tissues. Biochem Biophys Res Commun. 2005;332:392–7.Search in Google Scholar

[123] Singhal V, Maffazioli GD, Ackerman KE, Lee H, Elia EF, Woolley R, et al. Effect of chronic athletic activity on brown fat in young women. PLoS One. 2016;e015635311.Search in Google Scholar

Received: 2017-04-03
Accepted: 2017-05-11
Published Online: 2017-07-21

©2017 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 10.5.2024 from https://www.degruyter.com/document/doi/10.1515/hmbci-2017-0017/html
Scroll to top button