Human adipose dynamics and metabolic health

Bin Feng, Tracy Zhang, Haiyan Xu, Bin Feng, Tracy Zhang, Haiyan Xu

Abstract

The two types of adipose tissue in humans, white and brown, have distinct developmental origins and functions. Human white adipose tissue plays a pivotal role in maintaining whole-body energy homeostasis by storing triglycerides when energy is in surplus, releasing free fatty acids as a fuel during energy shortage, and secreting adipokines that are important for regulating lipid and glucose metabolism. The size of white adipose mass needs to be kept at a proper set point. Dramatic expansion of white fat mass causes obesity--now become a global epidemic disease--and increases the risk for the development of many life-threatening diseases. The absence of white adipose tissue or abnormal white adipose tissue redistribution leads to lipodystrophy, a condition often associated with metabolic disorders. Brown adipose tissue is a thermogenic organ whose mass is inversely correlated with body mass index and age. Therapeutic approaches targeting adipose tissue have been proven to be effective in improving obesity-related metabolic disorders, and promising new therapies could be developed in the near future.

© 2013 New York Academy of Sciences.

Figures

Figure 1
Figure 1
Origin and adipogenesis of white and brown adipocytes. Positive regulators involved in preadipocyte commitment and terminal differentiation of white, brite, and brown adipocytes are outlined.
Figure 2
Figure 2
BAT activities in humans Top panels, BAT activities measured by FDG-PET in a normal subject and a subject exposed to cold (from Ref. 48). Bottom panels, BAT activities in a normal subject and a pheochromocytoma patient with elevated circulating level of catecholamines (with permission from Ref. 57).
Figure 3
Figure 3
Histological images of WAT, brite cells, and BAT stained for UCP1. WAT and brite cell images are human (with permission from Ref. 45). The BAT image is mouse and from Hemmeryckx et al., with permission.
Figure 4
Figure 4
Changes of immune cell populations in adipose tissue in obesity. A Th1 response occurs in obese adipose tissue, featured with decreased populations of anti-inflammatory immune cells, including regulatory T (Treg) cells, M2 macrophages, eosinophils, and iNKT cells but increased populations of proinflammatory immune cells, such as neutrophils, M1 macrophages, mast cells, B2 cells, CD8+ T cells, and IFN-γ+ Th1 cells.
Figure 5
Figure 5
Mechanism of action for thiazolidinediones (TZDs). TZDs mainly improve adipose tissue insulin sensitivity by activating PPARγ in both adipocytes and infiltrated macrophages. Activation of PPARγ in adipose tissue promotes lipid storage in fat, decreases lipid deposit in muscle and liver through lowering circulating levels of FFA, stimulates secretion of adiponectin, and reduces secretion of proinflammatory factors, such as TNF-α, resistin, and MCP-1.

References

    1. Cristancho AG, Lazar MA. Forming functional fat: a growing understanding of adipocyte differentiation. Nat. Rev. Mol. Cell Biol. 2011;12:722–734.
    1. Poissonnet CM, Burdi AR, Garn SM. The chronology of adipose tissue appearance and distribution in the human fetus. Early Hum. Dev. 1984;10:1–11.
    1. Gesta S, Tseng YH, Kahn CR. Developmental origin of fat: tracking obesity to its source. Cell. 2007;131:242–256.
    1. Billon N, et al. The generation of adipocytes by the neural crest. Development. 2007;134:2283–2292.
    1. Ailhaud G, Grimaldi P, Negrel R. Cellular and molecular aspects of adipose tissue development. Annu. Rev. Nutr. 1992;12:207–233.
    1. Rosen ED, MacDougald OA. Adipocyte differentiation from the inside out. Nat. Rev. Mol. Cell Biol. 2006;7:885–896.
    1. Symonds ME, et al. Endocrine and nutritional regulation of fetal adipose tissue development. J. Endocrinol. 2003;179:293–299.
    1. Ibanez L, et al. Early development of adiposity and insulin resistance after catch-up weight gain in small-for-gestational-age children. J. Clin. Endocrinol. Metab. 2006;91:2153–2158.
    1. Jaquet D, et al. Insulin resistance early in adulthood in subjects born with intrauterine growth retardation. J. Clin. Endocrinol. Metab. 2000;85:1401–1406.
    1. Rigamonti A, et al. Rapid cellular turnover in adipose tissue. PLoS One. 2011;6:e17637.
    1. Spalding KL, et al. Dynamics of fat cell turnover in humans. Nature. 2008;453:783–787.
    1. Rosen ED, Spiegelman BM. Molecular regulation of adipogenesis. Annu. Rev. Cell Dev. Biol. 2000;16:145–171.
    1. Tang W, et al. White fat progenitor cells reside in the adipose vasculature. Science. 2008;322:583–586.
    1. Rodeheffer MS, Birsoy K, Friedman JM. Identification of white adipocyte progenitor cells in vivo. Cell. 2008;135:240–249.
    1. Huang H, et al. BMP signaling pathway is required for commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage. Proc. Natl. Acad. Sci. USA. 2009;106:12670–12675.
    1. Tang QQ, Lane MD. Adipogenesis: from stem cell to adipocyte. Annu. Rev. Biochem. 2012;81:715–736.
    1. Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid-activated transcription factor. Cell. 1994;79:1147–1156.
    1. Rosen ED, et al. PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro. Mol. Cell. 1999;4:611–617.
    1. Tanaka T, et al. Defective adipocyte differentiation in mice lacking the C/EBPbeta and/or C/EBPdelta gene. EMBO J. 1997;16:7432–7443.
    1. Mori T, et al. Role of Kruppel-like factor 15 (KLF15) in transcriptional regulation of adipogenesis. J. Biol. Chem. 2005;280:12867–12875.
    1. Linhart HG, et al. C/EBPalpha is required for differentiation of white, but not brown, adipose tissue. Proc. Natl. Acad. Sci. USA. 2001;98:12532–12537.
    1. Zhang JW, et al. Role of CREB in transcriptional regulation of CCAAT/enhancer-binding protein beta gene during adipogenesis. J. Biol. Chem. 2004;279:4471–4478.
    1. Chen Z, et al. Krox20 stimulates adipogenesis via C/EBPbeta-dependent and -independent mechanisms. Cell Metab. 2005;1:93–106.
    1. Birsoy K, Chen Z, Friedman J. Transcriptional regulation of adipogenesis by KLF4. Cell Metab. 2008;7:339–347.
    1. Oishi Y, et al. Kruppel-like transcription factor KLF5 is a key regulator of adipocyte differentiation. Cell Metab. 2005;1:27–39.
    1. Sue N, et al. Targeted disruption of the basic Kruppel-like factor gene (Klf3) reveals a role in adipogenesis. Mol. Cell Biol. 2008;28:3967–3978.
    1. Tontonoz P, Spiegelman BM. Fat and beyond: the diverse biology of PPARgamma. Annu. Rev. Biochem. 2008;77:289–312.
    1. Tong Q, et al. Function of GATA transcription factors in preadipocyte-adipocyte transition. Science. 2000;290:134–138.
    1. Ross SE, et al. Inhibition of adipogenesis by Wnt signaling. Science. 2000;289:950–953.
    1. James AW, et al. Sonic Hedgehog influences the balance of osteogenesis and adipogenesis in mouse adipose-derived stromal cells. Tissue Eng. Part A. 2010;16:2605–2616.
    1. Wu J, et al. The KLF2 transcription factor does not affect the formation of preadipocytes but inhibits their differentiation into adipocytes. Biochemistry. 2005;44:11098–11105.
    1. Romao JM, et al. MicroRNA regulation in mammalian adipogenesis. Exp. Biol. Med. 2011;236:997–1004.
    1. Ahima RS, Flier JS. Leptin. Annu. Rev. Physiol. 2000;62:413–437.
    1. Shetty S, Kusminski CM, Scherer PE. Adiponectin in health and disease: evaluation of adiponectin-targeted drug development strategies. Trends Pharmacol. Sci. 2009;30:234–239.
    1. Tchkonia T, et al. Fat depot origin affects adipogenesis in primary cultured and cloned human preadipocytes. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2002;282:R1286–R1296.
    1. Tchkonia T, et al. Fat depot-specific characteristics are retained in strains derived from single human preadipocytes. Diabetes. 2006;55:2571–2578.
    1. Wajchenberg BL. Subcutaneous and visceral adipose tissue: their relation to the metabolic syndrome. Endocr. Rev. 2000;21:697–738.
    1. Bahceci M, et al. The correlation between adiposity and adiponectin, tumor necrosis factor alpha, interleukin-6 and high sensitivity C-reactive protein levels. Is adipocyte size associated with inflammation in adults. J. Endocrinol. Invest. 2007;30:210–214.
    1. Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol. Rev. 2004;84:277–359.
    1. Sell H, Deshaies Y, Richard D. The brown adipocyte: update on its metabolic role. Int. J. Biochem. Cell Biol. 2004;36:2098–2104.
    1. Farmer SR. Molecular determinants of brown adipocyte formation and function. Genes Dev. 2008;22:1269–1275.
    1. Bargmann W, von Hehn G, Lindner E. On the cells of the brown fatty tissue and their innervation. Z. Zellforsch Mikrosk Anat. 1968;85:601–613.
    1. Merklin RJ. Growth and distribution of human fetal brown fat. Anat. Rec. 1974;178:637–645.
    1. Carter BW, Schucany WG. Brown adipose tissue in a newborn. Proc. Bayl. Univ. Med. Cent. 2008;21:328–330.
    1. Virtanen KA, et al. Functional brown adipose tissue in healthy adults. N. Engl. J. Med. 2009;360:1518–1525.
    1. Cypess AM, et al. Identification and importance of brown adipose tissue in adult humans. N. Engl. J. Med. 2009;360:1509–1517.
    1. van Marken Lichtenbelt WD, et al. Cold-activated brown adipose tissue in healthy men. N. Engl. J. Med. 2009;360:1500–1508.
    1. Cypess AM, et al. Cold but not sympathomimetics activates human brown adipose tissue in vivo. Proc. Natl. Acad. Sci. USA. 2012;109:10001–10005.
    1. Enerback S. Brown adipose tissue in humans. Int. J. Obes. 2010;34(Suppl 1):S43–S46.
    1. Nedergaard J, Bengtsson T, Cannon B. Unexpected evidence for active brown adipose tissue in adult humans. Am. J. Physiol. Endocrinol. Metab. 2007;293:E444–E452.
    1. Heaton JM. The distribution of brown adipose tissue in the human. J. Anat. 1972;112:35–39.
    1. Nedergaard J, Cannon B. The changed metabolic world with human brown adipose tissue: therapeutic visions. Cell Metab. 2010;11:268–272.
    1. Ouellet V, et al. Brown adipose tissue oxidative metabolism contributes to energy expenditure during acute cold exposure in humans. J. Clin. Invest. 2012;122:545–552.
    1. Orava J, et al. Different metabolic responses of human brown adipose tissue to activation by cold and insulin. Cell Metab. 2011;14:272–279.
    1. Ricquier D, Nechad M, Mory G. Ultrastructural and biochemical characterization of human brown adipose tissue in pheochromocytoma. J. Clin. Endocrinol. Metab. 1982;54:803–807.
    1. Fukuchi K, et al. Radionuclide imaging metabolic activity of brown adipose tissue in a patient with pheochromocytoma. Exp. Clin. Endocrinol. Diabetes. 2004;112:601–603.
    1. Wang Q, et al. Brown adipose tissue in humans is activated by elevated plasma catecholamines levels and is inversely related to central obesity. PLoS One. 2011;6:e21006.
    1. Huttunen P, Hirvonen J, Kinnula V. The occurrence of brown adipose tissue in outdoor workers. Eur. J. Appl. Physiol. Occup. Physiol. 1981;46:339–345.
    1. Kuji I, et al. Brown adipose tissue demonstrating intense FDG uptake in a patient with mediastinal pheochromocytoma. Ann. Nucl. Med. 2008;22:231–235.
    1. Wu J, et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell. 2012;150:366–376.
    1. Tseng YH, et al. New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature. 2008;454:1000–1004.
    1. Frontini A, Cinti S. Distribution and development of brown adipocytes in the murine and human adipose organ. Cell Metab. 2010;11:253–256.
    1. Seale P, et al. Transcriptional control of brown fat determination by PRDM16. Cell Metab. 2007;6:38–54.
    1. Uldry M, et al. Complementary action of the PGC-1 coactivators in mitochondrial biogenesis and brown fat differentiation. Cell Metab. 2006;3:333–341.
    1. Leone TC, et al. PGC-1alpha deficiency causes multi-system energy metabolic derangements: muscle dysfunction, abnormal weight control and hepatic steatosis. PLoS Biol. 2005;3:e101.
    1. Lin J, et al. Defects in adaptive energy metabolism with CNS-linked hyperactivity in PGC-1alpha null mice. Cell. 2004;119:121–135.
    1. Bostrom P, et al. A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature. 2012;481:463–468.
    1. Leonardsson G, et al. Nuclear receptor corepressor RIP140 regulates fat accumulation. Proc. Natl. Acad. Sci. USA. 2004;101:8437–8442.
    1. Kang S, et al. Effects of Wnt signaling on brown adipocyte differentiation and metabolism mediated by PGC-1alpha. Mol. Cell. Biol. 2005;25:1272–1282.
    1. Scime A, et al. Rb and p107 regulate preadipocyte differentiation into white versus brown fat through repression of PGC-1alpha. Cell Metab. 2005;2:283–295.
    1. Montague CT, et al. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature. 1997;387:903–908.
    1. Gibson WT, et al. Congenital leptin deficiency due to homozygosity for the Delta133G mutation: report of another case and evaluation of response to four years of leptin therapy. J. Clin. Endocrinol. Metab. 2004;89:4821–4826.
    1. Strobel A, et al. A leptin missense mutation associated with hypogonadism and morbid obesity. Nat. Genet. 1998;18:213–215.
    1. Farooqi IS, et al. Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N. Engl. J. Med. 1999;341:879–884.
    1. Farooqi IS, et al. Clinical and molecular genetic spectrum of congenital deficiency of the leptin receptor. N. Engl. J. Med. 2007;356:237–247.
    1. Clement K, et al. A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature. 1998;392:398–401.
    1. Farooqi IS, et al. Dominant and recessive inheritance of morbid obesity associated with melanocortin 4 receptor deficiency. J. Clin. Invest. 2000;106:271–279.
    1. Lubrano-Berthelier C, et al. Melanocortin 4 receptor mutations in a large cohort of severely obese adults: prevalence, functional classification, genotype-phenotype relationship, and lack of association with binge eating. J. Clin. Endocrinol. Metab. 2006;91:1811–1818.
    1. Krude H, et al. Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nat. Genet. 1998;19:155–157.
    1. Jackson RS, et al. Obesity and impaired prohormone processing associated with mutations in the human prohormone convertase 1 gene. Nat. Genet. 1997;16:303–306.
    1. Farooqi IS, et al. Hyperphagia and early-onset obesity due to a novel homozygous missense mutation in prohormone convertase 1/3. J. Clin. Endocrinol. Metab. 2007;92:3369–3373.
    1. Yeo GS, et al. A de novo mutation affecting human TrkB associated with severe obesity and developmental delay. Nat. Neurosci. 2004;7:1187–1189.
    1. Gray J, et al. Hyperphagia, severe obesity, impaired cognitive function, and hyperactivity associated with functional loss of one copy of the brain-derived neurotrophic factor (BDNF) gene. Diabetes. 2006;55:3366–3371.
    1. Han JC, et al. Brain-derived neurotrophic factor and obesity in the WAGR syndrome. N. Engl. J. Med. 2008;359:918–927.
    1. Hung CC, et al. Studies of the SIM1 gene in relation to human obesity and obesity-related traits. Int. J. Obes. 2007;31:429–434.
    1. Tobin JL, Beales PL. Bardet-Biedl syndrome: beyond the cilium. Pediatr. Nephrol. 2007;22:926–936.
    1. Ansley SJ, et al. Basal body dysfunction is a likely cause of pleiotropic Bardet-Biedl syndrome. Nature. 2003;425:628–633.
    1. Sahoo T, et al. Prader-Willi phenotype caused by paternal deficiency for the HBII-85 C/D box small nucleolar RNA cluster. Nat. Genet. 2008;40:719–721.
    1. de Smith AJ, et al. A deletion of the HBII-85 class of small nucleolar RNAs (snoRNAs) is associated with hyperphagia, obesity and hypogonadism. Hum. Mol. Genet. 2009;18:3257–3265.
    1. Bochukova EG, et al. Large, rare chromosomal deletions associated with severe early-onset obesity. Nature. 2010;463:666–670.
    1. Padoan A, et al. Differences in fat and lean mass proportions in normal and growth-restricted fetuses. Am. J. Obstet. Gynecol. 2004;191:1459–1464.
    1. Mazziotti G, Gazzaruso C, Giustina A. Diabetes in Cushing syndrome: basic and clinical aspects. Trends Endocrinol. Metab. 2011;22:499–506.
    1. Chanson P, Salenave S. Metabolic syndrome in Cushing's syndrome. Neuroendocrinology. 2010;92(Suppl 1):96–101.
    1. Kola B, et al. Changes in adenosine 5’-monophosphate-activated protein kinase as a mechanism of visceral obesity in Cushing's syndrome. J. Clin. Endocrinol. Metab. 2008;93:4969–4973.
    1. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444:860–867.
    1. Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance. J. Clin. Invest. 2006;116:1793–1801.
    1. Hotamisligil GS, Spiegelman BM. Tumor necrosis factor alpha: a key component of the obesity-diabetes link. Diabetes. 1994;43:1271–1278.
    1. Amrani A, et al. Interleukin-1 effect on glycemia in the non-obese diabetic mouse at the pre-diabetic stage. J. Endocrinol. 1996;148:139–148.
    1. Sartipy P, Loskutoff DJ. Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc. Natl. Acad. Sci. USA. 2003;100:7265–7270.
    1. Rotter V, Nagaev I, Smith U. Interleukin-6 (IL-6) induces insulin resistance in 3T3-L1 adipocytes and is, like IL-8 and tumor necrosis factor-alpha, overexpressed in human fat cells from insulin-resistant subjects. J. Biol. Chem. 2003;278:45777–45784.
    1. Schipper HS, et al. Adipose tissue-resident immune cells: key players in immunometabolism. Trends Endocrinol. Metab. 2012;23:407–415.
    1. Xu H, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Invest. 2003;112:1821–1830.
    1. Jiao P, et al. Obesity-related upregulation of monocyte chemotactic factors in adipocytes: involvement of nuclear factor-kappaB and c-Jun NH2-terminal kinase pathways. Diabetes. 2009;58:104–115.
    1. Nomiyama T, et al. Osteopontin mediates obesity-induced adipose tissue macrophage infiltration and insulin resistance in mice. J. Clin. Invest. 2007;117:2877–2888.
    1. Weisberg SP, et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J. Clin. Invest. 2006;116:115–124.
    1. Nara N, et al. Disruption of CXC motif chemokine ligand-14 in mice ameliorates obesity-induced insulin resistance. J. Biol. Chem. 2007;282:30794–30803.
    1. Feng B, et al. Clodronate liposomes improve metabolic profile and reduce visceral adipose macrophage content in diet-induced obese mice. PLoS One. 2011;6:e24358.
    1. Patsouris D, et al. Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab. 2008;8:301–309.
    1. Clement K, et al. Weight loss regulates inflammation-related genes in white adipose tissue of obese subjects. FASEB J. 2004;18:1657–1669.
    1. Cancello R, et al. Reduction of macrophage infiltration and chemoattractant gene expression changes in white adipose tissue of morbidly obese subjects after surgery-induced weight loss. Diabetes. 2005;54:2277–2286.
    1. Cote M, et al. Adiponectinemia in visceral obesity: impact on glucose tolerance and plasma lipoprotein and lipid levels in men. J. Clin. Endocrinol. Metab. 2005;90:1434–1439.
    1. Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling and obesity. J. Clin. Invest. 2011;121:2094–2101.
    1. Pasarica M, et al. Reduced adipose tissue oxygenation in human obesity: evidence for rarefaction, macrophage chemotaxis, and inflammation without an angiogenic response. Diabetes. 2009;58:718–725.
    1. Ye J. Emerging role of adipose tissue hypoxia in obesity and insulin resistance. Int. J. Obes. 2009;33:54–66.
    1. Cinti S, et al. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J. Lipid Res. 2005;46:2347–2355.
    1. Divoux A, et al. Fibrosis in human adipose tissue: composition, distribution, and link with lipid metabolism and fat mass loss. Diabetes. 2010;59:2817–2825.
    1. Khan T, et al. Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol. Cell Biol. 2009;29:1575–1591.
    1. Shaw AJ, McLean KA, Evans BA. Disorders of fat distribution in HIV infection. Int. J. STD AIDS. 1998;9:595–599.
    1. Giralt M, Domingo P, Villarroya F. Adipose tissue biology and HIV-infection. Best Pract. Res. Clin. Endocrinol. Metab. 2011;25:487–499.
    1. Giralt M, et al. HIV-1 infection alters gene expression in adipose tissue, which contributes to HIV- 1/HAART-associated lipodystrophy. Antivir. Ther. 2006;11:729–740.
    1. Bastard JP, et al. Association between altered expression of adipogenic factor SREBP1 in lipoatrophic adipose tissue from HIV-1-infected patients and abnormal adipocyte differentiation and insulin resistance. Lancet. 2002;359:1026–1031.
    1. Jan V, et al. Altered fat differentiation and adipocytokine expression are inter-related and linked to morphological changes and insulin resistance in HIV-1-infected lipodystrophic patients. Antivir. Ther. 2004;9:555–564.
    1. Grinspoon S, Carr A. Cardiovascular risk and body-fat abnormalities in HIV-infected adults. N. Engl. J. Med. 2005;352:48–62.
    1. Carr A, Cooper DA. Adverse effects of antiretroviral therapy. Lancet. 2000;356:1423–1430.
    1. Misra A, Garg A. Clinical features and metabolic derangements in acquired generalized lipodystrophy: case reports and review of the literature. Medicine. 2003;82:129–146.
    1. Garg A. Clinical review: Lipodystrophies: genetic and acquired body fat disorders. J. Clin. Endocrinol. Metab. 2011;96:3313–3325.
    1. Berardinelli W. An undiagnosed endocrinometabolic syndrome: report of 2 cases. J. Clin. Endocrinol. Metab. 1954;14:193–204.
    1. Seip M. Lipodystrophy and gigantism with associated endocrine manifestations. A new diencephalic syndrome. Acta Paediatr. 1959;48:555–574.
    1. Agarwal AK, et al. AGPAT2 is mutated in congenital generalized lipodystrophy linked to chromosome 9q34. Nat. Genet. 2002;31:21–23.
    1. Magre J, et al. Identification of the gene altered in Berardinelli-Seip congenital lipodystrophy on chromosome 11q13. Nat. Genet. 2001;28:365–370.
    1. Kim CA, et al. Association of a homozygous nonsense caveolin-1 mutation with Berardinelli-Seip congenital lipodystrophy. J. Clin. Endocrinol. Metab. 2008;93:1129–1134.
    1. Hayashi YK, et al. Human PTRF mutations cause secondary deficiency of caveolins resulting in muscular dystrophy with generalized lipodystrophy. J. Clin. Invest. 2009;119:2623–2633.
    1. Takeuchi K, Reue K. Biochemistry, physiology, and genetics of GPAT, AGPAT, and lipin enzymes in triglyceride synthesis. Am. J. Physiol. Endocrinol. Metab. 2009;296:E1195–E1209.
    1. Szymanski KM, et al. The lipodystrophy protein seipin is found at endoplasmic reticulum lipid droplet junctions and is important for droplet morphology. Proc. Natl. Acad. Sci. USA. 2007;104:20890–20895.
    1. Payne VA, et al. The human lipodystrophy gene BSCL2/seipin may be essential for normal adipocyte differentiation. Diabetes. 2008;57:2055–2060.
    1. Shastry S, et al. Congenital generalized lipodystrophy, type 4 (CGL4) associated with myopathy due to novel PTRF mutations. Am. J. Med. Genet A. 2010;152A:2245–2253.
    1. Simha V, et al. Novel subtype of congenital generalized lipodystrophy associated with muscular weakness and cervical spine instability. Am. J. Med. Genet A. 2008;146A:2318–2326.
    1. Shackleton S, et al. LMNA, encoding lamin A/C, is mutated in partial lipodystrophy. Nat. Genet. 2000;24:153–156.
    1. Semple RK, Chatterjee VK, O'Rahilly S. PPAR gamma and human metabolic disease. J. Clin. Invest. 2006;116:581–589.
    1. George S, et al. A family with severe insulin resistance and diabetes due to a mutation in AKT2. Science. 2004;304:1325–1328.
    1. Rubio-Cabezas O, et al. Partial lipodystrophy and insulin resistant diabetes in a patient with a homozygous nonsense mutation in CIDEC. EMBO Mol. Med. 2009;1:280–287.
    1. Cao H, Hegele RA. Nuclear lamin A/C R482Q mutation in canadian kindreds with Dunnigan-type familial partial lipodystrophy. Hum. Mol. Genet. 2000;9:109–112.
    1. Okumura T. Role of lipid droplet proteins in liver steatosis. J. Physiol. Biochem. 2011;67:629–636.
    1. Xu L, Zhou L, Li P. CIDE proteins and lipid metabolism. Arterioscler Thromb. Vasc. Biol. 2012;32:1094–1098.
    1. Salam GA. Lipoma excision. Am. Fam. Physician. 2002;65:901–904.
    1. Buisson P, et al. Cutaneous lipoma in children: 5 cases with Bannayan-Riley-Ruvalcaba syndrome. J. Pediatr. Surg. 2006;41:1601–1603.
    1. Arlotta P, et al. Transgenic mice expressing a truncated form of the high mobility group I-C protein develop adiposity and an abnormally high prevalence of lipomas. J. Biol. Chem. 2000;275:14394–1400.
    1. Dei Tos AP. Liposarcoma: new entities and evolving concepts. Ann. Diagn. Pathol. 2000;4:252–266.
    1. Brines OA, Johnson MH. Hibernoma, a special fatty tumor; report of a case. Am. J. Pathol. 1949;25:467–479.
    1. Furlong MA, Fanburg-Smith JC, Miettinen M. The morphologic spectrum of hibernoma: a clinicopathologic study of 170 cases. Am. J. Surg. Pathol. 2001;25:809–814.
    1. Honoki K, et al. Hibernoma of the axillary region: a rare benign adipocytic tumor. Rare Tumors. 2010;2:e7.
    1. Lehmann JM, et al. An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma (PPAR gamma) J. Biol. Chem. 1995;270:12953–12956.
    1. He W, et al. Adipose-specific peroxisome proliferator-activated receptor gamma knockout causes insulin resistance in fat and liver but not in muscle. Proc. Natl. Acad. Sci. USA. 2003;100:15712–15717.
    1. Barroso I, et al. Dominant negative mutations in human PPARgamma associated with severe insulin resistance, diabetes mellitus and hypertension. Nature. 1999;402:880–883.
    1. Raskin P, et al. Rosiglitazone short-term monotherapy lowers fasting and post-prandial glucose in patients with type II diabetes. Diabetologia. 2000;43:278–284.
    1. Okuno A, et al. Troglitazone increases the number of small adipocytes without the change of white adipose tissue mass in obese Zucker rats. J. Clin. Invest. 1998;101:1354–1361.
    1. Trujillo ME, Scherer PE. Adipose tissue-derived factors: impact on health and disease. Endocr. Rev. 2006;27:762–778.
    1. Combs TP, et al. A transgenic mouse with a deletion in the collagenous domain of adiponectin displays elevated circulating adiponectin and improved insulin sensitivity. Endocrinology. 2004;145:367–383.
    1. Hevener AL, et al. Macrophage PPAR gamma is required for normal skeletal muscle and hepatic insulin sensitivity and full antidiabetic effects of thiazolidinediones. J. Clin. Invest. 2007;117:1658–1669.
    1. Jiang C, Ting AT, Seed B. PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature. 1998;391:82–86.
    1. Marx N, Libby P, Plutzky J. Peroxisome proliferator-activated receptors (PPARs) and their role in the vessel wall: possible mediators of cardiovascular risk. J. Cardiovasc. Risk. 2001;8:203–210.
    1. Ghanim H, et al. Suppression of nuclear factor-kappaB and stimulation of inhibitor kappaB by troglitazone: evidence for an anti-inflammatory effect and a potential antiatherosclerotic effect in the obese. J. Clin. Endocrinol. Metab. 2001;86:1306–1312.
    1. Aljada A, et al. Nuclear factor-kappaB suppressive and inhibitor-kappaB stimulatory effects of troglitazone in obese patients with type 2 diabetes: evidence of an antiinflammatory action. J. Clin. Endocrinol. Metab. 2001;86:3250–3256.
    1. Mohanty P, et al. Evidence for a potent antiinflammatory effect of rosiglitazone. J. Clin. Endocrinol. Metab. 2004;89:2728–2735.
    1. Odegaard JI, et al. Macrophage-specific PPAR-gamma controls alternative activation and improves insulin resistance. Nature. 2007;447:1116–1120.
    1. Feuerer M, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat. Med. 2009;15:930–939.
    1. Cipolletta D, et al. PPAR-gamma is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature. 2012;486:549–553.
    1. Choi JH, et al. Antidiabetic actions of a non-agonist PPARgamma ligand blocking Cdk5-mediated phosphorylation. Nature. 2011;477:477–481.
    1. Pirat C, et al. Targeting peroxisome proliferator-activated receptors (PPARs): development of modulators. J. Med. Chem. 2012;55:4027–4061.
    1. Dunn FL, et al. Selective modulation of PPARgamma activity can lower plasma glucose without typical thiazolidinedione side-effects in patients with Type 2 diabetes. J. Diabetes Complications. 2011;25:151–158.
    1. Penumetcha M, Santanam N. Nutraceuticals as Ligands of PPARgamma. PPAR Res. 2012;2012:858352.
    1. Fleischman A, et al. Salsalate improves glycemia and inflammatory parameters in obese young adults. Diabetes Care. 2008;31:289–294.
    1. Goldfine AB, et al. The effects of salsalate on glycemic control in patients with type 2 diabetes: a randomized trial. Ann. Intern. Med. 2010;152:346–357.
    1. Romeo GR, Lee J, Shoelson SE. Metabolic syndrome, insulin resistance, and roles of inflammation – mechanisms and therapeutic targets. Arterioscler Thromb. Vasc. Biol. 2012;32:1771–1776.
    1. Berchtold LA, et al. IL-1 receptor antagonism and muscle gene expression in patients with type 2 diabetes. Eur. Cytokine Netw. 2009;20:81–87.
    1. Ofei F, et al. Effects of an engineered human anti-TNF-alpha antibody (CDP571) on insulin sensitivity and glycemic control in patients with NIDDM. Diabetes. 1996;45:881–885.
    1. Paquot N, et al. No increased insulin sensitivity after a single intravenous administration of a recombinant human tumor necrosis factor receptor: Fc fusion protein in obese insulin-resistant patients. J. Clin. Endocrinol. Metab. 2000;85:1316–1319.
    1. Rupnick MA, et al. Adipose tissue mass can be regulated through the vasculature. Proc. Natl. Acad. Sci. USA. 2002;99:10730–10735.
    1. Oike Y, Tabata M. Angiopoietin-like proteins–potential therapeutic targets for metabolic syndrome and cardiovascular disease. Circ. J. 2009;73:2192–2197.
    1. Kunduzova O, et al. Apelin/APJ signaling system: a potential link between adipose tissue and endothelial angiogenic processes. FASEB J. 2008;22:4146–4153.
    1. Kolonin MG, et al. Reversal of obesity by targeted ablation of adipose tissue. Nat. Med. 2004;10:625–632.
    1. Kim DH, Woods SC, Seeley RJ. Peptide designed to elicit apoptosis in adipose tissue endothelium reduces food intake and body weight. Diabetes. 2010;59:907–915.
    1. Cummings DE. Metabolic surgery for type 2 diabetes. Nat. Med. 2012;18:656–658.
    1. Mohammed BS, et al. Long-term effects of large-volume liposuction on metabolic risk factors for coronary heart disease. Obesity. 2008;16:2648–2651.
    1. Klein S. Is visceral fat responsible for the metabolic abnormalities associated with obesity?: implications of omentectomy. Diabetes Care. 2010;33:1693–1694.
    1. Astrup A, et al. Fat metabolism in the predisposition to obesity. Ann. N.Y. Acad. Sci. 1997;827:417–430.
    1. Bartlett J, Brunner M, Gough K. Deliberate poisoning with dinitrophenol (DNP): an unlicensed weight loss pill. Emerg. Med. J. 2010;27:159–160.
    1. Grundlingh J, et al. 2,4-dinitrophenol (DNP): a weight loss agent with significant acute toxicity and risk of death. J. Med. Toxicol. 2011;7:205–212.
    1. Dulloo AG. The search for compounds that stimulate thermogenesis in obesity management: from pharmaceuticals to functional food ingredients. Obes. Rev. 2011;12:866–883.
    1. Snitker S, et al. The sympathetic nervous system and obesity: role in aetiology and treatment. Obes. Rev. 2000;1:5–15.
    1. Acheson KJ, et al. Caffeine and coffee: their influence on metabolic rate and substrate utilization in normal weight and obese individuals. Am. J. Clin. Nutr. 1980;33:989–997.
    1. Astrup A, et al. Enhanced thermogenic responsiveness during chronic ephedrine treatment in man. Am. J. Clin. Nutr. 1985;42:83–94.
    1. Arch JR, et al. Atypical beta-adrenoceptor on brown adipocytes as target for anti-obesity drugs. Nature. 1984;309:163–165.
    1. Arch JR. The discovery of drugs for obesity, the metabolic effects of leptin and variable receptor pharmacology: perspectives from beta3-adrenoceptor agonists. Naunyn. Schmiedebergs Arch. Pharmacol. 2008;378:225–240.
    1. Rothwell NJ, Stock MJ. Diet-induced thermogenesis. Adv. Nutr. Res. 1983;5:201–220.
    1. Tran TT, Kahn CR. Transplantation of adipose tissue and stem cells: role in metabolism and disease. Nat. Rev. Endocrinol. 2010;6:195–213.
    1. Ahfeldt T, et al. Programming human pluripotent stem cells into white and brown adipocytes. Nat. Cell Biol. 2012;14:209–219.
    1. Gunawardana SC, Piston DW. Reversal of type 1 diabetes in mice by brown adipose tissue transplant. Diabetes. 2012;61:674–682.
    1. Timmons JA, et al. Is irisin a human exercise gene. Nature. 2012;488:E9–E10. discussion E10–1.
    1. Huh JY, et al. FNDC5 and irisin in humans: I. Predictors of circulating concentrations in serum and plasma and II. mRNA expression and circulating concentrations in response to weight loss and exercise. Metabolism. 2012;61:1725–1736.
    1. Hemmeryckx B, Hoylaerts MF, Lijnen HR. Effect of premature aging on murine adipose tissue. Exp. Gerontol. 2012;47:256–262.

Source: PubMed

3
Iratkozz fel