The Multifaceted Roles of Adipose Tissue-Therapeutic Targets for Diabetes and Beyond: The 2015 Banting Lecture

Philipp E Scherer, Philipp E Scherer

Abstract

The Banting Medal for Scientific Achievement is the highest scientific award of the American Diabetes Association (ADA). Given in memory of Sir Frederick Banting, one of the key investigators in the discovery of insulin, the Banting Medal is awarded annually for scientific excellence, recognizing significant long-term contributions to the understanding, treatment, or prevention of diabetes. Philipp E. Scherer, PhD, of the Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX, received the prestigious award at the ADA's 75th Scientific Sessions, 5-9 June 2015, in Boston, MA. He presented the Banting Lecture, "The Multifaceted Roles of Adipose Tissue-Therapeutic Targets for Diabetes and Beyond," on Sunday, 7 June 2015.A number of different cell types contribute to the cellular architecture of adipose tissue. Although the adipocyte is functionally making important contributions to systemic metabolic homeostatis, several additional cell types contribute a supportive role to bestow maximal flexibility on the tissue with respect to many biosynthetic and catabolic processes, depending on the metabolic state. These cells include vascular endothelial cells, a host of immune cells, and adipocyte precursor cells and fibroblasts. Combined, these cell types give rise to a tissue with remarkable flexibility with respect to expansion and contraction, while optimizing the ability of the tissue to act as an endocrine organ through the release of many protein factors, critically influencing systemic lipid homeostasis and biochemically contributing many metabolites. Using an example from each of these categories-adiponectin as a key adipokine, sphingolipids as critical mediators of insulin sensitivity, and uridine as an important metabolite contributed by the adipocyte to the systemic pool-I will discuss the emerging genesis of the adipocyte over the past 20 years from metabolic bystander to key driver of metabolic flexibility.

© 2016 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered.

Figures

Figure 1
Figure 1
The role of inflammation in the adipocyte. Genetic approaches to suppress local inflammatory responses in the adipocyte include a dominant-negative (dn) TNFα ligand that unproductively engages the TNF receptors, a potent adenoviral anti-inflammatory molecule called RID, and a traditional “super-repressor” of inflammation in the form of a mutant IκBα.
Figure 2
Figure 2
The consequences of a suppressed inflammatory response of the adipocyte. The process of adipose tissue expansion requires an inflammatory response as part of the healthy expansion process. By inhibiting these inflammatory events, adipogenesis is impaired, thereby reducing the number of fat cells available to accommodate any excess lipids, leading to ectopic lipid deposition in the liver. This also leads to a compromised intestinal barrier and the enhanced release of endotoxin into the system. Therefore, despite an early anti-inflammatory action at the level of the adipocyte, chronic systemic inflammation and severe systemic insulin resistance ensue in this setting.
Figure 3
Figure 3
The different types of adipocytes. Distinct precursor cells give rise to the different types of adipocytes, the classic white and brown adipocytes and the beige adipocytes that may transition to a “dormant” cell resembling a white adipocyte when their presence is not required. All these events require extensive remodeling of the extracellular matrix microenvironment of these cells.
Figure 4
Figure 4
The adipocyte as a professional secretory cell. Many distinct categories of secretory proteins have been reported to be released from adipocytes. Acrp30, adipocyte complement-related protein of 30 kDa; ASP, acylation-stimulating protein; ECM, extracellular matrix; IL-6, interleukin 6; MT1-MMP, membrane type 1 matrix metalloproteinase; PTX-3, pentraxin 3; RBP4, retinol-binding protein 4.

References

    1. Kasanicki MA, Pilch PF. Regulation of glucose-transporter function. Diabetes Care 1990;13:219–227
    1. Rutkowski JM, Stern JH, Scherer PE. The cell biology of fat expansion. J Cell Biol 2015;208:501–512
    1. Unger RH, Scherer PE. Gluttony, sloth and the metabolic syndrome: a roadmap to lipotoxicity. Trends Endocrinol Metab 2010;21:345–352
    1. Lowell BB, Spiegelman BM. Towards a molecular understanding of adaptive thermogenesis. Nature 2000;404:652–660
    1. Storlien L, Oakes ND, Kelley DE. Metabolic flexibility. Proc Nutr Soc 2004;63:363–368
    1. Samocha-Bonet D, Dixit VD, Kahn CR, et al. . Metabolically healthy and unhealthy obese--the 2013 Stock Conference report. Obes Rev 2014;15:697–708
    1. Patni N, Garg A. Congenital generalized lipodystrophies--new insights into metabolic dysfunction. Nat Rev Endocrinol 2015;11:522–534
    1. Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling and obesity. J Clin Invest 2011;121:2094–2101
    1. Rajala MW, Scherer PE. Minireview: The adipocyte--at the crossroads of energy homeostasis, inflammation, and atherosclerosis. Endocrinology 2003;144:3765–3773
    1. Sun K, Tordjman J, Clément K, Scherer PE. Fibrosis and adipose tissue dysfunction. Cell Metab 2013;18:470–477
    1. Pasarica M, Gowronska-Kozak B, Burk D, et al. . Adipose tissue collagen VI in obesity. J Clin Endocrinol Metab 2009;94:5155–5162
    1. Divoux A, Clément K. Architecture and the extracellular matrix: the still unappreciated components of the adipose tissue. Obes Rev 2011;12:e494–e503
    1. Khan T, Muise ES, Iyengar P, et al. . Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol Cell Biol 2009;29:1575–1591
    1. Lackey DE, Burk DH, Ali MR, et al. . Contributions of adipose tissue architectural and tensile properties toward defining healthy and unhealthy obesity. Am J Physiol Endocrinol Metab 2014;306:E233–E246
    1. Sun K, Wernstedt Asterholm I, Kusminski CM, et al. . Dichotomous effects of VEGF-A on adipose tissue dysfunction. Proc Natl Acad Sci U S A 2012;109:5874–5879
    1. Berg AH, Scherer PE. Adipose tissue, inflammation, and cardiovascular disease. Circ Res 2005;96:939–949
    1. Wernstedt Asterholm I, Tao C, Morley TS, et al. . Adipocyte inflammation is essential for healthy adipose tissue expansion and remodeling. Cell Metab 2014;20:103–118
    1. Nedergaard J, Cannon B. The changed metabolic world with human brown adipose tissue: therapeutic visions. Cell Metab 2010;11:268–272
    1. Ye R, Scherer PE. Adiponectin, driver or passenger on the road to insulin sensitivity? Mol Metab 2013;2:133–141
    1. Flier JS, Cook KS, Usher P, Spiegelman BM. Severely impaired adipsin expression in genetic and acquired obesity. Science 1987;237:405–408
    1. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 1993;259:87–91
    1. Halberg N, Wernstedt-Asterholm I, Scherer PE. The adipocyte as an endocrine cell. Endocrinol Metab Clin North Am 2008;37:753–768, x–xi
    1. Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem 1995;270:26746–26749
    1. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature 1994;372:425–432
    1. Steppan CM, Bailey ST, Bhat S, et al. . The hormone resistin links obesity to diabetes. Nature 2001;409:307–312
    1. Rajala MW, Lin Y, Ranalletta M, et al. . Cell type-specific expression and coregulation of murine resistin and resistin-like molecule-alpha in adipose tissue. Mol Endocrinol 2002;16:1920–1930
    1. Kharitonenkov A, Shiyanova TL, Koester A, et al. . FGF-21 as a novel metabolic regulator. J Clin Invest 2005;115:1627–1635
    1. Park J, Scherer PE. Adipocyte-derived endotrophin promotes malignant tumor progression. J Clin Invest 2012;122:4243–4256
    1. Arita Y, Kihara S, Ouchi N, et al. . Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Commun 1999;257:79–83
    1. Turer AT, Khera A, Ayers CR, et al. . Adipose tissue mass and location affect circulating adiponectin levels. Diabetologia 2011;54:2515–2524
    1. Ahl S, Guenther M, Zhao S, et al. . Adiponectin levels differentiate metabolically healthy vs unhealthy among obese and nonobese white individuals. J Clin Endocrinol Metab 2015;100:4172–4180
    1. Aguilar-Salinas CA, García EG, Robles L, et al. . High adiponectin concentrations are associated with the metabolically healthy obese phenotype. J Clin Endocrinol Metab 2008;93:4075–4079
    1. Kim JY, van de Wall E, Laplante M, et al. . Obesity-associated improvements in metabolic profile through expansion of adipose tissue. J Clin Invest 2007;117:2621–2637
    1. Kusminski CM, Holland WL, Sun K, et al. . MitoNEET-driven alterations in adipocyte mitochondrial activity reveal a crucial adaptive process that preserves insulin sensitivity in obesity. Nat Med 2012;18:1539–1549
    1. Pajvani UB, Hawkins M, Combs TP, et al. . Complex distribution, not absolute amount of adiponectin, correlates with thiazolidinedione-mediated improvement in insulin sensitivity. J Biol Chem 2004;279:12152–12162
    1. Nawrocki AR, Rajala MW, Tomas E, et al. . Mice lacking adiponectin show decreased hepatic insulin sensitivity and reduced responsiveness to peroxisome proliferator-activated receptor gamma agonists. J Biol Chem 2006;281:2654–2660
    1. Asterholm IW, Scherer PE. Enhanced metabolic flexibility associated with elevated adiponectin levels. Am J Pathol 2010;176:1364–1376
    1. Combs TP, Pajvani UB, Berg AH, 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. Pajvani UB, Trujillo ME, Combs TP, et al. . Fat apoptosis through targeted activation of caspase 8: a new mouse model of inducible and reversible lipoatrophy. Nat Med 2005;11:797–803
    1. Holland WL, Miller RA, Wang ZV, et al. . Receptor-mediated activation of ceramidase activity initiates the pleiotropic actions of adiponectin. Nat Med 2011;17:55–63
    1. Wang ZV, Mu J, Schraw TD, et al. . PANIC-ATTAC: a mouse model for inducible and reversible beta-cell ablation. Diabetes 2008;57:2137–2148
    1. Rutkowski JM, Wang ZV, Park AS, et al. . Adiponectin promotes functional recovery after podocyte ablation. J Am Soc Nephrol 2013;24:268–282
    1. Ye R, Holland WL, Gordillo R, et al. . Adiponectin is essential for lipid homeostasis and survival under insulin deficiency and promotes β-cell regeneration. eLife 2014;3:3
    1. Landskroner-Eiger S, Qian B, Muise ES, et al. . Proangiogenic contribution of adiponectin toward mammary tumor growth in vivo. Clin Cancer Res 2009;15:3265–3276
    1. Berg AH, Combs TP, Du X, Brownlee M, Scherer PE. The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nat Med 2001;7:947–953
    1. Kondo H, Shimomura I, Matsukawa Y, et al. . Association of adiponectin mutation with type 2 diabetes: a candidate gene for the insulin resistance syndrome. Diabetes 2002;51:2325–2328
    1. Tonelli J, Li W, Kishore P, et al. . Mechanisms of early insulin-sensitizing effects of thiazolidinediones in type 2 diabetes. Diabetes 2004;53:1621–1629
    1. Pischon T, Girman CJ, Hotamisligil GS, Rifai N, Hu FB, Rimm EB. Plasma adiponectin levels and risk of myocardial infarction in men. JAMA 2004;291:1730–1737
    1. Wang QA, Scherer PE. The AdipoChaser mouse: a model tracking adipogenesis in vivo. Adipocyte 2014;3:146–150
    1. Wang ZV, Deng Y, Wang QA, Sun K, Scherer PE. Identification and characterization of a promoter cassette conferring adipocyte-specific gene expression. Endocrinology 2010;151:2933–2939
    1. Okada-Iwabu M, Yamauchi T, Iwabu M, et al. . A small-molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature 2013;503:493–499
    1. Tanabe H, Fujii Y, Okada-Iwabu M, et al. . Crystal structures of the human adiponectin receptors. Nature 2015;520:312–316
    1. Kadowaki T, Yamauchi T, Okada-Iwabu M, Iwabu M. Adiponectin and its receptors: implications for obesity-associated diseases and longevity. Lancet Diabetes Endocrinol 2014;2:8–9
    1. Matsuzawa Y. Adiponectin: a key player in obesity related disorders. Curr Pharm Des 2010;16:1896–1901
    1. Holland WL, Summers SA. Sphingolipids, insulin resistance, and metabolic disease: new insights from in vivo manipulation of sphingolipid metabolism. Endocr Rev 2008;29:381–402
    1. Chaurasia B, Summers SA. Ceramides - lipotoxic inducers of metabolic disorders. Trends Endocrinol Metab 2015;26:538–550
    1. Holland WL, Adams AC, Brozinick JT, et al. . An FGF21-adiponectin-ceramide axis controls energy expenditure and insulin action in mice. Cell Metab 2013;17:790–797
    1. Warshauer JT, Lopez X, Gordillo R, et al. . Effect of pioglitazone on plasma ceramides in adults with metabolic syndrome. Diabetes Metab Res Rev 2015;31:734–744
    1. Jornayvaz FR, Shulman GI. Diacylglycerol activation of protein kinase Cε and hepatic insulin resistance. Cell Metab 2012;15:574–584
    1. Xia JY, Holland WL, Kusminski CM, et al. . Targeted induction of ceramide degradation leads to improved systemic metabolism and reduced hepatic steatosis. Cell Metab 2015;22:266–278
    1. Sun K, Kusminski CM, Luby-Phelps K, et al. . Brown adipose tissue derived VEGF-A modulates cold tolerance and energy expenditure. Mol Metab 2014;3:474–483
    1. Xue R, Wan Y, Zhang S, Zhang Q, Ye H, Li Y. Role of bone morphogenetic protein 4 in the differentiation of brown fat-like adipocytes. Am J Physiol Endocrinol Metab 2014;306:E363–E372
    1. Rao RR, Long JZ, White JP, et al. . Meteorin-like is a hormone that regulates immune-adipose interactions to increase beige fat thermogenesis. Cell 2014;157:1279–1291
    1. Boström P, Wu J, Jedrychowski MP, et al. . A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 2012;481:463–468
    1. Unger RH, Scherer PE, Holland WL. Dichotomous roles of leptin and adiponectin as enforcers against lipotoxicity during feast and famine. Mol Biol Cell 2013;24:3011–3015
    1. Choi JW, Yoo BK, Shin CY, et al. . Uridine prevents the glucose deprivation-induced death of immunostimulated astrocytes via the action of uridine phosphorylase. Neurosci Res 2006;56:111–118
    1. Jones ME. Pyrimidine nucleotide biosynthesis in animals: genes, enzymes, and regulation of UMP biosynthesis. Annu Rev Biochem 1980;49:253–279
    1. Deng Y, Wang ZV, Tao C, et al. . The Xbp1s/GalE axis links ER stress to postprandial hepatic metabolism. J Clin Invest 2013;123:455–468
    1. Wang ZV, Deng Y, Gao N, et al. . Spliced X-box binding protein 1 couples the unfolded protein response to hexosamine biosynthetic pathway. Cell 2014;156:1179–1192
    1. Williams KW, Liu T, Kong X, et al. . Xbp1s in Pomc neurons connects ER stress with energy balance and glucose homeostasis. Cell Metab 2014;20:471–482
    1. Swyryd EA, Seaver SS, Stark GR. N-(phosphonacetyl)-L-aspartate, a potent transition state analog inhibitor of aspartate transcarbamylase, blocks proliferation of mammalian cells in culture. J Biol Chem 1974;249:6945–6950
    1. Cortés VA, Curtis DE, Sukumaran S, et al. . Molecular mechanisms of hepatic steatosis and insulin resistance in the AGPAT2-deficient mouse model of congenital generalized lipodystrophy. Cell Metab 2009;9:165–176
    1. Perry RJ, Zhang D, Zhang XM, Boyer JL, Shulman GI. Controlled-release mitochondrial protonophore reverses diabetes and steatohepatitis in rats. Science 2015;347:1253–1256
    1. Perry RJ, Kim T, Zhang XM, et al. . Reversal of hypertriglyceridemia, fatty liver disease, and insulin resistance by a liver-targeted mitochondrial uncoupler. Cell Metab 2013;18:740–748
    1. Combs TP, Nagajyothi, Mukherjee S, de Almeida CJG, et al. The adipocyte as an important target cell for Trypanosoma cruzi infection. J Biol Chem 2005;280:24085–24094
    1. Park J, Kusminski CM, Chua SC, Scherer PE. Leptin receptor signaling supports cancer cell metabolism through suppression of mitochondrial respiration in vivo. Am J Pathol 2010;177:3133–3144
    1. Park J, Euhus DM, Scherer PE. Paracrine and endocrine effects of adipose tissue on cancer development and progression. Endocr Rev 2011;32:550–570

Source: PubMed

3
Subscribe