PPAR gamma 2 prevents lipotoxicity by controlling adipose tissue expandability and peripheral lipid metabolism

Gema Medina-Gomez, Sarah L Gray, Laxman Yetukuri, Kenju Shimomura, Sam Virtue, Mark Campbell, R Keira Curtis, Mercedes Jimenez-Linan, Margaret Blount, Giles S H Yeo, Miguel Lopez, Tuulikki Seppänen-Laakso, Frances M Ashcroft, Matej Oresic, Antonio Vidal-Puig, Gema Medina-Gomez, Sarah L Gray, Laxman Yetukuri, Kenju Shimomura, Sam Virtue, Mark Campbell, R Keira Curtis, Mercedes Jimenez-Linan, Margaret Blount, Giles S H Yeo, Miguel Lopez, Tuulikki Seppänen-Laakso, Frances M Ashcroft, Matej Oresic, Antonio Vidal-Puig

Abstract

Peroxisome proliferator activated receptor gamma 2 (PPARg2) is the nutritionally regulated isoform of PPARg. Ablation of PPARg2 in the ob/ob background, PPARg2(-/-) Lep(ob)/Lep(ob) (POKO mouse), resulted in decreased fat mass, severe insulin resistance, beta-cell failure, and dyslipidaemia. Our results indicate that the PPARg2 isoform plays an important role, mediating adipose tissue expansion in response to positive energy balance. Lipidomic analyses suggest that PPARg2 plays an important antilipotoxic role when induced ectopically in liver and muscle by facilitating deposition of fat as relatively harmless triacylglycerol species and thus preventing accumulation of reactive lipid species. Our data also indicate that PPARg2 may be required for the beta-cell hypertrophic adaptive response to insulin resistance. In summary, the PPARg2 isoform prevents lipotoxicity by (a) promoting adipose tissue expansion, (b) increasing the lipid-buffering capacity of peripheral organs, and (c) facilitating the adaptive proliferative response of beta-cells to insulin resistance.

Conflict of interest statement

Competing interests. The authors have declared that no competing interests exist.

Figures

Figure 1. Physiological Characterisation of POKO Mouse
Figure 1. Physiological Characterisation of POKO Mouse
(A) Body weights (black circles, WT; black squares, ob/ob; white circles, PPARg2 KO; white squares, POKO) are shown for males (left) or females (right) (n = 5–12). *, p < 0.05 POKO versus ob/ob and §, p < 0.01 POKO versus WT. (B) Food intake from 20-wk-old female mice (n = 4) is shown. (C) Body composition analysis from 20-wk-old females is shown: WT, ob/ob, PPARg2 KO, and POKO mice fed chow diet mice (n = 4–7). *, p < 0.05 POKO versus WT and ###, p < 0.001 POKO versus ob/ob. (D) Haematoxylin and eosin (H and E)-stained sections (10×) from epididymal WAT from 16-wk-old male WT, ob/ob, and POKO mice. (E) Percent relative cumulative frequency analysis (PRCF) from epididymal WAT adipocytes from 16-wk-old male WT, ob/ob, PPARg2 KO, and POKO mice. (n = 4–5).
Figure 2. Early Insulin Resistance in POKO…
Figure 2. Early Insulin Resistance in POKO Mice Independent of Body Weight
(A) Body weight and plasma glucose levels from three, four, and five-week-old female WT, ob/ob, PPARg2 KO, and POKO. *, p < 0.05; **, p < 0.01; ***, p < 0.001 POKO versus ob/ob. (B) Plasma glucose levels during ITT on 4-wk-old male (left) and female (right) mice on chow diet (black triangle, WT; white triangle, PPARg2 KO; black square, ob/ob; black diamond, POKO) (n = 7). *, p < 0.05; **, p < 0.01 POKO versus ob/ob. (C) Morphological analysis of H and E-stained sections (10×) in pancreas from 4-wk-old males ob/ob and POKO mice (n = 5).
Figure 3. Impaired β-Cell Function and Hepatic…
Figure 3. Impaired β-Cell Function and Hepatic Morphological Analyis in the POKO Mice
(A) H and E-stained sections (10×) and immunohistochemical (20×) analysis of insulin and glucagon in pancreas from 16-wk-old males WT, ob/ob, and POKO mice (n = 5). (B) Insulin content of islets isolated from POKO (black bars), and ob/ob (grey bars) mice. Each data point is the mean of six samples each of five islets. (C) Insulin secretion from islets isolated from POKO (black bars) and ob/ob (grey bars) mice in response to glucose (1, 16 mM) or glucose 16 mM + tolbutamide (200 μM). Data were collected from six samples each of five islets from three mice of each genotype. For each sample, insulin release was normalised to insulin content. *, p < 0.05; **, p < 0.01; ***, p < 0.001 POKO versus ob/ob. (D) H and E-stained sections (4×) in liver from 16-wk-old males WT, ob/ob, and POKO mice (n = 5).
Figure 4. Lipidomic and Gene Expression Analysis…
Figure 4. Lipidomic and Gene Expression Analysis of POKO WAT
(A) Lipidomic profiling of WAT from 16-wk-old males WT, ob/ob, and POKO mice. (B) Adipose tissue mRNA levels from different genes from 16-wk-old male WT, PPARg2 KO, ob/ob, and POKO mice (n = 6–8). *, p < 0.05; **, p <0.01; ***, p <0.001 POKO versus ob/ob.
Figure 5. Lipidomic and Gene Expression Analysis…
Figure 5. Lipidomic and Gene Expression Analysis in Islets and Liver from POKO Mice
Lipidomic profiling of islets (A) and liver (B) from 16-wk-old males WT, PPARg2 KO, ob/ob, and POKO mice. TG, TAGs; DAGs, diacylglycerols; SM, sphingomyelins. (C) Liver gene expression from 16-wk-old male WT, ob/ob, PPARg2 KO, and POKO mice fed chow (n = 6–8). *, p < 0.05; **, p, <0.01; ***, p <0.001 POKO versus ob/ob; ###, p < 0.001 POKO versus WT; ‡‡‡, p < 0.001 ob/ob versus WT.
Figure 6. Storage of Lipids—Antilipotoxic Role of…
Figure 6. Storage of Lipids—Antilipotoxic Role of PPARg2
Antilipotoxic role of PPARg2 mediated by (a) expansion of adipose tissue and facilitation of triglyceride deposition and (b) facilitating deposition of fat in liver, skeletal muscle, and pancreas in the form of TAG. Ob/Ob mice can induce PPARg2 expression in liver, muscle, and β-cell, facilitating deposition of excess of energy in these organs in the form of TAG. Absence of inducibility of PPARg2 in POKO mouse liver, muscle, and β-cells results in increased deposition of reactive lipid species and decreased TAG, leading to marked insulin resistance and β-cell failure.

References

    1. Rosen ED, Spiegelman BM. Molecular regulation of adipogenesis. Annu Rev Cell Dev Biol. 2000;16:145–171.
    1. Medina-Gomez G, Virtue S, Lelliott C, Boiani R, Campbell M, et al. The link between nutritional status and insulin sensitivity is dependent on the adipocyte-specific peroxisome proliferator-activated receptor-gamma2 isoform. Diabetes. 2005;54:1706–1716.
    1. Dandona P, Aljada A, Bandyopadhyay A. Inflammation: The link between insulin resistance, obesity and diabetes. Trends Immunol. 2004;25:4–7.
    1. Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Invest. 2005;115:1111–1119.
    1. Kraegen EW, Cooney GJ, Ye JM, Thompson AL, Furler SM. The role of lipids in the pathogenesis of muscle insulin resistance and beta cell failure in type II diabetes and obesity. Exp Clin Endocrinol Diabetes. 2001;2:S189–S201. 109 Suppl.
    1. Lelliott C, Vidal-Puig AJ. Lipotoxicity, an imbalance between lipogenesis de novo and fatty acid oxidation. Int J Obes Relat Metab Disord. 2004;4:S22–S28. 28 Suppl.
    1. Unger RH. Minireview: Weapons of lean body mass destruction: The role of ectopic lipids in the metabolic syndrome. Endocrinology. 2003;144:5159–5165.
    1. McGarry JD. Banting lecture 2001: Dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Diabetes. 2002;51:7–18.
    1. Lee Y, Hirose H, Ohneda M, Johnson JH, McGarry JD, et al. Beta-cell lipotoxicity in the pathogenesis of non-insulin-dependent diabetes mellitus of obese rats: Impairment in adipocyte-beta-cell relationships. Proc Natl Acad Sci U S A. 1994;91:10878–10882.
    1. Lupi R, Dotta F, Marselli L, Del Guerra S, Masini M, 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;51:1437–1442.
    1. Unger RH, Orci L. Lipoapoptosis: Its mechanism and its diseases. Biochim Biophys Acta. 2002;1585:202–212.
    1. Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, et al. PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro. Mol Cell. 1999;4:611–617.
    1. Koutnikova H, Cock TA, Watanabe M, Houten SM, Champy MF, et al. Compensation by the muscle limits the metabolic consequences of lipodystrophy in PPAR gamma hypomorphic mice. Proc Natl Acad Sci U S A. 2003;100:14457–14462.
    1. Barak Y, Nelson MC, Ong ES, Jones YZ, Ruiz-Lozano P, et al. PPAR gamma is required for placental, cardiac, and adipose tissue development. Mol Cell. 1999;4:585–595.
    1. Spiegelman BM. PPAR-gamma: Adipogenic regulator and thiazolidinedione receptor. Diabetes. 1998;47:507–514.
    1. Escher P, Braissant O, Basu-Modak S, Michalik L, Wahli W, et al. Rat PPARs: Quantitative analysis in adult rat tissues and regulation in fasting and refeeding. Endocrinology. 2001;142:4195–4202.
    1. Werman A, Hollenberg A, Solanes G, Bjorbaek C, Vidal-Puig AJ, et al. Ligand-independent activation domain in the N terminus of peroxisome proliferator-activated receptor gamma (PPARgamma). Differential activity of PPARgamma1 and −2 isoforms and influence of insulin. J Biol Chem. 1997;272:20230–20235.
    1. Vidal-Puig A, Jimenez-Linan M, Lowell BB, Hamann A, Hu E, et al. Regulation of PPAR gamma gene expression by nutrition and obesity in rodents. J Clin Invest. 1996;97:2553–2561.
    1. Vidal-Puig AJ, Considine RV, Jimenez-Linan M, Werman A, Pories WJ, et al. Peroxisome proliferator-activated receptor gene expression in human tissues. Effects of obesity, weight loss, and regulation by insulin and glucocorticoids. J Clin Invest. 1997;99:2416–2422.
    1. Ren D, Collingwood TN, Rebar EJ, Wolffe AP, Camp HS. PPARgamma knockdown by engineered transcription factors: Exogenous PPARgamma2 but not PPARgamma1 reactivates adipogenesis. Genes Dev. 2002;16:27–32.
    1. Zhang J, Fu M, Cui T, Xiong C, Xu K, et al. Selective disruption of PPAR{gamma}2 impairs the development of adipose tissue and insulin sensitivity. Proc Natl Acad Sci U S A. 2004;101:10703–10708.
    1. He W, Barak Y, Hevener A, Olson P, Liao D, 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 U S A. 2003;100:15712–15717.
    1. Gavrilova O, Haluzik M, Matsusue K, Cutson JJ, Johnson L, et al. Liver peroxisome proliferator-activated receptor gamma contributes to hepatic steatosis, triglyceride clearance, and regulation of body fat mass. J Biol Chem. 2003;278:34268–34276.
    1. Matsusue K, Haluzik M, Lambert G, Yim SH, Gavrilova O, et al. Liver-specific disruption of PPARgamma in leptin-deficient mice improves fatty liver but aggravates diabetic phenotypes. J Clin Invest. 2003;111:737–747.
    1. Norris AW, Chen L, Fisher SJ, Szanto I, Ristow M, et al. Muscle-specific PPARgamma-deficient mice develop increased adiposity and insulin resistance but respond to thiazolidinediones. J Clin Invest. 2003;112:608–618.
    1. Hevener AL, He W, Barak Y, Le J, Bandyopadhyay G, et al. Muscle-specific Pparg deletion causes insulin resistance. Nat Med. 2003;9:1491–1497.
    1. Braissant O, Foufelle F, Scotto C, Dauca M, Wahli W. Differential expression of peroxisome proliferator-activated receptors (PPARs): Tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat. Endocrinology. 1996;137:354–366.
    1. Dubois M, Pattou F, Kerr-Conte J, Gmyr V, Vandewalle B, et al. Expression of peroxisome proliferator-activated receptor gamma (PPARgamma) in normal human pancreatic islet cells. Diabetologia. 2000;43:1165–1169.
    1. Patane G, Anello M, Piro S, Vigneri R, Purrello F, et al. Role of ATP production and uncoupling protein-2 in the insulin secretory defect induced by chronic exposure to high glucose or free fatty acids and effects of peroxisome proliferator-activated receptor-gamma inhibition. Diabetes. 2002;51:2749–2756.
    1. Rosen ED, Kulkarni RN, Sarraf P, Ozcan U, Okada T, et al. Targeted elimination of peroxisome proliferator-activated receptor gamma in beta cells leads to abnormalities in islet mass without compromising glucose homeostasis. Mol Cell Biol. 2003;23:7222–7229.
    1. Matsui J, Terauchi Y, Kubota N, Takamoto I, Eto K, et al. Pioglitazone reduces islet triglyceride content and restores impaired glucose-stimulated insulin secretion in heterozygous peroxisome proliferator-activated receptor-gamma-deficient mice on a high-fat diet. Diabetes. 2004;53:2844–2854.
    1. Oresic M, Vidal-Puig A, Hanninen V. Metabolomic approaches to phenotype characterization and applications to complex diseases. Expert Rev Mol Diagn. 2006;6:575–585.
    1. Takabe W, Kanai Y, Chairoungdua A, Shibata N, Toi S, et al. Lysophosphatidylcholine enhances cytokine production of endothelial cells via induction of L-type amino acid transporter 1 and cell surface antigen 4F2. Arterioscler Thromb Vasc Biol. 2004;24:1640–1645.
    1. Zoeller RA, Lake AC, Nagan N, Gaposchkin DP, Legner MA, et al. Plasmalogens as endogenous antioxidants: Somatic cell mutants reveal the importance of the vinyl ether. Biochem J. 1999;338((Pt 3)):769–776.
    1. Yao ZM, Vance DE. The active synthesis of phosphatidylcholine is required for very low density lipoprotein secretion from rat hepatocytes. J Biol Chem. 1988;263:2998–3004.
    1. Patsouris D, Reddy JK, Muller M, Kersten S. Peroxisome proliferator-activated receptor alpha mediates the effects of high-fat diet on hepatic gene expression. Endocrinology. 2006;147:1508–1516.
    1. Puigserver P, Rhee J, Donovan J, Walkey CJ, Yoon JC, et al. Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction. Nature. 2003;423:550–555.
    1. Shimomura I, Hammer RE, Richardson JA, Ikemoto S, Bashmakov Y, et al. Insulin resistance and diabetes mellitus in transgenic mice expressing nuclear SREBP-1c in adipose tissue: Model for congenital generalized lipodystrophy. Genes Dev. 1998;12:3182–3194.
    1. Ross SR, Graves RA, Spiegelman BM. Targeted expression of a toxin gene to adipose tissue: Transgenic mice resistant to obesity. Genes Dev. 1993;7:1318–1324.
    1. Moitra J, Mason MM, Olive M, Krylov D, Gavrilova O, et al. Life without white fat: A transgenic mouse. Genes Dev. 1998;12:3168–3181.
    1. Gray SL, Nora ED, Grosse J, Manieri M, Stoeger T, et al. Leptin deficiency unmasks the deleterious effects of impaired peroxisome proliferator-activated receptor gamma function (P465L PPARgamma) in mice. Diabetes. 2006;55:2669–2677.
    1. Hummel KP, Dickie MM, Coleman DL. Diabetes, a new mutation in the mouse. Science. 1966;153:1127–1128.
    1. Leiter EH. The influence of genetic background on the expression of mutations at the diabetes locus in the mouse IV. Male lethal syndrome in CBA/Lt mice. Diabetes. 1981;30:1035–1044.
    1. Mori H, Ikegami H, Kawaguchi Y, Seino S, Yokoi N, et al. The Pro12 –>Ala substitution in PPAR-gamma is associated with resistance to development of diabetes in the general population: Possible involvement in impairment of insulin secretion in individuals with type 2 diabetes. Diabetes. 2001;50:891–894.
    1. Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Oxidative stress and stress-activated signaling pathways: A unifying hypothesis of type 2 diabetes. Endocr Rev. 2002;23:599–622.
    1. Haber CA, Lam TK, Yu Z, Gupta N, Goh T, et al. N-acetylcysteine and taurine prevent hyperglycemia-induced insulin resistance in vivo: Possible role of oxidative stress. Am J Physiol Endocrinol Metab. 2003;285:E744–E753.
    1. Hildebrandt W, Hamann A, Krakowski-Roosen H, Kinscherf R, Dugi K, et al. Effect of thiol antioxidant on body fat and insulin reactivity. J Mol Med. 2004;82:336–344.
    1. Fridlyand LE, Philipson LH. Reactive species and early manifestation of insulin resistance in type 2 diabetes. Diabetes Obes Metab. 2006;8:136–145.
    1. Bloch-Damti A, Bashan N. Proposed mechanisms for the induction of insulin resistance by oxidative stress. Antioxid Redox Signal. 2005;7:1553–1567.
    1. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, et al. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112:1796–1808.
    1. Xu H, Barnes GT, Yang Q, Tan G, Yang D, 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. Qi C, Pekala PH. Tumor necrosis factor-alpha-induced insulin resistance in adipocytes. Proc Soc Exp Biol Med. 2000;223:128–135.
    1. Herzig S, Long F, Jhala US, Hedrick S, Quinn R, et al. CREB regulates hepatic gluconeogenesis through the coactivator PGC-1. Nature. 2001;413:179–183.
    1. Yoon JC, Puigserver P, Chen G, Donovan J, Wu Z, et al. Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1. Nature. 2001;413:131–138.
    1. Hirasawa T, Ohara T, Makino S. Genetic typing of the mouse ob mutation by PCR and restriction enzyme analysis. Exp Anim. 1997;46:75–78.
    1. Elia M, Livesey G. Energy expenditure and fuel selection in biological systems: The theory and practice of calculations based on indirect calorimetry and tracer methods. World Rev Nutr Diet. 1992;70:68–131.
    1. Gundersen HJ, Osterby R. Optimizing sampling efficiency of stereological studies in biology: Or ‘do more less well!'. J Microsc. 1981;121:65–73.
    1. Vidal-Puig AJ, Grujic D, Zhang CY, Hagen T, Boss O, et al. Energy metabolism in uncoupling protein 3 gene knockout mice. J Biol Chem. 2000;275:16258–16266.
    1. Katajamaa M, Oresic M. Processing methods for differential analysis of LC/MS profile data. BMC Bioinformatics. 2005;6:179.

Source: PubMed

3
Subscribe