R. Villegas, R. Delahanty, S. Williams, H. Li, O. Brian et al., Genetic Variation and Insulin Resistance 642 in Middle-Aged Chinese MenAccepted:epub ahead of print, Ann Hum Genet, p.643, 2015.

A. Majithia, B. Tsuda, M. Agostini, K. Gnanapradeepan, and R. Rice, Prospective functional classification of all possible missense variants in PPARG, Nature Genetics, vol.371, issue.12, pp.1570-1575, 2016.
DOI : 10.1007/s00125-013-2962-5

M. Pawlak, P. Lefebvre, and B. Staels, Molecular mechanism of PPAR?? action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease, Journal of Hepatology, vol.62, issue.3, pp.720-649, 2015.
DOI : 10.1016/j.jhep.2014.10.039

A. Montagner, A. Polizzi, E. Fouché, S. Ducheix, and Y. Lippi, Liver PPAR? is crucial for whole-body 651

S. Kersten, Integrated physiology and systems biology of PPAR??, Molecular Metabolism, vol.3, issue.4, pp.354-371, 2014.
DOI : 10.1016/j.molmet.2014.02.002

A. Janssen, B. Betzel, G. Stoopen, F. Berends, and I. Janssen, The impact of PPAR?? activation on whole genome gene expression in human precision cut liver slices, BMC Genomics, vol.5, issue.1, pp.760-655, 2015.
DOI : 10.1186/1471-2164-11-16

C. Longuet, E. Sinclair, A. Maida, L. Baggio, and M. Maziarz, The glucagon receptor is required for 656

M. Shao, B. Shan, Y. Liu, Y. Deng, and C. Yan, Hepatic IRE1? regulates fasting-induced metabolic 658 adaptive programs through the XBP1s-PPAR? axis signalling, Nat Commun, vol.5, pp.3528-659, 2014.

F. Damiano, G. Gnoni, and L. Siculella, Citrate carrier promoter is target of peroxisome proliferator-activated receptor alpha and gamma in hepatocytes and adipocytes, The International Journal of Biochemistry & Cell Biology, vol.44, issue.4, pp.659-668, 2012.
DOI : 10.1016/j.biocel.2012.01.003

B. Knight, A. Hebbachi, D. Hauton, A. Brown, and D. Wiggins, A role for PPAR?? in the control of SREBP activity and lipid synthesis in the liver, Biochemical Journal, vol.389, issue.2, pp.413-421, 2005.
DOI : 10.1042/BJ20041896

D. Patel, B. Knight, D. Wiggins, S. Humphreys, and G. Gibbons, Disturbances in the normal 665 regulation of SREBP-sensitive genes in PPAR alpha-deficient mice, J Lipid Res, vol.42, pp.328-337, 2001.

S. Sengupta, T. Peterson, M. Laplante, S. Oh, and D. Sabatini, mTORC1 controls fasting-induced ketogenesis and its modulation by ageing, Nature, vol.287, issue.7327, pp.1100-1104, 2010.
DOI : 10.1152/ajpheart.01051.2001

S. Francque, A. Verrijken, S. Caron, J. Prawitt, and R. Paumelle, PPAR? gene expression correlates 669 with severity and histological treatment response in patients with non-alcoholic steatohepatitis, J, vol.670

L. Zheng, G. Lv, J. Sheng, and Y. Yang, Effect of miRNA-10b in regulating cellular steatosis level by 672 targeting PPAR-alpha expression, a novel mechanism for the pathogenesis of NAFLD, J Gastroenterol, p.673

S. Vernia, J. Cavanagh-kyros, L. Garcia-haro, G. Sabio, and T. Barrett, The PPAR?-FGF21 hormone 678 axis contributes to metabolic regulation by the hepatic JNK signaling pathway, Cell Metab, vol.67920, pp.512-525, 2014.

B. Staels, N. Vu-dac, V. Kosykh, R. Saladin, and J. Fruchart, Fibrates downregulate apolipoprotein C-III expression independent of induction of peroxisomal acyl coenzyme A oxidase. A potential mechanism for the hypolipidemic action of fibrates., Journal of Clinical Investigation, vol.95, issue.2, pp.705-712, 1995.
DOI : 10.1172/JCI117717

J. Crosby, G. Peloso, P. Auer, D. Crosslin, and N. Stitziel, Loss-of-function mutations in APOC3, 687 triglycerides, and coronary disease, N Engl J Med, vol.371, pp.22-31, 2014.

P. Dongiovanni, S. Petta, C. Maglio, A. Fracanzani, and R. Pipitone, Transmembrane 6 superfamily 692 member 2 gene variant disentangles nonalcoholic steatohepatitis from cardiovascular disease, p.693

N. Vu-dac, K. Schoonjans, V. Kosykh, J. Dallongeville, and J. Fruchart, Fibrates increase human 699 apolipoprotein A-II expression through activation of the peroxisome proliferator-activated receptor, p.700

T. Goto, J. Lee, A. Teraminami, Y. Kim, and S. Hirai, Activation of peroxisome proliferator-activated 708

P. Seale, Transcriptional Regulatory Circuits Controlling Brown Fat Development and Activation, Diabetes, vol.64, issue.7, p.711
DOI : 10.2337/db15-0203

URL : http://diabetes.diabetesjournals.org/content/diabetes/64/7/2369.full.pdf

L. Hue and H. Taegtmeyer, The Randle cycle revisited: a new head for an old hat, American Journal of Physiology-Endocrinology and Metabolism, vol.297, issue.3
DOI : 10.1152/ajpheart.00870.2007

A. Peeters and M. Baes, Role of PPAR? in Hepatic Carbohydrate Metabolism, PPAR Res, p.715, 2010.

F. Lalloyer, B. Vandewalle, F. Percevault, G. Torpier, and J. Kerr-conte, Peroxisome Proliferator-Activated Receptor ?? Improves Pancreatic Adaptation to Insulin Resistance in Obese Mice and Reduces Lipotoxicity in Human Islets, Diabetes, vol.55, issue.6, pp.1605-1613, 2006.
DOI : 10.2337/db06-0016

F. Oger, J. Dubois-chevalier, C. Gheeraert, S. Avner, and E. Durand, Peroxisome proliferator- 728 activated receptor ? regulates genes involved in insulin/insulin-like growth factor signaling and lipid 729 metabolism during adipogenesis through functionally distinct enhancer classes, J Biol Chem, vol.730289, pp.708-722, 2014.

R. Siersbaek, S. Baek, A. Rabiee, R. Nielsen, and S. Traynor, Molecular Architecture of Transcription Factor Hotspots in Early Adipogenesis, Cell Reports, vol.7, issue.5, pp.1434-1442, 2014.
DOI : 10.1016/j.celrep.2014.04.043

J. Dubois-chevalier, F. Oger, H. Dehondt, F. Firmin, and C. Gheeraert, A dynamic CTCF chromatin binding landscape promotes DNA hydroxymethylation and transcriptional induction of adipocyte differentiation, Nucleic Acids Research, vol.19, issue.17, pp.10943-10959, 2014.
DOI : 10.1101/gr.082800.108

L. Bacquer, O. Petroulakis, E. Paglialunga, S. Poulin, F. Richard et al., Elevated sensitivity to diet-induced obesity and insulin resistance in mice lacking 4E-BP1 and 4E-BP2, Journal of Clinical Investigation, vol.117, issue.2, pp.387-738, 2007.
DOI : 10.1172/JCI29528DS1

J. Kim and J. Chen, Regulation of Peroxisome Proliferator-Activated Receptor-?? Activity by Mammalian Target of Rapamycin and Amino Acids in Adipogenesis, Diabetes, vol.53, issue.11, pp.2748-2756, 2004.
DOI : 10.2337/diabetes.53.11.2748

J. Yu, S. Javorschi, A. Hevener, Y. Kruszynska, and R. Norman, The Effect of Thiazolidinediones on Plasma Adiponectin Levels in Normal, Obese, and Type 2 Diabetic Subjects, Diabetes, vol.51, issue.10, pp.2968-743, 2002.
DOI : 10.2337/diabetes.51.10.2968

C. Kallen and M. Lazar, Antidiabetic thiazolidinediones inhibit leptin (ob) gene expression in 3T3-L1 adipocytes., Proceedings of the National Academy of Sciences, vol.93, issue.12, pp.5793-5796, 1996.
DOI : 10.1073/pnas.93.12.5793

E. Ip, G. Farrell, P. Hall, G. Robertson, and I. Leclercq, Administration of the potent PPARalpha agonist, p.754

W. Greenstein, A. Majumdar, N. Yang, P. Subbaiah, P. Kineman et al., Hepatocyte-specific, p.756

M. Gao, Y. Ma, M. Alsaggar, and D. Liu, Dual Outcomes of Rosiglitazone Treatment on Fatty Liver, The AAPS Journal, vol.18, issue.4
DOI : 10.1208/s12248-016-9919-9

A. Sanyal, N. Chalasani, K. Kowdley, A. Mccullough, and A. Diehl, Pioglitazone, Vitamin E, or Placebo for Nonalcoholic Steatohepatitis, New England Journal of Medicine, vol.362, issue.18, pp.1675-1685, 2010.
DOI : 10.1056/NEJMoa0907929

V. Ratziu, P. Giral, S. Jacqueminet, C. F. Hartemann-heurtier, and A. , Rosiglitazone for 765 nonalcoholic steatohepatitis: one-year results of the randomized placebo-controlled Fatty Liver 766

M. Lu, D. Sarruf, S. Talukdar, S. Sharma, and P. Li, Brain PPAR-?? promotes obesity and is required for the insulin???sensitizing effect of thiazolidinediones, Nature Medicine, vol.17, issue.5, pp.618-622, 2011.
DOI : 10.1152/ajpendo.00668.2009

J. Fu, S. Gaetani, F. Oveisi, L. Verme, J. Serrano et al., Oleylethanolamide regulates feeding and body weight through activation of the nuclear receptor PPAR-??, Nature, vol.282, issue.6953, pp.90-93, 2003.
DOI : 10.1152/ajpendo.00072.2001

R. Kimura, N. Takahashi, T. Goto, K. Murota, and T. Kawada, Activation of peroxisome proliferator-activated receptor-?? (PPAR??) in proximal intestine improves postprandial lipidemia in obese diabetic KK-Ay mice, Obesity Research & Clinical Practice, vol.7, issue.5, pp.353-60, 2013.
DOI : 10.1016/j.orcp.2013.05.005

S. Colin, O. Briand, V. Touche, K. Wouters, and M. Baron, Activation of intestinal peroxisome 775 proliferator-activated receptor-? increases high-density lipoprotein production, Eur Heart J, vol.77634, pp.2566-2574, 2013.

S. Heinz, C. Benner, N. Spann, E. Bertolino, and Y. Lin, Simple Combinations of Lineage-Determining Transcription Factors Prime cis-Regulatory Elements Required for Macrophage and B Cell Identities, Molecular Cell, vol.38, issue.4, pp.576-589, 2010.
DOI : 10.1016/j.molcel.2010.05.004

J. Dispirito, B. Fang, F. Wang, and M. Lazar, Pruning of the Adipocyte Peroxisome Proliferator-Activated Receptor ?? Cistrome by Hematopoietic Master Regulator PU.1, Molecular and Cellular Biology, vol.33, issue.16, pp.3354-788, 2013.
DOI : 10.1128/MCB.00599-13

C. Settembre, D. Cegli, R. Mansueto, G. Saha, P. Vetrini et al., TFEB controls cellular lipid metabolism through a starvation-induced autoregulatory loop, Nature Cell Biology, vol.10, issue.6, pp.647-658, 2013.
DOI : 10.1016/j.cmet.2009.10.002

X. Yang, M. Downes, R. Yu, A. Bookout, and W. He, Nuclear Receptor Expression Links the Circadian Clock to Metabolism, Cell, vol.126, issue.4, pp.801-810, 2006.
DOI : 10.1016/j.cell.2006.06.050

D. Ratman, V. Mylka, N. Bougarne, M. Pawlak, and S. Caron, Chromatin recruitment of activated 802 AMPK drives fasting response genes co-controlled by GR and PPAR?, Nucleid Acids Res, vol.803, p.804, 2016.

J. Marino, L. Stechschulte, D. Stec, A. Nestor-kalinoski, and S. Coleman, Glucocorticoid receptor 805 ? induces hepatic steatosis by augmenting inflammation and inhibition of the peroxisome 806 proliferator-activated receptor (PPAR) ?Accepted:epub ahead of print, J Biol Chem, p.807, 2016.

F. Picard, M. Kurtev, N. Chung, A. Topark-ngarm, and T. Senawong, Erratum: Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-??, Nature, vol.272, issue.6993, pp.771-776, 2004.
DOI : 10.1074/jbc.272.3.2013

K. Kajita, T. Mune, T. Ikeda, M. Matsumoto, and Y. Uno, Effect of fasting on PPAR?? and AMPK activity in adipocytes, Diabetes Research and Clinical Practice, vol.81, issue.2, pp.144-149, 2008.
DOI : 10.1016/j.diabres.2008.05.003

M. Hatori, C. Vollmers, A. Zarrinpar, L. Ditacchio, and E. Bushong, Time-restricted feeding 812 without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet, Cell Metab, vol.81315, pp.848-860, 2012.

K. Eckel-mahan, V. Patel, S. De-mateo, R. Orozco-solis, and N. Ceglia, Reprogramming of the Circadian Clock by Nutritional Challenge, Cell, vol.155, issue.7, pp.1464-1478, 2013.
DOI : 10.1016/j.cell.2013.11.034

X. Liang, F. Bushman, and G. Fitzgerald, Rhythmicity of the intestinal microbiota is regulated by gender and the host circadian clock, Proceedings of the National Academy of Sciences, vol.5, issue.10, pp.10479-10484, 2015.
DOI : 10.1093/molbev/msp077

M. Murakami, P. Tognini, Y. Liu, K. Eckel-mahan, and P. Baldi, Gut microbiota directs PPAR?- 819 driven reprogramming of the liver circadian clock by nutritional challenge, EMBO Rep, vol.820, p.821, 2016.

T. Inagaki, P. Dutchak, G. Zhao, X. Ding, and L. Gautron, Endocrine Regulation of the Fasting Response by PPAR??-Mediated Induction of Fibroblast Growth Factor 21, Cell Metabolism, vol.5, issue.6, pp.415-823, 2007.
DOI : 10.1016/j.cmet.2007.05.003

X. Zhang, D. Yeung, M. Karpisek, D. Stejskal, and Z. Zhou, Serum FGF21 levels are increased in 825 obesity and are independently associated with the metabolic syndrome in humans, Diabetes, vol.82657, pp.1246-1253, 2008.

P. Dutchak, T. Katafuchi, A. Bookout, J. Choi, and R. Yu, Fibroblast growth factor-21 regulates 828

J. Jonker, J. Suh, A. Atkins, M. Ahmadian, and P. Li, A PPAR?-FGF1 axis is required for adaptive 830

W. So, Q. Cheng, L. Chen, C. Evans-molina, and A. Xu, High Glucose Represses ??-Klotho Expression and Impairs Fibroblast Growth Factor 21 Action in Mouse Pancreatic Islets: Involvement of Peroxisome Proliferator-Activated Receptor ?? Signaling, Diabetes, vol.62, issue.11, pp.3751-3759, 2013.
DOI : 10.2337/db13-0645

M. Donath and S. Shoelson, Type 2 diabetes as an inflammatory disease, Nature Reviews Immunology, vol.52, issue.2, pp.98-107, 2011.
DOI : 10.1007/s00125-008-1239-x

M. Bouhlel, B. Derudas, E. Rigamonti, R. Dièvart, and J. Brozek, PPARgamma activation primes 841 human monocytes into alternative M2 macrophages with anti-inflammatory properties, Cell Metab, vol.8426, pp.137-143, 2007.

S. Bapat, M. Suh, J. Fang, S. Liu, S. Zhang et al., Depletion of fat-resident Treg cells prevents 844 age-associated insulin resistance, Nature, vol.528, pp.137-141, 2015.

D. Cipolletta, P. Cohen, B. Spiegelman, C. Benoist, and D. Mathis, Appearance and disappearance of 846

D. Cipolletta, M. Feuerer, A. Li, N. Kamei, and J. Lee, PPAR-?? is a major driver of the accumulation and phenotype of adipose tissue Treg cells, Nature, vol.9, issue.7404, pp.549-553, 2012.
DOI : 10.1038/ni1008-1091

M. Angela, Y. Endo, H. Asou, T. Yamamoto, and D. Tumes, Fatty acid metabolic reprogramming 851 via mTOR-mediated inductions of PPAR? directs early activation of T cells, Nat Commun, vol.8527, pp.13683-853, 2016.

R. Mansouri, E. Baugé, B. Staels, and P. Gervois, Systemic and distal repercussions of liver-specific 854

G. Chinetti-gbaguidi and B. Staels, Lipid ligand-activated transcription factors regulating lipid storage and release in human macrophages, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1791, issue.6, pp.486-493, 2009.
DOI : 10.1016/j.bbalip.2009.01.009

P. Delerive, D. Bosscher, K. Besnard, S. , V. Berghe et al., Peroxisome Proliferator-activated Receptor ?? Negatively Regulates the Vascular Inflammatory Gene Response by Negative Cross-talk with Transcription Factors NF-??B and AP-1, Journal of Biological Chemistry, vol.91, issue.45, pp.32048-861, 1999.
DOI : 10.1073/pnas.95.5.2050

S. Chung, B. Kang, S. Kim, Y. Pak, and D. Cho, Oxidized low density lipoprotein inhibits 863 interleukin-12 production in lipopolysaccharide-activated mouse macrophages via direct interactions 864

D. Mogilenko, I. Kudriavtsev, V. Shavva, E. Dizhe, and E. Vilenskaya, Peroxisome proliferator- 870 activated receptor ? positively regulates complement C3 expression but inhibits tumor necrosis 871 factor ?-mediated activation of C3 gene in mammalian hepatic-derived cells, J Biol Chem, vol.872288, pp.1726-1738, 2013.

N. Bougarne, R. Paumelle, S. Caron, N. Hennuyer, and R. Mansouri, PPARalpha blocks 874 glucocorticoid receptor alpha-mediated transactivation but cooperates with the activated 875 glucocorticoid receptor alpha for transrepression on NF-kappaB, Proc Natl Acad Sci, vol.876106, pp.7397-7402, 2009.

G. Pascual, A. Fong, S. Ogawa, A. Gamliel, and A. Li, A SUMOylation-dependent pathway mediates transrepression of inflammatory response genes by PPAR-??, Nature, vol.2, issue.7059, pp.759-879, 2005.
DOI : 10.1038/35056591

R. Stienstra, S. Mandard, N. Tan, W. Wahli, and C. Trautwein, The Interleukin-1 receptor antagonist is a direct target gene of PPAR?? in liver, Journal of Hepatology, vol.46, issue.5, pp.869-877, 2007.
DOI : 10.1016/j.jhep.2006.11.019

C. Meier, R. Chicheportiche, C. Juge-aubry, M. Dreyer, and J. Dayer, REGULATION OF THE INTERLEUKIN-1 RECEPTOR ANTAGONIST IN THP-1 CELLS BY LIGANDS OF THE PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR ??, Cytokine, vol.18, issue.6, pp.320-328, 2002.
DOI : 10.1006/cyto.2002.1945

A. Othman, R. Benghozi, I. Alecu, Y. Wei, and E. Niesor, Fenofibrate lowers atypical sphingolipids in plasma of dyslipidemic patients: A novel approach for treating diabetic neuropathy?, Journal of Clinical Lipidology, vol.9, issue.4, pp.568-575, 2015.
DOI : 10.1016/j.jacl.2015.03.011

D. Gorden, D. Myers, P. Ivanova, E. Fahy, and M. Maurya, Biomarkers of NAFLD progression: a lipidomics approach to an epidemic, Journal of Lipid Research, vol.1841, issue.3, pp.722-736, 2015.
DOI : 10.1038/nrgastro.2012.256

J. Skogsberg, K. Kannisto, T. Cassel, A. Hamsten, and P. Eriksson, Evidence that peroxisome 896 proliferator-activated receptor delta influences cholesterol metabolism in men, Arterioscler Thromb, vol.897

M. Vänttinen, P. Nuutila, T. Kuulasmaa, J. Pihlajamäki, and K. Hällsten, Single Nucleotide Polymorphisms in the Peroxisome Proliferator-Activated Receptor ?? Gene Are Associated With Skeletal Muscle Glucose Uptake, Diabetes, vol.54, issue.12, pp.3587-3591, 2005.
DOI : 10.2337/diabetes.54.12.3587

L. Andrulionyte, P. Peltola, J. Chiasson, and M. Laakso, Single nucleotide polymorphisms of PPARD in 902 combination with the Gly482Ser substitution of PGC-1A and the Pro12Ala substitution of PPARG2 903 predict the conversion from impaired glucose tolerance to type 2 diabetes: the STOP-NIDDM trial, p.904

U. Risérus, D. Sprecher, T. Johnson, E. Olson, and S. Hirschberg, Activation of peroxisome 906 proliferator-activated receptor (PPAR)delta promotes reversal of multiple metabolic abnormalities, 907 reduces oxidative stress, and increases fatty acid oxidation in moderately obese men, Diabetes, vol.90857, pp.332-339, 2008.

H. Bays, S. Schwartz, T. Littlejohn, B. Kerzner, and R. Krauss, MBX-8025, A Novel Peroxisome Proliferator Receptor-?? Agonist: Lipid and Other Metabolic Effects in Dyslipidemic Overweight Patients Treated with and without Atorvastatin, The Journal of Clinical Endocrinology & Metabolism, vol.96, issue.9, pp.2889-2897, 2011.
DOI : 10.1210/jc.2011-1061

W. Oliver, J. Shenk, M. Snaith, C. Russell, and K. Plunket, A selective peroxisome 913 proliferator-activated receptor delta agonist promotes reverse cholesterol transport, Proc Natl Acad, p.914

X. Li, J. Li, X. Lu, H. Ma, and H. Shi, Treatment with PPAR? agonist alleviates non-alcoholic fatty liver 916 disease by modulating glucose and fatty acid metabolic enzymes in a rat model, Int J Mol Med, vol.91736, pp.767-775, 2015.

Y. Wang, C. Lee, S. Tiep, R. Yu, and J. Ham, Peroxisome-Proliferator-Activated Receptor ?? Activates Fat Metabolism to Prevent Obesity, Cell, vol.113, issue.2, pp.159-170, 2003.
DOI : 10.1016/S0092-8674(03)00269-1

URL : https://doi.org/10.1016/s0092-8674(03)00269-1

T. Tanaka, J. Yamamoto, S. Iwasaki, H. Asaba, and H. Hamura, Activation of peroxisome proliferator-activated receptor ?? induces fatty acid ??-oxidation in skeletal muscle and attenuates metabolic syndrome, Proceedings of the National Academy of Sciences, vol.300, issue.5622, pp.15924-15929, 2003.
DOI : 10.1126/science.1082889

L. Roberts, A. Murray, D. Menassa, T. Ashmore, and A. Nicholls, The contrasting roles of PPAR? 924

E. Barroso, R. Rodríguez-calvo, L. Serrano-marco, A. Astudillo, and J. Balsinde, The PPAR?/? 933 activator GW501516 prevents the down-regulation of AMPK caused by a high-fat diet in liver and 934 amplifies the PGC-1?-Lipin 1-PPAR? pathway leading to increased fatty acid oxidation, p.935

C. Vrins, A. Van-der-velde, K. Van-den-oever, J. Levels, and S. Huet, Peroxisome proliferator-activated receptor delta activation leads to increased transintestinal cholesterol efflux, Journal of Lipid Research, vol.19, issue.10, pp.2046-2054, 2009.
DOI : 10.1152/ajpgi.90231.2008

URL : http://www.jlr.org/content/50/10/2046.full.pdf

P. Thulin, B. Glinghammar, J. Skogsberg, K. Lundell, and E. Ehrenborg, PPARdelta increases expression of 943 the human apolipoprotein A-II gene in human liver cells, Int J Mol Med, vol.21, pp.819-824, 2008.

K. Chehaibi, L. Cedó, J. Metso, X. Palomer, and D. Santos, PPAR-??/?? activation promotes phospholipid transfer protein expression, Biochemical Pharmacology, vol.94, issue.2, pp.101-108, 2015.
DOI : 10.1016/j.bcp.2015.01.016

E. Barroso, R. Rodríguez-rodríguez, M. Chacón, E. Maymó-masip, and L. Ferrer, PPAR?/? 947 ameliorates fructose-induced insulin resistance in adipocytes by preventing Nrf2 activation, Biochim, vol.948

K. Kang, S. Reilly, V. Karabacak, M. Gangl, and K. Fitzgerald, Adipocyte-Derived Th2 Cytokines and Myeloid PPAR?? Regulate Macrophage Polarization and Insulin Sensitivity, Cell Metabolism, vol.7, issue.6, pp.485-495, 2008.
DOI : 10.1016/j.cmet.2008.04.002

URL : https://doi.org/10.1016/j.cmet.2008.04.002

T. Coll, D. Alvarez-guardia, E. Barroso, A. Gómez-foix, and X. Palomer, Activation of peroxisome 956 proliferator-activated receptor-{delta} by GW501516 prevents fatty acid-induced nuclear factor- 957

S. Luquet, J. Lopez-soriano, D. Holst, A. Fredenrich, and J. Melki, Peroxisome proliferator-activated receptor ?? controls muscle development and oxidative capability, The FASEB Journal, vol.17, issue.15, pp.2299-2301, 2003.
DOI : 10.1096/fj.03-0269fje

M. Schuler, F. Ali, C. Chambon, D. Duteil, and J. Bornert, PGC1?? expression is controlled in skeletal muscles by PPAR??, whose ablation results in fiber-type switching, obesity, and type 2 diabetes, Cell Metabolism, vol.4, issue.5, pp.407-414, 2006.
DOI : 10.1016/j.cmet.2006.10.003

Z. Gan, J. Rumsey, B. Hazen, L. Lai, and T. Leone, Nuclear receptor/microRNA circuitry links muscle fiber type to energy metabolism, Journal of Clinical Investigation, vol.123, issue.6, pp.2564-2575, 2013.
DOI : 10.1172/JCI67652DS1

URL : http://www.jci.org/articles/view/67652/files/pdf

P. Albers, A. Pedersen, J. Birk, D. Kristensen, and B. Vind, Human muscle fiber type-specific 967

A. Oberbach, Y. Bossenz, S. Lehmann, J. Niebauer, and V. Adams, Altered Fiber Distribution and Fiber-Specific Glycolytic and Oxidative Enzyme Activity in Skeletal Muscle of Patients With Type 2 Diabetes, Diabetes Care, vol.29, issue.4, pp.895-900, 2006.
DOI : 10.2337/diacare.29.04.06.dc05-1854

C. Lee, P. Olson, A. Hevener, I. Mehl, and L. Chong, PPAR?? regulates glucose metabolism and insulin sensitivity, Proceedings of the National Academy of Sciences, vol.3, issue.4, pp.3444-3449, 2006.
DOI : 10.2337/diabetes.54.4.935

URL : http://www.pnas.org/content/103/9/3444.full.pdf

S. Liu, B. Hatano, M. Zhao, Y. C. Kang, and K. , Role of Peroxisome Proliferator-activated Receptor ??/?? in Hepatic Metabolic Regulation, Journal of Biological Chemistry, vol.42, issue.2, pp.1237-1247, 2011.
DOI : 10.1038/nm1166

M. Daoudi, N. Hennuyer, M. Borland, V. Touche, and C. Duhem, PPAR??/?? Activation Induces Enteroendocrine L Cell GLP-1 Production, Gastroenterology, vol.140, issue.5, pp.1564-1574, 2011.
DOI : 10.1053/j.gastro.2011.01.045

A. Sahebkar, G. Chew, and G. Watts, New peroxisome proliferator-activated receptor agonists: potential treatments for atherogenic dyslipidemia and non-alcoholic fatty liver disease, Expert Opinion on Pharmacotherapy, vol.97, issue.1, p.981
DOI : 10.1136/hrt.2010.204990

N. Hennuyer, I. Duplan, C. Paquet, J. Vanhoutte, and E. Woitrain, The novel selective PPAR?? modulator (SPPARM??) pemafibrate improves dyslipidemia, enhances reverse cholesterol transport and decreases inflammation and atherosclerosis, Atherosclerosis, vol.249, pp.200-208, 2016.
DOI : 10.1016/j.atherosclerosis.2016.03.003

S. Ishibashi, S. Yamashita, H. Arai, E. Araki, and K. Yokote, Effects of K-877, a novel selective PPAR?? modulator (SPPARM??), in dyslipidaemic patients: A randomized, double blind, active- and placebo-controlled, phase 2 trial, Atherosclerosis, vol.249, pp.36-43, 2016.
DOI : 10.1016/j.atherosclerosis.2016.02.029

A. Khera, J. Millar, G. Ruotolo, M. Wang, and D. Rader, Potent peroxisome proliferator-activated receptor-?? agonist treatment increases cholesterol efflux capacity in humans with the metabolic syndrome, European Heart Journal, vol.35, issue.27, pp.3020-3022, 2015.
DOI : 10.1093/eurheartj/ehu105

F. Dunn, L. Higgins, J. Fredrickson, and A. Depaoli, Selective modulation of PPAR? activity can lower 998 plasma glucose without typical thiazolidinedione side-effects in patients with Type 2 diabetes, p.999

C. Pollock, O. Rodriguez, P. Martin, C. Albanese, and X. Li, Induction of metastatic gastric cancer 1001 by peroxisome proliferator-activated receptor? activation, PPAR Res, pp.571783-1002, 2010.

B. He, Z. Ning, Z. Li, S. Shan, and D. Pan, In vitro and in vivo characterizations of chiglitazar, a 1003 newly identified PPAR pan-agonist, PPAR Res, vol.2012, pp.546548-1004, 2012.

N. Ruzehaji, C. Frantz, M. Ponsoye, J. Avouac, and S. Pezet, Pan PPAR agonist IVA337 is effective in prevention and treatment of experimental skin fibrosis, Annals of the Rheumatic Diseases, vol.185, issue.18, pp.2175-2183, 2016.
DOI : 10.1016/j.ajpath.2015.01.018

M. Wright, M. Bortolini, M. Tadayyon, and M. Bopst, Minireview: Challenges and Opportunities in Development of PPAR Agonists, Molecular Endocrinology, vol.28, issue.11, pp.1756-1768, 2014.
DOI : 10.1210/me.2013-1427

. Randomized, Double-blind Study to Evaluate the Safety and Efficacy of Saroglitazar 2 and 4 mg 1013

R. Jani, V. Pai, P. Jha, G. Jariwala, and S. Mukhopadhyay, A Multicenter, Prospective, Randomized, Double-Blind Study to Evaluate the Safety and Efficacy of Saroglitazar 2 and 4???mg Compared with Placebo in Type 2 Diabetes Mellitus Patients Having Hypertriglyceridemia Not Controlled with Atorvastatin Therapy (PRESS VI), Diabetes Technology & Therapeutics, vol.16, issue.2, pp.63-71, 2014.
DOI : 10.1089/dia.2013.0253

T. Goto, R. Nakayama, M. Yamanaka, M. Takata, and T. Takazawa, Effects of DSP-8658, a novel 1020 selective peroxisome proliferator-activated receptors a/? modulator, on adipogenesis and glucose 1021 metabolism in diabetic obese mice, Exp Clin Endocrinol Diabetes, vol.123, pp.492-499, 2015.

M. Delmedico, D. Severynse-stevens, and W. Oliver, DB959 is a novel, dual PPAR ?/? agonist which 1023 controls glucose and regulates triglycerides and HDLc in animal models of T2D and dyslipidemia, pp.69-1024

B. Cariou, Y. Zaïr, B. Staels, and E. Bruckert, Effects of the New Dual PPAR??/?? Agonist GFT505 on Lipid and Glucose Homeostasis in Abdominally Obese Patients With Combined Dyslipidemia or Impaired Glucose Metabolism, Diabetes Care, vol.34, issue.9, pp.2008-2014, 2011.
DOI : 10.2337/dc11-0093

B. Cariou, R. Hanf, S. Lambert-porcheron, Y. Zaïr, and V. Sauvinet, Dual Peroxisome Proliferator-Activated Receptor ??/?? Agonist GFT505 Improves Hepatic and Peripheral Insulin Sensitivity in Abdominally Obese Subjects, Diabetes Care, vol.36, issue.10, pp.2923-2930, 2013.
DOI : 10.2337/dc12-2012

B. Staels, M. Maes, and A. Zambon, Fibrates and future PPAR?? agonists in the treatment of cardiovascular disease, Nature Clinical Practice Cardiovascular Medicine, vol.99, issue.9, pp.542-553, 2008.
DOI : 10.1161/01.CIR.102.1.21