A. Afshin, M. H. Forouzanfar, M. B. Reitsma, P. Sur, K. Estep et al., Health effects of overweight and obesity in 195 countries over 25 years, New England Journal of Medicine, vol.377, issue.1, pp.13-27, 2017.

E. A. Finkelstein, O. A. Khavjou, H. Thompson, J. G. Trogdon, L. Pan et al., Obesity and severe obesity forecasts through 2030, American Journal of Preventive Medicine, vol.42, issue.6, pp.563-570, 2012.
DOI : 10.1016/j.amepre.2011.10.026

J. P. Boyle, A. A. Honeycutt, K. M. Narayan, T. J. Hoerger, L. S. Geiss et al., Projection of diabetes burden through 2050: impact of changing demography and disease prevalence in the, Diabetes Care, vol.24, issue.11, pp.1936-1940, 2001.

L. Sjöström, Review of the key results from the Swedish Obese Subjects (SOS) trial -a prospective controlled intervention study of bariatric surgery, Journal of Internal Medicine, vol.273, issue.3, pp.219-234, 2013.

M. A. Pearen and G. E. Muscat, Orphan nuclear receptors and the regulation of nutrient metabolism: understanding obesity, Physiology (Bethesda), vol.27, issue.3, pp.156-166, 2012.

I. G. Schulman, Nuclear receptors as drug targets for metabolic disease, Advanced Drug Delivery Reviews, vol.62, issue.13, pp.1307-1315, 2010.
DOI : 10.1016/j.addr.2010.07.002

URL : http://europepmc.org/articles/pmc2987515?pdf=render

V. K. Dhiman, M. J. Bolt, and K. P. White, Nuclear receptors in cancer e uncovering new and evolving roles through genomic analysis, Nature Reviews Genetics, vol.19, issue.3, pp.160-174, 2018.
DOI : 10.1038/nrg.2017.102

P. Germain, B. Staels, C. Dacquet, M. Spedding, and V. Laudet, Overview of nomenclature of nuclear receptors, Pharmacological Reviews, vol.58, issue.4, pp.685-704, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00187929

N. Tanaka, T. Aoyama, S. Kimura, and F. J. Gonzalez, Targeting nuclear receptors for the treatment of fatty liver disease, Pharmacology & Therapeutics, vol.179, pp.142-157, 2017.

H. I. Swanson, T. Wada, W. Xie, B. Renga, A. Zampella et al., Role of nuclear receptors in lipid dysfunction and obesity-related diseases, Drug Metabolism & Disposition, vol.41, issue.1, pp.1-11, 2013.

A. M. Jetten, Retinoid-related orphan receptors (RORs): critical roles in development, immunity, circadian rhythm, and cellular metabolism, Nuclear Receptor Signaling, vol.7, p.3, 2009.
DOI : 10.1621/nrs.07003

URL : http://europepmc.org/articles/pmc2670432?pdf=render

R. L. Fitzsimmons, P. Lau, and G. E. Muscat, Retinoid-related orphan receptor alpha and the regulation of lipid homeostasis, The Journal of Steroid Biochemistry and Molecular Biology, vol.130, issue.3e5, pp.159-168, 2012.

D. P. Marciano, M. R. Chang, C. A. Corzo, D. Goswami, V. Q. Lam et al., The therapeutic potential of nuclear receptor modulators for treatment of metabolic disorders: PPARg, RORs, and Rev-erbs, Cell Metabolism, vol.19, issue.2, pp.193-208, 2014.

I. Dussault, D. Fawcett, A. Matthyssen, J. A. Bader, and V. Giguère, Orphan nuclear receptor ROR alpha-deficient mice display the cerebellar defects of staggerer, Mechanisms of Development, vol.70, issue.1e2, pp.147-153, 1998.

P. Lau, R. L. Fitzsimmons, S. Raichur, S. C. Wang, A. Lechtken et al., The orphan nuclear receptor, RORalpha, regulates gene expression that controls lipid metabolism: staggerer (SG/SG) mice are resistant to diet-induced obesity, Journal of Biological Chemistry, vol.283, issue.26, pp.18411-18421, 2008.

S. Kadiri, C. Monnier, M. Ganbold, T. Ledent, J. Capeau et al., The nuclear retinoid-related orphan receptor-a regulates adipose tissue glyceroneogenesis in addition to hepatic gluconeogenesis, American Journal of Physiology Endocrinology and Metabolism, vol.309, issue.2, pp.105-114, 2015.

Y. H. Han, H. J. Kim, H. Na, M. W. Nam, J. Y. Kim et al., RORa induces KLF4-mediated M2 polarization in the liver macrophages that protect against nonalcoholic steatohepatitis, Cell Reports, vol.20, issue.1, pp.124-135, 2017.
DOI : 10.1016/j.celrep.2017.06.017

URL : https://doi.org/10.1016/j.celrep.2017.06.017

E. J. Kim, Y. S. Yoon, S. Hong, H. Y. Son, T. Y. Na et al., Retinoic acid receptor-related orphan receptor a-induced activation of adenosine monophosphate-activated protein kinase results in attenuation of hepatic steatosis, Hepatology, vol.55, issue.5, pp.1379-1388, 2012.

Y. H. Han, H. J. Kim, E. J. Kim, K. S. Kim, S. Hong et al., RORa decreases oxidative stress through the induction of SOD2 and GPx1 expression and thereby protects against nonalcoholic steatohepatitis in mice, Antioxidants and Redox Signaling, vol.21, issue.15, pp.2083-2094, 2014.

H. J. Kim, Y. H. Han, H. Na, J. Y. Kim, T. Kim et al., Liverspecific deletion of RORa aggravates diet-induced nonalcoholic steatohepatitis by inducing mitochondrial dysfunction, Scientific Reports, vol.7, issue.1, p.16041, 2017.
DOI : 10.1038/s41598-017-16077-y

URL : https://www.nature.com/articles/s41598-017-16077-y.pdf

K. Kim, K. Boo, Y. S. Yu, S. K. Oh, H. Kim et al., RORa controls hepatic lipid homeostasis via negative regulation of PPARg transcriptional network, Nature Communications, vol.8, issue.1, p.162, 2017.

A. Molinaro, R. Caesar, L. M. Holm, V. Tremaroli, P. D. Cani et al., Host-microbiota interaction induces bi-phasic inflammation and glucose intolerance in mice, Molecular Metabolism, vol.6, issue.11, pp.1371-1380, 2017.
DOI : 10.1016/j.molmet.2017.08.016

URL : https://doi.org/10.1016/j.molmet.2017.08.016

R. Homan and M. K. Anderson, Rapid separation and quantitation of combined neutral and polar lipid classes by high-performance liquid chromatography and evaporative light-scattering mass detection, Journal of Chromatography B Biomedical Sciences and Applications, vol.708, issue.1e2, pp.21-26, 1998.
DOI : 10.1016/s0378-4347(97)00651-8

L. Löfgren, G. B. Forsberg, and M. Ståhlman, The BUME method: a new rapid and simple chloroform-free method for total lipid extraction of animal tissue, Scientific Reports, vol.6, p.27688, 2016.

W. Zhang, R. M. Sargis, P. A. Volden, C. M. Carmean, X. J. Sun et al., PCB 126 and other dioxin-like PCBs specifically suppress hepatic PEPCK expression via the aryl hydrocarbon receptor, PLoS One, vol.7, issue.5, p.37103, 2012.
DOI : 10.1371/journal.pone.0037103

URL : https://journals.plos.org/plosone/article/file?id=10.1371/journal.pone.0037103&type=printable

A. Koh, M. N. Lee, Y. R. Yang, H. Jeong, J. Ghim et al., C1-Ten is a protein tyrosine phosphatase of insulin receptor substrate 1 (IRS-1), regulating IRS-1 stability and muscle atrophy, Molecular and Cellular Biology, vol.33, issue.8, pp.1608-1620, 2013.

T. Wada, H. S. Kang, M. Angers, H. Gong, S. Bhatia et al., Identification of oxysterol 7alpha-hydroxylase (Cyp7b1) as a novel retinoid-related orphan receptor alpha (RORalpha) (NR1F1) target gene and a functional cross-talk between RORalpha and liver X receptor (NR1H3), Molecular Pharmacology, vol.73, issue.3, pp.891-899, 2008.

P. Delerive, W. W. Chin, and C. S. Suen, Identification of Reverb(alpha) as a novel ROR(alpha) target gene, Journal of Biological Chemistry, vol.277, issue.38, pp.35013-35018, 2002.

R. C. Bortolin, A. R. Vargas, J. Gasparotto, P. R. Chaves, C. E. Schnorr et al., A new animal diet based on human Western diet is a robust diet-induced obesity model: comparison to high-fat and cafeteria diets in term of metabolic and gut microbiota disruption, International Journal of Obesity (Lond), vol.42, issue.3, pp.525-534, 2017.

C. K. Fleissner, N. Huebel, A. El-bary, M. M. Loh, G. Klaus et al., Absence of intestinal microbiota does not protect mice from dietinduced obesity, British Journal of Nutrition, vol.104, issue.6, pp.919-929, 2010.

Y. Zhang, R. Papazyan, M. Damle, B. Fang, J. Jager et al., The hepatic circadian clock fine-tunes the lipogenic response to feeding through RORa/g, Genes & Development, vol.31, issue.12, pp.1202-1211, 2017.

X. Zhao, H. Cho, R. T. Yu, A. R. Atkins, M. Downes et al., Nuclear receptors rock around the clock, EMBO Reports, vol.15, issue.5, pp.518-528, 2014.
DOI : 10.1002/embr.201338271

URL : http://embor.embopress.org/content/15/5/518.full.pdf