P. Alonso-magdalena, E. Vieira, S. Soriano, L. Menes, D. Burks et al., Bisphenol A exposure during pregnancy disrupts glucose homeostasis in mothers and adult male offspring, Environ. Health Perspect, vol.118, pp.1243-1250, 2010.

L. B. Barber, J. E. Loyo-rosales, C. P. Rice, T. A. Minarik, and A. K. Oskouie, Endocrine disrupting alkylphenolic chemicals and other contaminants in wastewater treatment plant effluents, urban streams, and fish in the Great Lakes and Upper Mississippi River Regions, Sci. Total Environ, vol.517, pp.195-206, 2015.

L. P. Bechmann, R. A. Hannivoort, G. Gerken, G. S. Hotamisligil, M. Trauner et al., The interaction of hepatic lipid and glucose metabolism in liver diseases, J. Hepatol, vol.56, pp.952-964, 2012.

P. Bedossa, Histological Assessment of NAFLD, Dig. Dis. Sci, vol.61, pp.1348-1355, 2016.

B. D. Blair, J. P. Crago, C. J. Hedman, and R. D. Klaper, Pharmaceuticals and personal care products found in the Great Lakes above concentrations of environmental concern, Chemosphere, vol.93, pp.2116-2123, 2013.

A. R. Blaustein and B. A. Bancroft, Amphibian Population Declines: Evolutionary Considerations, BioScience, vol.57, pp.437-444, 2007.

A. M. Bolger, M. Lohse, and B. Usadel, Trimmomatic: a flexible trimmer for Illumina sequence data, Bioinforma. Oxf. Engl, vol.30, pp.2114-2120, 2014.

S. Brooks, C. R. Tyler, and J. P. Sumpter, Egg quality in fish: what makes a good egg?, Rev. Fish Biol. Fish, vol.7, pp.387-416, 1997.

R. K. Browne, Reproduction and larval rearing of amphibians, ILAR Journal/National Research Council, pp.214-234, 2007.

A. Bulteau, M. Ikeda-saito, and L. I. Szweda, Redox-Dependent Modulation of Aconitase Activity in Intact Mitochondria, Biochemistry, vol.42, pp.14846-14855, 2003.

T. K. Felder, H. Oberkofler, R. Weitgasser, V. Mackevics, F. Krempler et al., The SREBF-1 locus is associated with type 2 diabetes and plasma adiponectin levels in a middle-aged Austrian population, Int. J. Obes, vol.31, pp.1099-1103, 2007.

P. Ferré and F. Foufelle, Hepatic steatosis: a role for de novo lipogenesis and the transcription factor SREBP-1c, Diabetes Obes. Metab, vol.12, pp.83-92, 2010.

J. M. Galloway, B. E. Haggard, M. T. Meyers, and W. R. Green, Occurrence of pharmaceuticals and other organic wastewater constituents in selected streams in northern Arkansas, USGS Numbered Series No. 2005-5140), Scientific Investigations Report, 2004.

B. Garait, K. Couturier, S. Servais, D. Letexier, D. Perrin et al., Fat intake reverses the beneficial effects of low caloric intake on skeletal muscle mitochondrial H(2)O(2) production. Free Radic, Biol. Med, vol.39, pp.1249-1261, 2005.
URL : https://hal.archives-ouvertes.fr/inserm-00388735

M. Gardner, S. Comber, M. D. Scrimshaw, E. Cartmell, J. Lester et al., The significance of hazardous chemicals in wastewater treatment works effluents, Sci. Total Environ, vol.437, pp.363-372, 2012.

A. A. Godoy and F. Kummrow, What do we know about the ecotoxicology of pharmaceutical and personal care product mixtures? A critical review, Crit. Rev. Environ. Sci. Technol, vol.47, pp.1453-1496, 2017.

F. Grün, H. Watanabe, Z. Zamanian, L. Maeda, K. Arima et al., Endocrine-disrupting organotin compounds are potent inducers of adipogenesis in vertebrates, Mol. Endocrinol, vol.20, pp.2141-2155, 2006.

Y. Guan, J. Gao, Y. Zhang, S. Chen, C. Yuan et al., Effects of bisphenol A on lipid metabolism in rare minnow Gobiocypris rarus, Comp. Biochem. Physiol. C, vol.179, pp.144-149, 2016.

X. Han, C. Liu, Y. Xue, J. Wang, C. Xue et al., Long-term fatty liver-induced insulin resistance in orotic acid-induced nonalcoholic fatty liver rats, 2016.

, Biosci. Biotechnol. Biochem, vol.80, pp.735-743

T. Hayes, K. Haston, M. Tsui, A. Hoang, C. Haeffele et al., Herbicides: Feminization of male frogs in the wild, Nature, vol.419, pp.895-896, 2002.

T. B. Hayes, L. L. Anderson, V. R. Beasley, S. R. De-solla, T. Iguchi et al., Demasculinization and feminization of male gonads by atrazine: Consistent effects across vertebrate classes, J. Steroid Biochem. Mol. Biol, vol.127, pp.64-73, 2011.

T. B. Hayes, P. Case, S. Chui, D. Chung, C. Haeffele et al., Pesticide Mixtures, Endocrine Disruption, and Amphibian Declines: Are We Underestimating the Impact?, Environ. Health Perspect, vol.114, pp.40-50, 2006.

T. B. Hayes, P. Falso, S. Gallipeau, and M. Stice, The cause of global amphibian declines: a developmental endocrinologist's perspective, J. Exp. Biol, vol.213, pp.921-933, 2010.

M. Hua, H. Su, T. Yao, M. Kuo, M. Lai et al., Alternation of plasma fatty acids composition and desaturase activities in children with liver steatosis, PloS One, vol.12, 2017.

D. W. Huang, B. T. Sherman, and R. A. Lempicki, Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists, Nucleic Acids Res, vol.37, pp.1-13, 2009.

Y. Jiang, W. Xia, Y. Zhu, X. Li, D. Wang et al., Mitochondrial dysfunction in early life resulted from perinatal bisphenol A exposure contributes to hepatic steatosis in rat offspring, Toxicol. Lett, vol.228, pp.85-92, 2014.

M. Kaplan, Amphibians rarely give earliest warning of pollution, Nat. News, 2009.

A. Kashiwagi, K. Utsumi, K. Kashiwagi, S. Ohta, K. Sugihara et al., Effects of Endocrine Disrupting Chemicals on Amphibian Metamorphosis and Mitochondrial Membrane Permeability Transition, J. Health Sci, vol.54, pp.273-280, 2008.

M. Kasuga, Insulin resistance and pancreatic ? cell failure, J. Clin. Invest, vol.116, pp.1756-1760, 2006.

W. Kloas, R. Urbatzka, R. Opitz, S. Würtz, T. Behrends et al., Endocrine Disruption in Aquatic Vertebrates, Ann. N. Y. Acad. Sci, vol.1163, pp.187-200, 2009.

C. Koliaki, J. Szendroedi, K. Kaul, T. Jelenik, P. Nowotny et al., Adaptation of Hepatic Mitochondrial Function in Humans with Non-Alcoholic Fatty Liver Is Lost in Steatohepatitis, Cell Metab, vol.21, pp.739-746, 2015.

M. Kuramoto, Correlations of quantitative parameters of fecundity in amphibians, Evolution, vol.32, pp.287-296, 1978.

V. S. Langlois, P. Duarte-guterman, S. Ing, B. D. Pauli, G. M. Cooke et al., Fadrozole and finasteride exposures modulate sex steroid-and thyroid hormone-related gene expression in Silurana (Xenopus) tropicalis early larval development, Gen. Comp. Endocrinol, vol.166, pp.417-427, 2010.

C. W. Levy, A. Roujeinikova, S. Sedelnikova, P. J. Baker, A. R. Stuitje et al., Molecular basis of triclosan activity, Nature, vol.398, pp.383-384, 1999.

A. Lindström, I. J. Buerge, T. Poiger, P. Bergqvist, M. D. Müller et al., Occurrence and environmental behavior of the bactericide triclosan and its methyl derivative in surface waters and in wastewater, Environ. Sci. Technol, vol.36, pp.2322-2329, 2002.

D. C. Mcavoy, B. Schatowitz, M. Jacob, A. Hauk, and W. S. Eckhoff, Measurement of triclosan in wastewater treatment systems, Environ. Toxicol. Chem, vol.21, pp.1323-1329, 2009.

V. Miksztowicz, D. Lucero, V. Zago, L. Cacciagiú, G. Lopez et al., Hepatic lipase activity is increased in nonalcoholic fatty liver disease beyond insulin resistance, Diabetes Metab. Res. Rev, vol.28, pp.535-541, 2012.

T. Møretrø, G. S. Høiby-pettersen, O. Habimana, E. Heir, and S. Langsrud, Assessment of the antibacterial activity of a triclosan-containing cutting board, Int. J. Food Microbiol, vol.146, pp.157-162, 2011.

G. Mosconi, O. Carnevali, M. F. Franzoni, E. Cottone, I. Lutz et al., Environmental Estrogens and Reproductive Biology in Amphibians, Gen. Comp. Endocrinol, vol.126, pp.125-129, 2002.

B. J. Moyer, I. Y. Rojas, J. S. Kerley-hamilton, H. F. Hazlett, K. V. Nemani et al., Inhibition of the aryl hydrocarbon receptor prevents Western diet-induced obesity. Model for AHR activation by kynurenine via oxidized-LDL, TLR2/4, TGF?, and IDO1, Toxicol. Appl. Pharmacol, vol.300, pp.13-24, 2016.

T. Mukai, M. Egawa, T. Takeuchi, H. Yamashita, and T. Kusudo, Silencing of FABP1 ameliorates hepatic steatosis, inflammation, and oxidative stress in mice with nonalcoholic fatty liver disease, FEBS Open Bio, vol.7, pp.1009-1016, 2017.

A. Naik, R. Ko?ir, and D. Rozman, Genomic aspects of NAFLD pathogenesis, Genomics, SI:Clinical and Translational Genomics, vol.102, pp.84-95, 2013.

F. Neuschäfer-rube, A. Schraplau, B. Schewe, S. Lieske, J. Krützfeldt et al., Arylhydrocarbon receptor-dependent mIndy (Slc13a5) induction as possible contributor to benzo[a]pyrene-induced lipid accumulation in hepatocytes, Toxicology, vol.337, pp.1-9, 2015.

H. F. Olivares-rubio, R. Dzul-caamal, M. E. Gallegos-rangel, R. L. Madera-sandoval, M. L. Domínguez-lópez et al., Relationship between biomarkers and endocrine-disrupting compounds in wild Girardnichthys viviparus from two lakes with different degrees of pollution, Ecotoxicol. Lond. Engl, vol.24, pp.664-685, 2015.

T. Otoda, T. Takamura, H. Misu, T. Ota, S. Murata et al., Proteasome dysfunction mediates obesity-induced endoplasmic reticulum stress and insulin resistance in the liver, Diabetes, vol.62, pp.811-824, 2013.

A. M. Petersen and T. T. Gleeson, Characterization of circannual patterns of metabolic recovery from activity in Rana catesbeiana at 15°C, J. Exp. Biol, vol.210, pp.1786-1797, 2007.

K. F. Petersen, S. Dufour, D. B. Savage, S. Bilz, G. Solomon et al., The role of skeletal muscle insulin resistance in the pathogenesis of the metabolic syndrome, Proc. Natl. Acad. Sci. U. S. A, vol.104, pp.12587-12594, 2007.

M. W. Pfaffl, A new mathematical model for relative quantification in real-time RT-PCR, Nucleic Acids Res, vol.29, p.45, 2001.

A. Piccoli, J. Fiori, V. Andrisano, and M. Orioli, Determination of triclosan in personal health care products by liquid chromatography (HPLC). Il Farm, vol.57, pp.369-372, 2002.

G. Ramakrishnan, A. Arjuman, S. Suneja, C. Das, and N. C. Chandra, The association between insulin and low-density lipoprotein receptors, Diab. Vasc. Dis. Res, vol.9, pp.196-204, 2012.

C. Regnault, M. Usal, S. Veyrenc, K. Couturier, C. Batandier et al., Unexpected metabolic disorders induced by endocrine disruptors in Xenopus tropicalis provide new lead for understanding amphibian decline, Proc. Natl. Acad. Sci. U, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01833911

S. , , vol.115, pp.4416-4425

C. Regnault, J. Willison, S. Veyrenc, A. Airieau, P. Méresse et al., Metabolic and immune impairments induced by the endocrine disruptors benzo[a]pyrene and triclosan in Xenopus tropicalis, Chemosphere, vol.155, pp.519-527, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01351542

C. Regnault, I. A. Worms, C. Oger-desfeux, C. Melodelima, S. Veyrenc et al., Impaired liver function in Xenopus tropicalis exposed to benzo[a]pyrene: transcriptomic and metabolic evidence, BMC Genomics, vol.15, 2014.

R. A. Relyea, A cocktail of contaminants: how mixtures of pesticides at low concentrations affect aquatic communities, Oecologia, vol.159, pp.363-376, 2009.

K. Roelants, D. J. Gower, M. Wilkinson, S. P. Loader, S. D. Biju et al., Global patterns of diversification in the history of modern amphibians, Proc. Natl. Acad. Sci, vol.104, pp.887-892, 2007.

S. Rousset, M. Alves-guerra, J. Mozo, B. Miroux, A. Cassard-doulcier et al., The biology of mitochondrial uncoupling proteins, Diabetes, vol.53, issue.1, pp.130-135, 2004.

C. T. Rumrill, D. E. Scott, and S. L. Lance, Delayed effects and complex life cycles: How the larval aquatic environment influences terrestrial performance and survival, Environ. Toxicol. Chem, vol.37, pp.2660-2669, 2018.

A. I. Saeed, V. Sharov, J. White, J. Li, W. Liang et al., TM4: a free, open-source system for microarray data management and analysis, BioTechniques, vol.34, pp.374-378, 2003.

S. Santangeli, V. Notarstefano, F. Maradonna, E. Giorgini, G. Gioacchini et al., Effects od diethylene glycol dibenzoate and Bisphenol A on the lipid metabolism of Danio rerio, Sci. Total Environ, vol.636, pp.641-655, 2018.

H. Shi, P. Zhu, and S. Guo, Effects of tributyltin on metamorphosis and gonadal differentiation of Xenopus laevis at environmentally relevant concentrations, Toxicol. Ind. Health, vol.30, pp.297-303, 2014.

C. Showell and F. L. Conlon, Natural Mating and Tadpole Husbandry in the Western Clawed Frog Xenopus tropicalis, Cold Spring Harb. Protoc, p.5292, 2009.

D. C. Smith, Adult Recruitment in Chorus Frogs: Effects of Size and Date at Metamorphosis, Ecology, vol.68, pp.344-350, 1987.

K. Srogi, Monitoring of environmental exposure to polycyclic aromatic hydrocarbons: a review, Environ. Chem. Lett, vol.5, pp.169-195, 2007.

M. Susiarjo, F. Xin, A. Bansal, M. Stefaniak, C. Li et al., Bisphenol a exposure disrupts metabolic health across multiple generations in the mouse, Endocrinology, vol.156, pp.2049-2058, 2015.

P. A. Srere, Citrate synthase, Methods Enzymol, vol.13, pp.3-5, 1969.

Y. Takahashi and T. Fukusato, Histopathology of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis, World J. Gastroenterol, vol.20, pp.15539-15548, 2014.

D. Teubner, M. Paulus, M. Veith, and R. Klein, Biometric parameters of the bream (Abramis brama) as indicators for long-term changes in fish health and environmental quality--data from the German ESB, Environ. Sci. Pollut. Res. Int, vol.22, pp.1620-1627, 2015.

B. D. Todd, C. M. Bergeron, M. J. Hepner, and W. A. Hopkins, Aquatic and terrestrial stressors in amphibians: a test of the double jeopardy hypothesis based on maternally and trophically derived contaminants, Environ. Toxicol. Chem, vol.30, pp.2277-2284, 2011.

M. Trapido and I. Veldre, On polynuclear aromatic hydrocarbons contamination levels in the ecosystem of Lake Peipsi in the 1970s-1980s, Hydrobiologia, vol.338, pp.185-190, 1996.

C. Trapnell, L. Pachter, and S. L. Salzberg, TopHat: discovering splice junctions with RNA-Seq, Bioinforma. Oxf. Engl, vol.25, pp.1105-1111, 2009.

C. Trapnell, A. Roberts, L. Goff, G. Pertea, D. Kim et al., Differential gene and transcript expression analysis of RNAseq experiments with TopHat and Cufflinks, Nat. Protoc, vol.7, pp.562-578, 2012.

F. Ucar, S. Sezer, S. Erdogan, S. Akyol, F. Armutcu et al., The relationship between oxidative stress and nonalcoholic fatty liver disease: Its effects on the development of nonalcoholic steatohepatitis, Redox Rep, vol.18, pp.127-133, 2013.

, Endocrine Disruptor Screening Program: Second List of Chemicals for Tier 1 Screening, 2010.

R. Urbatzka, S. Bottero, A. Mandich, I. Lutz, and W. Kloas, Endocrine disrupters with (anti)estrogenic and (anti)androgenic modes of action affecting reproductive biology of Xenopus laevis: I. Effects on sex steroid levels and biomarker expression, 2007.

, Biochem. Physiol. Part C Toxicol. Pharmacol, vol.144, pp.310-318

R. Urbatzka, I. Lutz, R. Opitz, and W. Kloas, Luteinizing hormone, follicle stimulating hormone, and gonadotropin releasing hormone mRNA expression of Xenopus laevis in response to endocrine disrupting compounds affecting reproductive biology, Gen. Comp. Endocrinol, vol.146, pp.119-125, 2006.

M. N. Vansaun, A. M. Mendonsa, and D. L. Gorden, Hepatocellular Proliferation Correlates with Inflammatory Cell and Cytokine Changes in a Murine Model of Nonalchoholic Fatty Liver Disease, PLOS ONE, vol.8, p.73054, 2013.

N. Veldhoen, R. C. Skirrow, H. Osachoff, H. Wigmore, D. J. Clapson et al., The bactericidal agent triclosan modulates thyroid hormone-associated gene expression and disrupts postembryonic anuran development, 2006.

, Aquat. Toxicol, vol.80, pp.217-227

G. Vial, H. Dubouchaud, K. Couturier, C. Cottet-rousselle, N. Taleux et al., Effects of a high-fat diet on energy metabolism and ROS production in rat liver, J. Hepatol, vol.54, pp.348-356, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00628497

Q. Wang, L. Jiang, J. Wang, S. Li, Y. Yu et al., Abrogation of hepatic ATP-citrate lyase protects against fatty liver and ameliorates hyperglycemia in leptin receptor-deficient mice, Hepatol. Baltim. Md, vol.49, pp.1166-1175, 2009.

A. Weir, W. E. Moiles, B. Brockman, C. S. Mattick, K. Mcclellan et al., Concentrations of Hydrophobic Organic Pollutants in U.S. Wastewater Treatment Plants and in Receiving Surface Waters Modeled from EPA Biosolids Monitoring Data, Contaminants of Emerging Concern in the Environment: Ecological and Human Health Considerations, pp.421-436, 2010.

D. M. Whitacre, Reviews of Environmental Contamination and Toxicology, vol.206, 2010.

K. Yamaguchi, L. Yang, S. Mccall, J. Huang, X. X. Yu et al., Inhibiting triglyceride synthesis improves hepatic steatosis but exacerbates liver damage and fibrosis in obese mice with nonalcoholic steatohepatitis, Hepatol. Baltim. Md, vol.45, pp.1366-1374, 2007.

Y. Yuan, L. Zhao, Y. Chen, J. F. Moorhead, Z. Varghese et al., Advanced glycation end products (AGEs) increase human mesangial foam cell formation by increasing Golgi SCAP glycosylation in vitro, Am. J. Physiol, 2011.

, Physiol, vol.301, pp.236-243