L. Micallef and P. Rodgers, eulerAPE: Drawing Area-Proportional 3-Venn Diagrams Using Ellipses, PLoS ONE, vol.29, issue.7, p.101717, 2014.
DOI : 10.1371/journal.pone.0101717.g013

T. Ly, J. Wang, R. Pereira, K. Rojas, and X. Peng, Activation of the Ran GTPase Is Subject to Growth Factor Regulation and Can Give Rise to Cellular Transformation, Journal of Biological Chemistry, vol.285, issue.8, pp.5815-5826, 2010.
DOI : 10.1074/jbc.M109.071886

J. Seok, H. Warren, A. Cuenca, M. Mindrinos, and H. Baker, Genomic responses in mouse models poorly mimic human inflammatory diseases, Proceedings of the National Academy of Sciences, vol.110, issue.9, pp.3507-3512, 2007.
DOI : 10.1073/pnas.1222878110

P. Jennings, A. Limonciel, L. Felice, M. Leonard, J. Lamba et al., An overview of transcriptional regulation in response to toxicological insult Genetic contribution to variable human CYP3A-mediated metabolism, Arch Toxicol Adv Drug Deliv Rev, vol.87, issue.54, pp.49-721271, 2002.

J. Slatter, I. Templeton, J. Castle, A. Kulkarni, and T. Rushmore, Compendium of gene expression profiles comprising a baseline model of the human liver drug metabolism transcriptome, Xenobiotica, vol.35, issue.2, pp.938-962, 2006.
DOI : 10.1080/00498250600861728

J. Villeneuve and V. Pichette, Cytochrome P450 and Liver Diseases, Current Drug Metabolism, vol.5, issue.3, pp.273-282, 2004.
DOI : 10.2174/1389200043335531

L. Yang, E. Price, C. Chang, Y. Li, and Y. Huang, Gene expression variability in human hepatic drug metabolizing enzymes and transporters Expression profiling of interindividual variability following xenobiotic exposures in primary human hepatocyte cultures, PLoS One Toxicol Appl Pharmacol, vol.8, issue.231, pp.216-224, 2008.

L. Guo, S. Dial, L. Shi, W. Branham, and J. Liu, Similarities and Differences in the Expression of Drug-Metabolizing Enzymes between Human Hepatic Cell Lines and Primary Human Hepatocytes, Drug Metabolism and Disposition, vol.39, issue.3, pp.528-538, 2011.
DOI : 10.1124/dmd.110.035873

A. Sjogren, M. Liljevald, B. Glinghammar, J. Sagemark, and X. Li, Critical differences in toxicity mechanisms in induced pluripotent stem cell-derived hepatocytes, hepatic cell lines and primary hepatocytes, Archives of Toxicology, vol.124, issue.2, pp.1427-1437, 2014.
DOI : 10.1007/s00204-014-1265-z

J. Vrba, M. Havlikova, D. Gerhardova, and J. Ulrichova, Palmatine activates AhR and upregulates CYP1A activity in HepG2 cells but not in human hepatocytes, Toxicology in Vitro, vol.28, issue.4, pp.693-699, 2014.
DOI : 10.1016/j.tiv.2014.02.008

S. Kim, E. Dere, L. Burgoon, C. Chang, and T. Zacharewski, Comparative Analysis of AhR-Mediated TCDD-Elicited Gene Expression in Human Liver Adult Stem Cells, Toxicological Sciences, vol.112, issue.1, pp.229-244, 2009.
DOI : 10.1093/toxsci/kfp189

W. Kim, Y. In, J. Kim, H. Cho, and J. Kim, Quantitative relationship of dioxin-responsive gene expression to dioxin response element in Hep3B and HepG2 human hepatocarcinoma cell lines, Toxicology Letters, vol.165, issue.2, pp.174-181, 2006.
DOI : 10.1016/j.toxlet.2006.03.007

S. Safe and M. Wormke, Inhibitory Aryl Hydrocarbon Receptor???Estrogen Receptor ?? Cross-Talk and Mechanisms of Action, Chemical Research in Toxicology, vol.16, issue.7, pp.807-816, 2003.
DOI : 10.1021/tx034036r

E. Swedenborg and I. Pongratz, AhR and ARNT modulate ER signaling, Toxicology, vol.268, issue.3, pp.132-138, 2010.
DOI : 10.1016/j.tox.2009.09.007

M. Marques, L. Laflamme, and L. Gaudreau, Estrogen receptor ?? can selectively repress dioxin receptor-mediated gene expression by targeting DNA methylation, Nucleic Acids Research, vol.41, issue.17, pp.8094-8102, 2013.
DOI : 10.1093/nar/gkt595

S. Mutka, L. Green, E. Verderber, J. Richards, and D. Looker, ADH IB Expression, but Not ADH III, Is Decreased in Human Lung Cancer, PLoS ONE, vol.16, issue.12, pp.52995-88, 2012.
DOI : 10.1371/journal.pone.0052995.t001

R. Wei, M. Zhang, H. Rao, H. Pu, and H. Zhang, Identification of ADH4 as a novel and potential prognostic marker in hepatocellular carcinoma, Medical Oncology, vol.23, issue.3???4, pp.2737-2743, 2012.
DOI : 10.1007/s12032-011-0126-3

C. Nilsson and H. Hakansson, The Retinoid Signaling System ??? A Target in Dioxin Toxicity, Critical Reviews in Toxicology, vol.28, issue.3, pp.211-232, 2002.
DOI : 10.1038/330444a0

J. Novak, M. Benisek, and K. Hilscherova, Disruption of retinoid transport, metabolism and signaling by environmental pollutants, Environment International, vol.34, issue.6, pp.898-913, 2008.
DOI : 10.1016/j.envint.2007.12.024

N. Fletcher, A. Hanberg, and H. Hakansson, Hepatic Vitamin A Depletion Is a Sensitive Marker of 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) Exposure in Four Rodent Species, Toxicological Sciences, vol.62, issue.1, pp.166-175, 2001.
DOI : 10.1093/toxsci/62.1.166

N. Fletcher, D. Wahlstrom, R. Lundberg, C. Nilsson, and K. Nilsson, 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) alters the mRNA expression of critical genes associated with cholesterol metabolism, bile acid biosynthesis, and bile transport in rat liver: A microarray study, Toxicology and Applied Pharmacology, vol.207, issue.1, pp.1-24, 2005.
DOI : 10.1016/j.taap.2004.12.003

K. Su, N. Sabeva, J. Liu, Y. Wang, and S. Bhatnagar, The ABCG5 ABCG8 Sterol Transporter Opposes the Development of Fatty Liver Disease and Loss of Glycemic Control Independently of Phytosterol Accumulation, Journal of Biological Chemistry, vol.287, issue.34, pp.28564-28575, 2012.
DOI : 10.1074/jbc.M112.360081

P. Lefebvre, B. Cariou, L. F. Kuipers, F. Staels, and B. , Role of Bile Acids and Bile Acid Receptors in Metabolic Regulation, Physiological Reviews, vol.89, issue.1, pp.147-191, 2009.
DOI : 10.1152/physrev.00010.2008

S. Kodama, R. Moore, Y. Yamamoto, and M. Negishi, Human nuclear pregnane X receptor cross-talk with CREB to repress cAMP activation of the glucose-6-phosphatase gene, Biochemical Journal, vol.407, issue.3, pp.373-381, 2007.
DOI : 10.1042/BJ20070481

S. Diani-moore, R. P. Li, X. Mondal, P. Youn, and D. , Identification of the Aryl Hydrocarbon Receptor Target Gene TiPARP as a Mediator of Suppression of Hepatic Gluconeogenesis by 2,3,7,8-Tetrachlorodibenzo-p-dioxin and of Nicotinamide as a Corrective Agent for This Effect, Journal of Biological Chemistry, vol.285, issue.50, pp.38801-38810, 2010.
DOI : 10.1074/jbc.M110.131573

A. Silverstone, P. Rosenbaum, R. Weinstock, S. Bartell, and H. Foushee, Polychlorinated Biphenyl (PCB) Exposure and Diabetes: Results from the Anniston Community Health Survey, Environmental Health Perspectives, vol.120, issue.5, pp.727-732, 2012.
DOI : 10.1289/ehp.1104247

R. Clapp, M. Jacobs, and E. Loechler, Environmental and Occupational Causes of Cancer: New Evidence 2005-2007, Reviews on Environmental Health, vol.23, issue.1, pp.1-37, 2008.
DOI : 10.1515/REVEH.2008.23.1.1

D. Consonni, A. Pesatori, C. Zocchetti, R. Sindaco, D. Oro et al., Mortality in a Population Exposed to Dioxin after the Seveso, Italy, Accident in 1976: 25 Years of Follow-Up, American Journal of Epidemiology, vol.167, issue.7, pp.847-858, 2008.
DOI : 10.1093/aje/kwm371

H. Inadera, Developmental origins of obesity and type 2 diabetes: molecular aspects and role of chemicals, Environmental Health and Preventive Medicine, vol.304, issue.3, pp.185-197, 2013.
DOI : 10.1016/j.mce.2009.02.021

P. Mann, Selected Lesions of Dioxin in Laboratory Rodents, Toxicologic Pathology, vol.98, issue.4, pp.72-79, 1997.
DOI : 10.1177/019262339702500114

F. El-sabeawy, E. Enan, and B. Lasley, Biochemical and toxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin in immature male and female chickens, Comparative Biochemistry and Physiology Part C: Toxicology & Pharmacology, vol.129, issue.4, pp.317-327, 2001.
DOI : 10.1016/S1532-0456(01)00199-5

J. Zodrow, J. Stegeman, and R. Tanguay, Histological analysis of acute toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in zebrafish, Aquatic Toxicology, vol.66, issue.1, pp.25-38, 2004.
DOI : 10.1016/j.aquatox.2003.07.002

B. Wahlang, J. Beier, H. Clair, H. Bellis-jones, and K. Falkner, Toxicant-associated Steatohepatitis, Toxicologic Pathology, vol.66, issue.2, pp.343-360, 2013.
DOI : 10.1111/j.1365-2036.2011.04724.x

K. Taylor, R. Novak, H. Anderson, L. Birnbaum, and C. Blystone, Evaluation of the Association between Persistent Organic Pollutants (POPs) and Diabetes in Epidemiological Studies: A National Toxicology Program Workshop Review, Environmental Health Perspectives, vol.121, issue.7, pp.774-783, 2013.
DOI : 10.1289/ehp.1205502

D. Tata and V. , Association of Dioxin and Other Persistent Organic Pollutants (POPs) with Diabetes: Epidemiological Evidence and New Mechanisms of Beta Cell Dysfunction, International Journal of Molecular Sciences, vol.15, issue.5, pp.7787-7811, 2014.
DOI : 10.3390/ijms15057787