T. Suzuki and M. Yamamoto, Stress-sensing mechanisms and the physiological roles of the Keap1???Nrf2 system during cellular stress, Journal of Biological Chemistry, vol.17, issue.41, 2017.
DOI : 10.1126/scisignal.aam5711

M. Xue, H. Momiji, N. Rabbani, G. Barker, T. Bretschneider et al., Frequency Modulated Translocational Oscillations of Nrf2 Mediate the Antioxidant Response Element Cytoprotective Transcriptional Response, Antioxidants & Redox Signaling, vol.23, issue.7, pp.613-629, 2014.
DOI : 10.1089/ars.2014.5962

J. D. Hayes and A. T. Dinkova-kostova, The Nrf2 regulatory network provides an interface between redox and intermediary metabolism, Trends in Biochemical Sciences, vol.39, issue.4, 2014.
DOI : 10.1016/j.tibs.2014.02.002

M. Baradat, I. Jouanin, S. Dalleau, S. Tache, M. Gieules et al., (E)-nonenal metabolism differs in Apc(+/+) cells and in Apc(Min/+) cells: it may explain colon cancer promotion by heme iron, Chem Res Toxicol, vol.24, pp.4-6, 2011.

S. Dalleau, M. Baradat, F. Gueraud, and L. Huc, Cell death and diseases related to oxidative stress:4-hydroxynonenal (HNE) in the balance, Cell Death & Differentiation, vol.14, issue.132
DOI : 10.1016/j.devcel.2007.12.002

R. Surya, C. Héliès-toussaint, O. C. Martin, T. Gauthier, F. Guéraud et al., Red meat and colorectal cancer: Nrf2-dependent antioxidant response contributes to the resistance of preneoplastic colon cells to fecal water of hemoglobin- and beef-fed rats, Carcinogenesis, vol.133, issue.6, pp.37-635, 2016.
DOI : 10.1021/ja407407h

V. Calabrese, C. Cornelius, A. T. Dinkova-kostova, E. J. Calabrese, and M. P. Mattson, Cellular Stress Responses, The Hormesis Paradigm, and Vitagenes: Novel Targets for Therapeutic Intervention in Neurodegenerative Disorders, Antioxidants & Redox Signaling, vol.13, issue.11, pp.1763-1811, 2010.
DOI : 10.1089/ars.2009.3074

H. K. Bayele, E. S. Debnam, and K. S. Srai, Nrf2 transcriptional derepression from Keap1 by dietary polyphenols, Biochemical and Biophysical Research Communications, vol.469, issue.3, 2016.
DOI : 10.1016/j.bbrc.2015.11.103

C. Köhle and K. W. Bock, Coordinate regulation of Phase I and II xenobiotic metabolisms by the Ah receptor and Nrf2, Biochemical Pharmacology, vol.73, issue.12, 2007.
DOI : 10.1016/j.bcp.2007.01.009

H. Dong, R. R. Bonala, N. Suzuki, F. Johnson, A. P. Grollman et al., Mutagenic potential of benzo[a]pyrene-derived DNA adducts positioned in codon 273 of the human P53 gene, Biochemistry (Mosc, pp.43-15922, 1021.

T. D. Phillips, M. Richardson, Y. L. Cheng, L. He, T. J. Mcdonald et al., Mechanistic relationships between hepatic genotoxicity and carcinogenicity in male B6C3F1 mice treated with polycyclic aromatic hydrocarbon mixtures, Archives of Toxicology, vol.249, issue.3, pp.967-977, 2015.
DOI : 10.1016/j.taap.2010.09.019

S. Yi, J. Hong, H. Ohrr, and J. Yi, Agent Orange exposure and disease prevalence in Korean Vietnam veterans: The Korean veterans health study, Environmental Research, vol.133, pp.56-65, 2014.
DOI : 10.1016/j.envres.2014.04.027

D. N. Das, P. P. Naik, S. Mukhopadhyay, P. K. Panda, N. Sinha et al., Elimination of dysfunctional mitochondria through mitophagy suppresses benzo[a]pyrene-induced apoptosis, Free Radic, Biol. Med, vol.112, pp.452-463, 2017.

S. Lin, Z. Yang, H. Liu, and Z. Cai, Metabolomic analysis of liver and skeletal muscle tissues in C57BL/6J and DBA/2J mice exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin, Molecular BioSystems, vol.205, issue.83, pp.1956-1965, 2011.
DOI : 10.1016/j.taap.2004.09.014

R. Nault, K. A. Fader, T. A. Lydic, and T. R. Zacharewski, -dioxin, Chemical Research in Toxicology, vol.30, issue.4, pp.1060-1075, 2017.
DOI : 10.1021/acs.chemrestox.6b00430

R. B. Robey, J. Weisz, N. Kuemmerle, A. C. Salzberg, A. Berg et al., Metabolic reprogramming and dysregulated metabolism: cause, consequence and/or enabler of environmental carcinogenesis?, Carcinogenesis, vol.124, issue.1, p.36, 2015.
DOI : 10.1093/jnci/90.1.67

A. Ambolet-camoit, C. Ottolenghi, A. Leblanc, M. J. Kim, F. Letourneur et al., Two persistent organic pollutants which act through different xenosensors (alpha-endosulfan and 2,3,7,8 tetrachlorodibenzo-p-dioxin) interact in a mixture and downregulate multiple genes involved in human hepatocyte lipid and glucose metabolism, Biochimie, vol.116, pp.79-91, 2015.
DOI : 10.1016/j.biochi.2015.07.003

URL : https://hal.archives-ouvertes.fr/hal-01174215

E. Dere, R. Lo, T. Celius, J. Matthews, and T. R. Zacharewski, Integration of Genome-Wide Computation DRE Search, AhR ChIP-chip and Gene Expression Analyses of TCDD-Elicited Responses in the Mouse Liver, BMC Genomics, vol.19, issue.Suppl 2, pp.365-375, 2011.
DOI : 10.1101/gr.092759.109

S. Sato, H. Shirakawa, S. Tomita, Y. Ohsaki, K. Haketa et al., Low-dose dioxins alter gene expression related to cholesterol biosynthesis, lipogenesis, and glucose metabolism through the aryl hydrocarbon receptor-mediated pathway in mouse liver, Toxicology and Applied Pharmacology, vol.229, issue.1, pp.10-19, 2008.
DOI : 10.1016/j.taap.2007.12.029

R. Nault, K. A. Fader, D. A. Ammendolia, P. Dornbos, D. Potter et al., Dose-Dependent Metabolic Reprogramming and Differential Gene Expression in TCDD-Elicited Hepatic Fibrosis, Toxicological Sciences, vol.6, issue.346, pp.253-266, 2016.
DOI : 10.1289/ehp.1409055

Y. Wei, L. Zhao, W. He, J. Yang, C. Geng et al., Benzo[a]pyrene promotes gastric cancer cell proliferation and metastasis likely through the Aryl hydrocarbon receptor M A

A. C. Accepted, ERK-dependent induction of MMP9 and c-myc, Int. J. Oncol, vol.49, 2016.

R. Ho, C. Chronis, and K. Plath, Mechanistic insights into reprogramming to induced pluripotency, Journal of Cellular Physiology, vol.4, issue.4, pp.868-878, 2011.
DOI : 10.1016/j.stem.2009.04.005

P. S. Ward and C. B. Thompson, Metabolic Reprogramming: A Cancer Hallmark Even Warburg Did Not Anticipate, Cancer Cell, vol.21, issue.3, pp.297-308, 2012.
DOI : 10.1016/j.ccr.2012.02.014

S. U. Vorrink and F. E. Domann, Regulatory crosstalk and interference between the xenobiotic and hypoxia sensing pathways at the AhR-ARNT-HIF1?? signaling node, Chemico-Biological Interactions, vol.218, 2014.
DOI : 10.1016/j.cbi.2014.05.001

A. Al-dhfyan, A. Alhoshani, and H. M. Korashy, Aryl hydrocarbon receptor/cytochrome P450 1A1 pathway mediates breast cancer stem cells expansion through PTEN inhibition and ?-Catenin and Akt activation, Mol. Cancer, vol.16
DOI : 10.1186/s12943-016-0570-y

URL : https://molecular-cancer.biomedcentral.com/track/pdf/10.1186/s12943-016-0570-y?site=molecular-cancer.biomedcentral.com

L. Huc, X. Tekpli, J. A. Holme, M. Rissel, A. Solhaug et al., c-Jun NH2-Terminal Kinase-Related Na+/H+ Exchanger Isoform 1 Activation Controls Hexokinase II Expression in Benzo(a)Pyrene-Induced Apoptosis, Cancer Research, vol.67, issue.4, pp.1696-705, 2007.
DOI : 10.1158/0008-5472.CAN-06-2327

URL : https://hal.archives-ouvertes.fr/hal-00690320

K. Hardonnière, E. Saunier, A. Lemarié, M. Fernier, I. Gallais et al., The environmental carcinogen benzo[a]pyrene induces a Warburg-like metabolic reprogramming dependent on NHE1 and associated with cell survival, Scientific Reports, vol.34, issue.1
DOI : 10.1038/onc.2014.321

K. Hardonnière, L. Huc, O. Sergent, J. A. Holme, and D. , Lagadic-Gossmann, Environmental carcinogenesis and pH homeostasis: Not only a matter of dysregulated metabolism, Semin. Cancer Biol, vol.43, 2017.

L. Huc, L. Sparfel, M. Rissel, M. Dimanche-boitrel, A. Guillouzo et al., exchange as a new target for toxic polycyclic aromatic hydrocarbons, The FASEB Journal, vol.18, issue.2, pp.344-346, 2004.
DOI : 10.1096/fj.03-0316fje

K. Hardonnière, M. Fernier, I. Gallais, B. Mograbi, N. Podechard et al., Role for the ATPase inhibitory factor 1 in the environmental carcinogen-induced Warburg phenotype, Scientific Reports, vol.13, issue.8, pp.41598-41615
DOI : 10.1016/j.semcancer.2017.01.001

D. M. Tappenden, S. G. Lynn, R. B. Crawford, K. Lee, A. Vengellur et al., The aryl hydrocarbon receptor interacts with ATP5??1, a subunit of the ATP synthase complex, and modulates mitochondrial function, Toxicology and Applied Pharmacology, vol.254, issue.3, pp.299-310, 2011.
DOI : 10.1016/j.taap.2011.05.004

F. J. Quintana and D. H. Sherr, Aryl Hydrocarbon Receptor Control of Adaptive Immunity, Pharmacological Reviews, vol.65, issue.4, 2013.
DOI : 10.1124/pr.113.007823

A. A. Fernández-ramos, C. Marchetti-laurent, V. Poindessous, S. Antonio, C. Petitgas et al., A comprehensive characterization of the impact of mycophenolic acid on the metabolism of Jurkat T cells, Scientific Reports, vol.25, issue.1, pp.41598-41615
DOI : 10.1038/srep30776

L. Almeida, M. Lochner, L. Berod, and T. Sparwasser, Metabolic pathways in T cell activation and lineage differentiation, Seminars in Immunology, vol.28, issue.5, 2016.
DOI : 10.1016/j.smim.2016.10.009

D. Jestaedt, M. Schrenk, M. Weller, G. J. Jugold, C. L. Guillemin et al., An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor, Nature, pp.478-197, 1038.

J. D. Mezrich, J. H. Fechner, X. Zhang, B. P. Johnson, W. J. Burlingham et al., An Interaction between Kynurenine and the Aryl Hydrocarbon Receptor Can Generate Regulatory T Cells, The Journal of Immunology, vol.185, issue.6, pp.3190-3198, 1950.
DOI : 10.4049/jimmunol.0903670

C. Schiering, E. Wincent, A. Metidji, A. Iseppon, Y. Li et al., Feedback control of AHR signalling regulates intestinal immunity, Nature, vol.7, issue.7640, pp.542-242, 1038.
DOI : 10.1186/1471-213X-1-4

M. G. Vander-heiden, L. C. Cantley, and C. B. Thompson, Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation, Science, vol.26, issue.1, pp.1029-1033, 2009.
DOI : 10.1038/nrc2536

L. M. Solis, C. Behrens, W. Dong, M. Suraokar, N. C. Ozburn et al., Nrf2 and Keap1 Abnormalities in Non-Small Cell Lung Carcinoma and Association with Clinicopathologic Features, Clinical Cancer Research, vol.16, issue.14, pp.3743-3753, 2010.
DOI : 10.1158/1078-0432.CCR-09-3352

Y. Mitsuishi, K. Taguchi, Y. Kawatani, T. Shibata, T. Nukiwa et al., Nrf2 Redirects Glucose and Glutamine into Anabolic Pathways in Metabolic Reprogramming, Nrf2 Redirects Glucose and Glutamine into Anabolic Pathways in Metabolic Reprogramming, pp.66-79, 2012.
DOI : 10.1016/j.ccr.2012.05.016

V. I. Sayin, S. E. Leboeuf, S. X. Singh, S. M. Davidson, D. Biancur et al., Author response, eLife, vol.15, issue.6, 2017.
DOI : 10.7554/eLife.28083.018

J. L. Clarke, J. B. Murray, B. K. Park, and I. M. Copple, Roles of Nrf2 in drug and chemical toxicity, Current Opinion in Toxicology, vol.1, 2016.
DOI : 10.1016/j.cotox.2016.10.004

J. Adam, E. Hatipoglu, L. O. Flaherty, N. Ternette, N. Sahgal et al., Renal Cyst Formation in Fh1-Deficient Mice Is Independent of the Hif/Phd Pathway: Roles for Fumarate in KEAP1 Succination and Nrf2 Signaling, Cancer Cell, vol.20, issue.4, pp.524-537, 2011.
DOI : 10.1016/j.ccr.2011.09.006

M. Komatsu, H. Kurokawa, S. Waguri, K. Taguchi, A. Kobayashi et al., The selective autophagy substrate p62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1, Nature Cell Biology, vol.17, pp.213-22310, 1038.
DOI : 10.1016/S0002-9440(10)64369-6

A. Lau, Y. Zheng, S. Tao, H. Wang, S. A. Whitman et al., Arsenic Inhibits Autophagic Flux, Activating the Nrf2-Keap1 Pathway in a p62-Dependent Manner, Molecular and Cellular Biology, vol.33, issue.12, pp.2436-244601748, 2013.
DOI : 10.1128/MCB.01748-12

A. Morandi, M. L. Taddei, P. Chiarugi, and E. Giannoni, Targeting the Metabolic Reprogramming That Controls Epithelial-to-Mesenchymal Transition in Aggressive Tumors, Front, Oncol, vol.7

L. Bui, C. Tomkiewicz, A. Chevallier, S. Pierre, A. Bats et al., Nedd9/Hef1/Cas-L mediates the effects of environmental pollutants on cell migration and plasticity, Oncogene, vol.16, issue.41, pp.28-3642, 2009.
DOI : 10.1615/CritRevImmunol.v26.i5.20

S. Pierre, A. Bats, A. Chevallier, L. Bui, A. Ambolet-camoit et al., Induction of the Ras activator Son of Sevenless 1 by environmental pollutants mediates their effects on cellular proliferation, Biochemical Pharmacology, vol.81, issue.2, pp.304-313, 2011.
DOI : 10.1016/j.bcp.2010.10.003

URL : https://hal.archives-ouvertes.fr/hal-00649887

C. Tomkiewicz, L. Herry, L. Bui, C. Métayer, M. Bourdeloux et al., The aryl hydrocarbon receptor regulates focal adhesion sites through a non-genomic FAK/Src pathway, Oncogene, vol.31, issue.14, pp.1811-1820, 2013.
DOI : 10.1038/onc.2011.314