, Fabienne Béguet-Crespel 1 , Hervé M. Blottière 1, 4 and Nicolas Lapaque 1*, vol.1

M. Institute, A. Inra, and U. Paris,

. University-grenoble-alpes, . Cnrs, . Cea, and . Ibs,

I. Metagenopolis and U. Paris-saclay,

T. S. Postler and S. Ghosh, Understanding the Holobiont: How Microbial Metabolites Affect Human Health and Shape the Immune System, Cell metabolism, vol.26, pp.110-130, 2017.

J. Gao, Impact of the Gut Microbiota on Intestinal Immunity Mediated by Tryptophan Metabolism. Front Cell Infect Microbiol, vol.8, 2018.

F. J. Quintana and D. H. Sherr, Aryl hydrocarbon receptor control of adaptive immunity, Pharmacol Rev, vol.65, pp.1148-1161, 2013.

M. Veldhoen and C. Ferreira, Influence of nutrient-derived metabolites on lymphocyte immunity, Nature medicine, vol.21, pp.709-718, 2015.

R. Barouki, M. Aggerbeck, L. Aggerbeck, and X. Coumoul, The aryl hydrocarbon receptor system, Drug Metabol Drug Interact, vol.27, pp.3-8, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02190803

T. D. Hubbard, Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles, Scientific reports, vol.5, p.12689, 2015.

T. D. Hubbard, I. A. Murray, and G. H. Perdew, Indole and Tryptophan Metabolism: Endogenous and Dietary Routes to Ah Receptor Activation, Drug Metab Dispos, vol.43, pp.1522-1535, 2015.

U. H. Jin, Microbiome-derived tryptophan metabolites and their aryl hydrocarbon receptordependent agonist and antagonist activities, Mol Pharmacol, vol.85, pp.777-788, 2014.

Y. Fujii-kuriyama, M. Ema, J. Mimura, and K. Sogawa, Ah receptor: a novel ligand-activated transcription factor, Exp Clin Immunogenet, vol.11, pp.65-74, 1994.

O. Hankinson, The aryl hydrocarbon receptor complex, Annu Rev Pharmacol Toxicol, vol.35, pp.307-340, 1995.

B. Lamas, CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands, Nature medicine, vol.22, pp.598-605, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02785302

I. Monteleone, Aryl hydrocarbon receptor-induced signals up-regulate IL-22 production and inhibit inflammation in the gastrointestinal tract, Gastroenterology, vol.141, p.231, 2011.

K. Furumatsu, A role of the aryl hydrocarbon receptor in attenuation of colitis, Dig Dis Sci, vol.56, pp.2532-2544, 2011.

J. A. Goettel, AHR Activation Is Protective against Colitis Driven by T Cells in Humanized Mice, Cell reports, vol.17, pp.1318-1329, 2016.

Z. Liu, Aryl hydrocarbon receptor activation maintained the intestinal epithelial barrier function through Notch1 dependent signaling pathway, Int J Mol Med, vol.41, pp.1560-1572, 2018.

M. Yu, Aryl Hydrocarbon Receptor Activation Modulates Intestinal Epithelial Barrier Function by Maintaining Tight Junction Integrity, Int J Biol Sci, vol.14, pp.69-77, 2018.

C. Schiering, A. Vonk, S. Das, B. Stockinger, and E. Wincent, Cytochrome P4501-inhibiting chemicals amplify aryl hydrocarbon receptor activation and IL-22 production in T helper 17 cells, Biochem Pharmacol, vol.151, pp.47-58, 2018.

C. Schiering, Feedback control of AHR signalling regulates intestinal immunity, Nature, vol.542, pp.242-245, 2017.

T. Zelante, Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22, Immunity, vol.39, pp.372-385, 2013.

R. Sonowal, Indoles from commensal bacteria extend healthspan, Proceedings of the National Academy of Sciences of the United States of America, vol.114, pp.7506-7515, 2017.

M. Vital, A. C. Howe, and J. Tiedje, Revealing the bacterial butyrate synthesis pathways by analyzing (meta)genomic data, vol.5, p.889, 2014.

J. H. Cummings, Short chain fatty acids in the human colon, Gut, vol.22, pp.763-779, 1981.

J. H. Cummings, E. W. Pomare, W. J. Branch, C. P. Naylor, and G. T. Macfarlane, Short chain fatty acids in human large intestine, portal, hepatic and venous blood, Gut, vol.28, pp.1221-1227, 1987.

F. Blachier, F. Mariotti, J. F. Huneau, and D. Tome, Effects of amino acid-derived luminal metabolites on the colonic epithelium and physiopathological consequences, Amino Acids, vol.33, pp.547-562, 2007.
URL : https://hal.archives-ouvertes.fr/hal-01186779

U. E. Schaible and S. H. Kaufmann, A nutritive view on the host-pathogen interplay, Trends in microbiology, vol.13, pp.373-380, 2005.

M. Cuff, J. Dyer, M. Jones, and S. Shirazi-beechey, The human colonic monocarboxylate transporter Isoform 1: its potential importance to colonic tissue homeostasis, Gastroenterology, vol.128, pp.676-686, 2005.

U. H. Jin, Short Chain Fatty Acids Enhance Aryl Hydrocarbon (Ah) Responsiveness in Mouse Colonocytes and Caco-2 Human Colon Cancer Cells, Scientific reports, vol.7, p.10163, 2017.

P. Gallinari, S. Di-marco, P. Jones, M. Pallaoro, and C. Steinkuhler, HDACs, histone deacetylation and gene transcription: from molecular biology to cancer therapeutics, Cell Res, vol.17, pp.195-211, 2007.

M. Kinoshita, Y. Suzuki, and Y. Saito, Butyrate reduces colonic paracellular permeability by enhancing PPARgamma activation, Biochem Biophys Res Commun, vol.293, pp.827-831, 2002.

A. and S. , Short-chain fatty acids stimulate angiopoietin-like 4 synthesis in human colon adenocarcinoma cells by activating peroxisome proliferator-activated receptor gamma, Mol Cell Biol, vol.33, pp.1303-1316, 2013.

Z. Yin, E. C. Henry, and T. A. Gasiewicz, Epigallocatechin-3-gallate is a novel Hsp90 inhibitor, Biochemistry, vol.48, pp.336-345, 2009.

A. Perkins, A Structural Switch between Agonist and Antagonist Bound Conformations for a Ligand-Optimized Model of the Human Aryl Hydrocarbon Receptor Ligand Binding Domain, Biology (Basel), vol.3, pp.645-669, 2014.

K. J. Smith, Identification of a high-affinity ligand that exhibits complete aryl hydrocarbon receptor antagonism, J Pharmacol Exp Ther, vol.338, pp.318-327, 2011.

C. M. Palermo, C. A. Westlake, and T. A. Gasiewicz, Epigallocatechin gallate inhibits aryl hydrocarbon receptor gene transcription through an indirect mechanism involving binding to a 90 kDa heat shock protein, Biochemistry, vol.44, pp.5041-5052, 2005.

E. C. Henry and T. A. Gasiewicz, Agonist but not antagonist ligands induce conformational change in the mouse aryl hydrocarbon receptor as detected by partial proteolysis, Mol Pharmacol, vol.63, pp.392-400, 2003.

L. Bonati, D. Corrada, S. G. Tagliabue, and S. Motta, Molecular modeling of the AhR structure and interactions can shed light on ligand-dependent activation and transformation mechanisms, Curr Opin Toxicol, vol.2, pp.42-49, 2017.

W. H. Bisson, Modeling of the aryl hydrocarbon receptor (AhR) ligand binding domain and its utility in virtual ligand screening to predict new AhR ligands, J Med Chem, vol.52, pp.5635-5641, 2009.

P. J. Erbel, Solution structure of the alpha-subunit of human chorionic gonadotropin, Eur J Biochem, vol.260, pp.490-498, 1999.

B. Stockinger, P. Di-meglio, M. Gialitakis, and J. H. Duarte, The aryl hydrocarbon receptor: multitasking in the immune system, Annu Rev Immunol, vol.32, pp.403-432, 2014.

J. Qiu, Group 3 innate lymphoid cells inhibit T-cell-mediated intestinal inflammation through aryl hydrocarbon receptor signaling and regulation of microflora, Immunity, vol.39, pp.386-399, 2013.

J. Qiu and L. Zhou, Aryl hydrocarbon receptor promotes RORgammat(+) group 3 ILCs and controls intestinal immunity and inflammation, Seminars in immunopathology, vol.35, pp.657-670, 2013.

S. Fukumoto, Identification of a probiotic bacteria-derived activator of the aryl hydrocarbon receptor that inhibits colitis, Immunol Cell Biol, vol.92, pp.460-465, 2014.

P. Moura-alves, AhR sensing of bacterial pigments regulates antibacterial defence, Nature, vol.512, pp.387-392, 2014.

K. Daly and S. P. Shirazi-beechey, Microarray analysis of butyrate regulated genes in colonic epithelial cells, DNA Cell Biol, vol.25, pp.49-62, 2006.

P. M. Garrison, J. M. Rogers, W. R. Brackney, and M. S. Denison, Effects of histone deacetylase inhibitors on the Ah receptor gene promoter, Arch Biochem Biophys, vol.374, pp.161-171, 2000.

R. K. Gill, Expression and membrane localization of MCT isoforms along the length of the human intestine, Am J Physiol Cell Physiol, vol.289, pp.846-852, 2005.

C. Hadjiagapiou, L. Schmidt, P. K. Dudeja, T. J. Layden, and K. Ramaswamy, Mechanism(s) of butyrate transport in Caco-2 cells: role of monocarboxylate transporter 1, American journal of physiology. Gastrointestinal and liver physiology, vol.279, pp.775-780, 2000.

R. Thibault, Butyrate utilization by the colonic mucosa in inflammatory bowel diseases: a transport deficiency, Inflammatory bowel diseases, vol.16, pp.684-695, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01173382

C. A. Flaveny, I. A. Murray, C. R. Chiaro, and G. H. Perdew, Ligand selectivity and gene regulation by the human aryl hydrocarbon receptor in transgenic mice, Mol Pharmacol, vol.75, pp.1412-1420, 2009.

H. Sokol, Low counts of Faecalibacterium prausnitzii in colitis microbiota, Inflammatory bowel diseases, vol.15, pp.1183-1189, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00657435

K. Machiels, A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis, Gut, vol.63, pp.1275-1283, 2014.

R. Kumari, V. Ahuja, and J. Paul, Fluctuations in butyrate-producing bacteria in ulcerative colitis patients of North India, World J Gastroenterol, vol.19, pp.3404-3414, 2013.

K. Cushing, D. M. Alvarado, and M. A. Ciorba, Butyrate and Mucosal Inflammation: New Scientific Evidence Supports Clinical Observation, Clin Transl Gastroenterol, vol.6, p.108, 2015.

E. E. Alexeev, Microbiota-Derived Indole Metabolites Promote Human and Murine Intestinal Homeostasis through Regulation of Interleukin-10 Receptor, Am J Pathol, 2018.

L. Zheng, Microbial-Derived Butyrate Promotes Epithelial Barrier Function through IL-10 Receptor-Dependent Repression of Claudin-2, Journal of immunology, vol.199, pp.2976-2984, 2017.

R. E. Hungate, The anaerobic mesophilic cellulolytic bacteria, Bacteriol Rev, vol.14, pp.1-49, 1950.

O. Lakhdari, Identification of NF-kappaB modulation capabilities within human intestinal commensal bacteria, Journal of biomedicine & biotechnology, p.282356, 2011.

C. Bourriaud, Lactate is mainly fermented to butyrate by human intestinal microfloras but interindividual variation is evident, J Appl Microbiol, vol.99, pp.201-212, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02680381

G. C. Van-zundert, The HADDOCK2.2 Web Server: User-Friendly Integrative Modeling of Biomolecular Complexes, J Mol Biol, vol.428, pp.720-725, 2016.

T. Wassenaar, Structural Biology on the Grid. Journal of Grid Computing, vol.10, pp.743-767, 2012.

D. Wu, N. Potluri, J. Lu, Y. Kim, and F. Rastinejad, Structural integration in hypoxia-inducible factors, Nature, vol.524, pp.303-308, 2015.

M. Kallberg, Template-based protein structure modeling using the RaptorX web server, Nat Protoc, vol.7, pp.1511-1522, 2012.

J. Ma, J. Peng, S. Wang, and J. Xu, A conditional neural fields model for protein threading, Bioinformatics, vol.28, pp.59-66, 2012.

E. Chovancova, CAVER 3.0: a tool for the analysis of transport pathways in dynamic protein structures, PLoS Comput Biol, vol.8, 2012.

B. K. Ho and F. Gruswitz, HOLLOW: generating accurate representations of channel and interior surfaces in molecular structures, BMC Struct Biol, vol.8, p.49, 2008.

E. E. Alexeev, J. M. Lanis, D. J. Kao, E. L. Campbell, C. J. Kelly et al., , 2018.

, Microbiota-Derived Indole Metabolites Promote Human and Murine Intestinal Homeostasis through Regulation of Interleukin-10 Receptor

T. Bansal, R. C. Alaniz, T. K. Wood, and A. Jayaraman, The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation, Proceedings of the National Academy of Sciences, vol.107, issue.1, pp.228-233, 2010.

E. Biagi, M. Candela, S. Turroni, P. Garagnani, C. Franceschi et al., Ageing and gut microbes: Perspectives for health maintenance and longevity, Pharmacological Research, vol.69, issue.1, pp.11-20, 2013.

B. Biavati and P. Mattarelli, The Family Bifidobacteriaceae, The Prokaryotes, pp.322-382, 2006.

,

N. Cerf-bensussan and V. Gaboriau-routhiau, The immune system and the gut microbiota: Friends or foes? Nature Reviews Immunology, 2010.

D. Vuyst, L. Leroy, and F. , Cross-feeding between bifidobacteria and butyrate-producing colon bacteria explains bifdobacterial competitiveness, butyrate production, and gas production, International Journal of Food Microbiology, vol.149, issue.1, pp.73-80, 2011.

D. Di-gioia, I. Aloisio, G. Mazzola, and B. Biavati, Bifidobacteria: their impact on gut microbiota composition and their applications as probiotics in infants, Applied Microbiology and Biotechnology, vol.98, issue.2, pp.563-577, 2014.

J. B. Ewaschuk, H. Diaz, L. Meddings, B. Diederichs, A. Dmytrash et al., Secreted bioactive factors from Bifidobacterium infantis enhance epithelial cell barrier function, American Journal of Physiology-Gastrointestinal and Liver Physiology, vol.295, issue.5, pp.1025-1034, 2008.

C. A. Flaveny, I. A. Murray, C. R. Chiaro, and G. H. Perdew, Ligand Selectivity and Gene Regulation by the Human Aryl Hydrocarbon Receptor in Transgenic Mice, Molecular Pharmacology, vol.75, issue.6, pp.1412-1420, 2009.

Y. Fujii-kuriyama, M. Ema, J. Mimura, and K. Sogawa, Ah receptor: a novel ligand-activated transcription factor, Experimental and Clinical Immunogenetics, vol.11, issue.2-3, pp.65-74, 1994.

S. Fukuda, H. Toh, K. Hase, K. Oshima, Y. Nakanishi et al., , 2011.

, Bifidobacteria can protect from enteropathogenic infection through production of acetate, Nature, issue.7331, pp.543-549

S. Fukumoto, T. Toshimitsu, S. Matsuoka, A. Maruyama, K. Oh-oka et al., Identification of a probiotic bacteria-derived activator of the aryl hydrocarbon receptor that inhibits colitis, Immunology and Cell Biology, vol.92, issue.5, pp.460-465, 2014.

N. Gagliani, M. C. Vesely, A. Iseppon, L. Brockmann, H. Xu et al., , 2015.

, Th17 cells transdifferentiate into regulatory T cells during resolution of inflammation, Nature, pp.1-5

V. Grimm, C. Westermann, and C. U. Riedel, Bifidobacteria-host interactions--an update on colonisation factors, BioMed Research International, p.960826, 2014.

O. Hankinson, The aryl hydrocarbon receptor complex, Annual Review of Pharmacology and Toxicology, vol.35, issue.1, pp.307-340, 1995.

T. Hashimoto, T. Perlot, A. Rehman, J. Trichereau, H. Ishiguro et al., ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation, Nature, vol.487, pp.477-483, 2012.

C. Hsieh, T. Osaka, E. Moriyama, Y. Date, J. Kikuchi et al., Strengthening of the intestinal epithelial tight junction by Bifidobacterium bifidum, Physiological Reports, vol.3, issue.3, 2015.

T. D. Hubbard, I. A. Murray, W. H. Bisson, T. S. Lahoti, K. Gowda et al., Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles, 2015.

C. Schiering, A. Vonk, S. Das, B. Stockinger, and E. Wincent, Cytochrome P4501-inhibiting chemicals amplify aryl hydrocarbon receptor activation and IL-22 production in T helper 17 cells, Biochemical Pharmacology, vol.151, pp.47-58, 2018.

Y. Shimada, M. Kinoshita, K. Harada, M. Mizutani, K. Masahata et al., , 2013.

, Commensal bacteria-dependent indole production enhances epithelial barrier function in the colon, PLoS ONE, vol.8, issue.11

M. Spiljar, D. Merkler, and M. Trajkovski, The Immune System Bridges the Gut Microbiota with Systemic Energy Homeostasis: Focus on TLRs, Mucosal Barrier, and SCFAs, Frontiers in Immunology, vol.8, p.1353, 2017.

B. Stockinger, P. Meglio, . Di, M. Gialitakis, and J. H. Duarte, The Aryl Hydrocarbon Receptor: Multitasking in the Immune System, Annual Review of Immunology, vol.32, issue.1, pp.403-432, 2014.

N. Voreades, A. Kozil, and T. L. Weir, Diet and the development of the human intestinal microbiome, Frontiers in Microbiology, vol.5, 2014.

J. M. Wells, O. Rossi, M. Meijerink, and P. Van-baarlen, Epithelial crosstalk at the microbiotamucosal interface, Proceedings of the, p.108, 2011.

, , pp.4607-4614

M. Yu, Q. Wang, Y. Ma, L. Li, K. Yu et al., Aryl Hydrocarbon Receptor Activation Modulates Intestinal Epithelial Barrier Function by Maintaining Tight Junction Integrity, International Journal of Biological Sciences, vol.14, issue.1, pp.69-77, 2018.

S. Yu and N. Gao, Compartmentalizing intestinal epithelial cell toll-like receptors for immune surveillance, Cellular and Molecular Life Sciences, vol.72, issue.17, pp.3343-3353, 2015.

T. Zelante, R. G. Iannitti, C. Cunha, A. Deluca, G. Giovannini et al., Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22, Immunity, vol.39, issue.6, pp.372-385, 2013.

T. Zelante, G. Pieraccini, L. Scaringi, F. Aversa, and L. Romani, Learning from other diseases: protection and pathology in chronic fungal infections, Seminars in Immunopathology, 2016.

. Bibliography,

B. D. Abbott, L. S. Birnbaum, and G. H. Perdew, Developmental expression of two members of a new class of transcription factors: I. Expression of aryl hydrocarbon receptor in the C57BL/6N mouse embryo, Developmental Dynamics, vol.204, issue.2, pp.133-143, 1995.

C. C. Abnet, Two forms of aryl hydrocarbon receptor type 2 in rainbow trout (Oncorhynchus mykiss). Evidence for differential expression and enhancer specificity, The Journal of biological chemistry, vol.274, issue.21, pp.15159-66, 1999.

J. Adachi, Indirubin and Indigo are Potent Aryl Hydrocarbon Receptor Ligands Present in Human Urine, Journal of Biological Chemistry, vol.276, issue.34, pp.31475-31478, 2001.

M. Al-waiz, The exogenous origin of trimethylamine in the mouse, Metabolism, vol.41, issue.2, pp.135-136, 1992.

M. Aldunate, Antimicrobial and immune modulatory effects of lactic acid and short chain fatty acids produced by vaginal microbiota associated with eubiosis and bacterial vaginosis, Frontiers in physiology, vol.6, p.1, 2015.

A. and S. , Short-Chain Fatty Acids Stimulate Angiopoietin-Like 4 Synthesis in Human Colon Adenocarcinoma Cells by Activating Peroxisome Proliferator-Activated Receptor, Molecular and Cellular Biology, vol.33, issue.7, pp.1303-1316, 2013.

E. E. Alexeev, Microbiota-Derived Indole Metabolites Promote Human and Murine Intestinal Homeostasis through Regulation of Interleukin-10 Receptor, The American Journal of Pathology, 2018.

J. Amar, Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: molecular mechanisms and probiotic treatment, EMBO Molecular Medicine, vol.3, issue.9, pp.559-572, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00801348

P. Andersson, A constitutively active dioxin/aryl hydrocarbon receptor induces stomach tumors, Proceedings of the National Academy of Sciences of the United States of America, vol.99, pp.9990-9995, 2002.

C. Antonsson, Distinct roles of the molecular chaperone hsp90 in modulating dioxin receptor function via the basic helix-loop-helix and PAS domains, Molecular and cellular biology, vol.15, issue.2, pp.756-65, 1995.

J. Aron-wisnewsky and K. Clément, The gut microbiome, diet, and links to cardiometabolic and chronic disorders, Nature Reviews Nephrology, vol.12, issue.3, pp.169-181, 2016.

M. Arumugam, Enterotypes of the human gut microbiome, Nature, vol.473, issue.7346, pp.174-180, 2011.
URL : https://hal.archives-ouvertes.fr/cea-00903625

C. Atuma, The adherent gastrointestinal mucus gel layer: thickness and physical state in vivo, American Journal of Physiology-Gastrointestinal and Liver Physiology, vol.280, issue.5, pp.922-929, 2001.

T. Baba, Structure and Expression of the Ah Receptor Repressor Gene, Journal of Biological Chemistry, vol.276, issue.35, pp.33101-33110, 2001.

J. Bach, The Effect of Infections on Susceptibility to Autoimmune and Allergic Diseases, New England Journal of Medicine, vol.347, issue.12, pp.911-920, 2002.

F. Bäckhed, The gut microbiota as an environmental factor that regulates fat storage, Proceedings of the National Academy of Sciences of the United States of America, vol.101, pp.15718-15741, 2004.

M. Backlund and M. Ingelman-sundberg, Regulation of aryl hydrocarbon receptor signal transduction by protein tyrosine kinases, Cellular Signalling, vol.17, issue.1, pp.39-48, 2005.

S. G. Bacsi, S. Reisz-porszasz, and O. Hankinson, Orientation of the heterodimeric aryl hydrocarbon (dioxin) receptor complex on its asymmetric DNA recognition sequence, Molecular Pharmacology, vol.47, issue.3, pp.432-438, 1995.

P. A. Baeuerle and V. R. Baichwal, NF-kappa B as a frequent target for immunosuppressive and antiinflammatory molecules, Advances in immunology, vol.65, pp.111-148, 1997.

T. Bansal, The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation, Proceedings of the National Academy of Sciences, vol.107, issue.1, pp.228-233, 2010.

C. Barnaba, The catalytic function of cytochrome P450 is entwined with its membrane-bound nature, vol.6, p.662, 1000.

R. Barouki, X. Coumoul, and P. M. Fernandez-salguero, The aryl hydrocarbon receptor, more than a xenobiotic-interacting protein, FEBS Letters, vol.581, issue.19, pp.3608-3615, 2007.
URL : https://hal.archives-ouvertes.fr/hal-02175734

M. E. Bashir, Toll-like receptor 4 signaling by intestinal microbes influences susceptibility to food allergy, Journal of immunology, vol.172, issue.11, pp.6978-87, 1950.

R. L. Batterham and S. R. Bloom, The Gut Hormone Peptide YY Regulates Appetite, Annals of the New York Academy of Sciences, vol.994, issue.1, pp.162-168, 2003.

T. V. Beischlag and G. H. Perdew, ERa-AHR-ARNT protein-protein interactions mediate estradioldependent transrepression of dioxin-inducible gene transcription, Journal of Biological Chemistry, vol.280, issue.22, pp.21607-21611, 2005.

P. E. Bendheim, Development of indole-3-propionic acid (OXIGON TM ) for alzheimer's disease, Journal of Molecular Neuroscience, vol.19, issue.1-2, pp.213-217, 2002.

J. M. Benson and D. M. Shepherd, Aryl hydrocarbon receptor activation by TCDD reduces inflammation associated with Crohn's disease. Toxicological sciences : an official journal of the Society of Toxicology, vol.120, pp.68-78, 2011.

L. Bergander, Metabolic fate of the Ah receptor ligand 6-formylindolo[3,2-b]carbazole, Chemico-Biological Interactions, vol.149, issue.2-3, pp.151-164, 2004.

J. H. Bernink, Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues, Nature Immunology, vol.14, issue.3, pp.221-229, 2013.

E. Biagi, Through Ageing, and Beyond: Gut Microbiota and Inflammatory Status in Seniors and Centenarians N. Ahmed, vol.5, p.10667, 2010.

W. H. Bisson, Modeling of the aryl hydrocarbon receptor (AhR) ligand binding domain and its utility in virtual ligand screening to predict new AhR ligands, Journal of Medicinal Chemistry, vol.52, issue.18, pp.5635-5641, 2009.

L. F. Bjeldanes, Aromatic hydrocarbon responsiveness-receptor agonists generated from indole-3-carbinol in vitro and in vivo: comparisons with 2,3,7,8-tetrachlorodibenzo-p-dioxin, Proceedings of the National Academy of Sciences of the United States of America, vol.88, pp.9543-9547, 1991.

M. Bjursell, Improved glucose control and reduced body fat mass in free fatty acid receptor 2-deficient mice fed a high-fat diet, American Journal of Physiology-Endocrinology and Metabolism, vol.300, issue.1, pp.211-220, 2011.

H. P. Browne, Culturing of "unculturable" human microbiota reveals novel taxa and extensive sporulation, Nature, vol.533, issue.7604, pp.543-546, 2016.

S. Buonocore, Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology, Nature, vol.464, issue.7293, pp.1371-1375, 2010.

R. A. Butler, An aryl hydrocarbon receptor (AHR) homologue from the soft-shell clam, Mya arenaria: evidence that invertebrate AHR homologues lack 2,3,7,8-tetrachlorodibenzo-p-dioxin and ?naphthoflavone binding, Gene, vol.278, issue.1-2, pp.223-234, 2001.

R. Cabrera-rubio, Microbiome diversity in the bronchial tracts of patients with chronic obstructive pulmonary disease, Journal of clinical microbiology, vol.50, issue.11, pp.3562-3570, 2012.

A. Camelo, IL-33, IL-25, and TSLP induce a distinct phenotypic and activation profile in human type 2 innate lymphoid cells, Blood Advances, vol.1, issue.10, pp.577-589, 2017.

B. L. Cantarel, The Carbohydrate-Active EnZymes database (CAZy): an expert resource for Glycogenomics, Nucleic Acids Research, vol.37, pp.233-238, 2009.

E. Cario, G. Gerken, and D. K. Podolsky, Toll-like receptor 2 enhances ZO-1-associated intestinal epithelial barrier integrity via protein kinase C, Gastroenterology, vol.127, issue.1, pp.224-262, 2004.

L. A. Carver and C. A. Bradfield, Ligand-dependent interaction of the aryl hydrocarbon receptor with a novel immunophilin homolog in vivo, Journal of Biological Chemistry, vol.272, issue.17, pp.11452-11458, 1997.

L. A. Carver, V. Jackiw, and C. A. Bradfields, The 90-kDa Heat Shock Protein Is Essential for Ah Receptor Signaling in a Yeast Expression System, THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol.269, issue.48, pp.30109-30112, 1994.

C. Y. Chang and A. Puga, Constitutive activation of the aromatic hydrocarbon receptor, Molecular and cellular biology, vol.18, issue.1, pp.525-560, 1998.

A. Chapman-smith, J. K. Lutwyche, and M. L. Whitelaw, Contribution of the Per/Arnt/Sim (PAS) Domains to DNA Binding by the Basic Helix-Loop-Helix PAS Transcriptional Regulators, Journal of Biological Chemistry, vol.279, issue.7, pp.5353-5362, 2004.

L. Chatelier and E. , Richness of human gut microbiome correlates with metabolic markers, Nature, vol.500, issue.7464, pp.541-546, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01190602

H. S. Chen, S. S. Singh, and G. H. Perdew, The Ah receptor is a sensitive target of geldanamycininduced protein turnover, Archives of Biochemistry and Biophysics, vol.348, issue.1, pp.190-198, 1997.

X. Chen, TGR5: a novel target for weight maintenance and glucose metabolism, Experimental diabetes research, p.853501, 2011.

Y. Chen, Lysine propionylation and butyrylation are novel post-translational modifications in histones, Molecular & cellular proteomics : MCP, vol.6, issue.5, pp.812-821, 2007.

C. Chimerel, Bacterial metabolite indole modulates incretin secretion from intestinal enteroendocrine L cells, Cell reports, vol.9, issue.4, pp.1202-1210, 2014.

I. Chinen, The aryl hydrocarbon receptor/microRNA-212/132 axis in T cells regulates IL-10 production to maintain intestinal homeostasis, International Immunology, vol.27, issue.8, pp.405-415, 2015.

L. J. Cohen, Functional metagenomic discovery of bacterial effectors in the human microbiome and isolation of commendamide, a GPCR G2A/132 agonist, Proceedings of the National Academy of Sciences, vol.112, issue.35, pp.4825-4834, 2015.

M. C. Collado, Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid, Scientific Reports, vol.6, issue.1, p.23129, 2016.

P. I. Costea, Enterotypes in the landscape of gut microbial community composition, Nature Microbiology, vol.3, issue.1, pp.8-16, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02629150

A. Cotillard, Dietary intervention impact on gut microbial gene richness, Nature, vol.500, issue.7464, pp.585-588, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01001543

P. Coumailleau, Definition of a minimal domain of the dioxin receptor that is associated with Hsp90 and maintains wild type ligand binding affinity and specificity, Journal of Biological Chemistry, vol.270, issue.42, pp.25291-25300, 1995.

A. Couturier-maillard, NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer, Journal of Clinical Investigation, vol.123, issue.2, pp.700-711, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00944194

M. B. Cox, C. A. Miller, and . Iii, Cooperation of heat shock protein 90 and p23 in aryl hydrocarbon receptor signaling, Cell stress & chaperones, vol.9, issue.1, pp.4-20, 2004.

S. Craciun and E. P. Balskus, Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme, vol.109, pp.21307-21312, 2012.

J. H. Cummings, Short chain fatty acids in human large intestine, portal, hepatic and venous blood, Gut, vol.28, issue.10, pp.1221-1228, 1987.

K. Daly and S. P. Shirazi-beechey, Microarray analysis of butyrate regulated genes in colonic epithelial cells, DNA and cell biology, vol.25, issue.1, pp.49-62, 2006.

A. Darfeuille-michaud, Adherent-invasive Escherichia coli: a putative new E. coli pathotype associated with Crohn's disease, International Journal of Medical Microbiology, vol.292, pp.185-193, 2002.

D. Vadder and F. , Microbiota-Generated Metabolites Promote Metabolic Benefits via Gut-Brain Neural Circuits, Cell, vol.156, issue.1-2, pp.84-96, 2014.

M. S. Denison and S. R. Nagy, Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annual review of pharmacology and toxicology, vol.43, pp.309-334, 2003.

M. Derrien, The Mucin degrader Akkermansia muciniphila is an abundant resident of the human intestinal tract, Applied and environmental microbiology, vol.74, issue.5, pp.1646-1654, 2008.

P. Desreumaux, Attenuation of colon inflammation through activators of the retinoid X receptor (RXR)/peroxisome proliferator-activated receptor gamma (PPARgamma) heterodimer. A basis for new therapeutic strategies, The Journal of experimental medicine, vol.193, issue.7, pp.827-838, 2001.

L. Dethlefsen, M. Mcfall-ngai, and D. A. Relman, An ecological and evolutionary perspective on human-microbe mutualism and disease, Nature, issue.7164, pp.811-818, 2007.

L. Dethlefsen and D. A. Relman, Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation, Proceedings of the National Academy of Sciences, vol.108, issue.Supplement_1, pp.4554-4561, 2011.

P. Dimeglio, Activation of the aryl hydrocarbon receptor dampens the severity of inflammatory skin conditions, Immunity, vol.40, pp.989-1001, 2014.

L. Ding, Ligand-independent activation of estrogen receptor alpha by XBP-1, Nucleic acids research, vol.31, issue.18, pp.5266-74, 2003.

G. P. Donaldson, S. M. Lee, and S. K. Mazmanian, Gut biogeography of the bacterial microbiota, Nature reviews. Microbiology, vol.14, issue.1, pp.20-32, 2016.

D. Donnelly, The structure and function of the glucagon-like peptide-1 receptor and its ligands, British Journal of Pharmacology, vol.166, issue.1, pp.27-41, 2012.

M. K. El-tanani and C. D. Green, Two Separate Mechanisms for Ligand-Independent Activation of the Estrogen Receptor, Molecular Endocrinology, vol.11, issue.7, pp.928-937, 1997.

M. Ema, Dioxin binding activities of polymorphic forms of mouse and human aryl hydrocarbon receptors, J Biol.Chem, vol.269, issue.44, pp.27337-27343, 1994.

C. Esser and A. Rannug, The Aryl Hydrocarbon Receptor in Barrier Organ Physiology, Immunology, and Toxicology. Pharmacological reviews, vol.67, issue.2, pp.259-279, 2015.

B. R. Evans, Repression of aryl hydrocarbon receptor (AHR) signaling by AHR repressor: role of DNA binding and competition for AHR nuclear translocator, Molecular Pharmacology, vol.73, issue.2, pp.387-398, 2008.

A. Everard, Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity, Proceedings of the National Academy of Sciences, vol.110, issue.22, pp.9066-9071, 2013.

R. Fellows, Microbiota derived short chain fatty acids promote histone crotonylation in the colon through histone deacetylases, Nature Communications, vol.9, issue.1, p.105, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01759208

A. Ferguson, Intraepithelial lymphocytes of the small intestine, Gut, vol.18, issue.11, pp.921-958, 1977.

P. M. Fernandez-salguero, Immune System Impairment and Hepatic Fibrosis in Mice Lacking the Dioxin-Binding Ah Receptor, Science, vol.268, pp.722-726, 1995.
URL : https://hal.archives-ouvertes.fr/hal-02714504

P. M. Fernandez-salguero, Lesions of Aryl-hydrocarbon Receptor-deficient Mice, Vet Pathol, vol.34, pp.605-614, 1997.

C. De-filippo, Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa, Proceedings of the National Academy of Sciences of the United States of America, vol.107, pp.14691-14697, 2010.

C. T. Fitzgerald, D. W. Nebert, and A. Puga, Regulation of mouse Ah receptor (Ahr) gene basal expression by members of the Sp family of transcription factors, DNA and cell biology, vol.17, issue.9, pp.811-833, 1998.

C. A. Flaveny, Ligand Selectivity and Gene Regulation by the Human Aryl Hydrocarbon Receptor in Transgenic Mice, Molecular Pharmacology, vol.75, issue.6, pp.1412-1420, 2009.

H. J. Flint, Polysaccharide utilization by gut bacteria: potential for new insights from genomic analysis, Nature reviews. Microbiology, vol.6, issue.2, pp.121-152, 2008.

E. A. Franzosa, Identifying personal microbiomes using metagenomic codes, Proceedings of the National Academy of Sciences of the United States of America, vol.112, pp.2930-2938, 2015.

A. C. Freitas and J. E. Hill, Quantification, isolation and characterization of Bifidobacterium from the vaginal microbiomes of reproductive aged women, Anaerobe, vol.47, pp.145-156, 2017.

M. Frericks, M. Meissner, and C. Esser, Microarray analysis of the AHR system: Tissue-specific flexibility in signal and target genes, Toxicology and Applied Pharmacology, vol.220, issue.3, pp.320-332, 2007.

J. L. Fribourgh and C. L. Partch, Assembly and function of bHLH-PAS complexes, vol.114, pp.5330-5332, 2017.

Y. Fujii-kuriyama, Ah receptor: a novel ligand-activated transcription factor, Experimental and clinical immunogenetics, vol.11, issue.2-3, pp.65-74, 1994.

S. Fukumoto, Identification of a probiotic bacteria-derived activator of the aryl hydrocarbon receptor that inhibits colitis, Immunology and cell biology, vol.92, issue.5, pp.460-465, 2014.

K. Furumatsu, A Role of the Aryl Hydrocarbon Receptor in Attenuation of Colitis, Digestive Diseases and Sciences, vol.56, issue.9, pp.2532-2544, 2011.

J. Gao, Impact of the Gut Microbiota on Intestinal Immunity Mediated by Tryptophan Metabolism, Frontiers in Cellular and Infection Microbiology, vol.8, p.13, 2018.

M. A. Garcia, W. J. Nelson, and N. Chavez, Cell-Cell Junctions Organize Structural and Signaling Networks, Cold Spring Harbor Perspectives in Biology, vol.10, issue.4, p.29181, 2017.

F. Gavini, Differences in the Distribution of Bifidobacterial and Enterobacterial Species in Human Faecal Microflora of Three Different (Children, Adults, Elderly) Age Groups, Microbial Ecology in Health and Disease, vol.13, issue.1, pp.40-45, 2001.
URL : https://hal.archives-ouvertes.fr/hal-00416238

A. Geirnaert, Butyrate-producing bacteria supplemented in vitro to Crohn's disease patient microbiota increased butyrate production and enhanced intestinal epithelial barrier integrity, Scientific Reports, vol.7, issue.1, p.11450, 2017.

S. Gerbal-chaloin, Role of CYP3A4 in the regulation of the aryl hydrocarbon receptor by omeprazole sulphide, Cellular Signalling, vol.18, issue.5, pp.740-750, 2006.

F. Gerbe, Distinct ATOH1 and Neurog3 requirements define tuft cells as a new secretory cell type in the intestinal epithelium, The Journal of cell biology, vol.192, issue.5, pp.767-80, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02459483

F. Gerbe and P. Jay, Intestinal tuft cells: Epithelial sentinels linking luminal cues to the immune system, Mucosal Immunology, vol.9, issue.6, pp.1353-1359, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02459450

A. Geremia, IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease, The Journal of Experimental Medicine, vol.208, issue.6, pp.1127-1133, 2011.

M. Ghotbaddini and J. B. Powell, The AhR Ligand, TCDD, Regulates Androgen Receptor Activity Differently in Androgen-Sensitive versus Castration-Resistant Human Prostate Cancer Cells, International Journal of Environmental Research and Public Health, vol.12, issue.7, pp.7506-7524, 2015.

E. M. Gillam, Formation of indigo by recombinant mammalian cytochrome P450, Biochemical and Biophysical Research Communications, vol.265, issue.2, pp.469-472, 1999.

S. E. Girardin and I. G. Boneca, Nod1 detects a unique muropeptide from gram-negative bacterial peptidoglycan, Science, vol.300, issue.5625, pp.1584-1587, 2003.

S. E. Girardin and I. G. Boneca, Nod2 is a general sensor of peptidoglycan through muramyl dipeptide (MDP) detection, Journal of Biological Chemistry, vol.278, issue.11, pp.8869-8872, 2003.

C. Goudot, Aryl Hydrocarbon Receptor Controls Monocyte Differentiation into Dendritic Cells versus Macrophages, Immunity, vol.47, issue.3, pp.582-596, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01613828

M. Grönlund, Maternal breast-milk and intestinal bifidobacteria guide the compositional development of the Bifidobacterium microbiota in infants at risk of allergic disease, Clinical & Experimental Allergy, vol.37, issue.12, pp.1764-1772, 2007.

G. Duperat and V. , Characterization of the human mucin gene MUC5AC: a consensus cysteine-rich domain for 11p15 mucin genes?, The Biochemical journal, pp.211-220, 1995.

C. W. Ha, Y. Y. Lam, and A. J. Holmes, Mechanistic links between gut microbial community dynamics, microbial functions and metabolic health, World Journal of Gastroenterology, vol.20, issue.44, pp.16498-16517, 2014.

T. Haarmann-stemmann, Analysis of the transcriptional regulation and molecular function of the aryl hydrocarbon receptor repressor in human cell lines. Drug metabolism and disposition: the biological fate of chemicals, vol.35, pp.2262-2269, 2007.

T. Haarmann-stemmann, H. Bothe, and J. Abel, Growth factors, cytokines and their receptors as downstream targets of arylhydrocarbon receptor (AhR) signaling pathways, Biochemical Pharmacology, vol.77, issue.4, pp.508-520, 2009.

K. Haas, Aryl Hydrocarbon Receptor in Keratinocytes Is Essential for Murine Skin Barrier Integrity, Journal of Investigative Dermatology, vol.136, issue.11, pp.2260-2269, 2016.

M. E. Hahn, Aryl hydrocarbon receptors: Diversity and evolution, Chemico-Biological Interactions, vol.141, issue.1-2, pp.131-160, 2002.

M. E. Hahn, L. L. Allan, and D. H. Sherr, Regulation of constitutive and inducible AHR signaling: Complex interactions involving the AHR repressor, 2009.

T. Y. Halim, Lung Natural Helper Cells Are a Critical Source of Th2 Cell-Type Cytokines in Protease Allergen-Induced Airway Inflammation, Immunity, vol.36, issue.3, pp.451-463, 2012.

L. Halme, Family and twin studies in inflammatory bowel disease, World journal of gastroenterology, vol.12, issue.23, pp.3668-72, 2006.

H. Hamada, Identification of multiple isolated lymphoid follicles on the antimesenteric wall of the mouse small intestine, Journal of immunology, vol.168, issue.1, pp.57-64, 1950.

B. Han, Aryl Hydrocarbon Receptor Activation in Intestinal Obstruction Ameliorates Intestinal Barrier Dysfunction Via Suppression of MLCK-MLC Phosphorylation Pathway, SHOCK, vol.46, issue.3, pp.319-328, 2016.

E. H. Han, Endosulfan induces cyp1a1 expression mediated through aryl hydrocarbon receptor signal transduction by protein kinase c, Toxicological Research, vol.31, issue.4, pp.339-345, 2015.

N. Hao, Identification of residues in the N-terminal PAS domains important for dimerization of Arnt and AhR, Nucleic Acids Research, vol.39, issue.9, pp.3695-3709, 2011.

P. A. Harper, C. L. Golas, and A. B. Okey, Characterization of the Ah Receptor and Aryl Hydrocarbon Hydroxylase Induction by 2,3,7,8-Tetrachlorodibenzo-p-dioxin and Benz(a)anthracene in the Human A431 Squamous Cell Carcinoma Line, Cancer Research, vol.48, issue.9, pp.2388-2395, 1988.

T. Hashimoto, ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation, Nature, vol.487, pp.477-483, 2012.

S. Heath-pagliuso, Activation of the Ah receptor by tryptophan and tryptophan metabolites, Biochemistry, vol.37, issue.33, pp.11508-11515, 1998.

S. E. Heid, R. S. Pollenz, and H. I. Swanson, Role of heat shock protein 90 dissociation in mediating agonist-induced activation of the aryl hydrocarbon receptor, Mol Pharmacol, vol.57, issue.1, pp.82-92, 2000.

E. C. Henry, A potential endogenous ligand for the aryl hydrocarbon receptor has potent agonist activity in vitro and in vivo, Archives of Biochemistry and Biophysics, vol.450, issue.1, pp.67-77, 2006.

Y. Higashimura, Propionate promotes fatty acid oxidation through the up-regulation of peroxisome proliferator-activated receptor ? in intestinal epithelial cells, Journal of Nutritional Science and Vitaminology, issue.6, pp.511-515, 2015.

B. Hippe, Faecalibacterium prausnitzii phylotypes in type two diabetic, obese, and lean control subjects, Beneficial Microbes, vol.7, issue.4, pp.511-517, 2016.

P. Holzer, F. Reichmann, and A. Farzi, Neuropeptide Y, peptide YY and pancreatic polypeptide in the gut-brain axis, Neuropeptides, vol.46, issue.6, pp.261-274, 2012.

L. V. Hooper, You AhR what you eat: linking diet and immunity, Cell, vol.147, issue.3, pp.489-91, 2011.

L. Hooper and A. J. Macpherson, Immune adaptations that maintain homeostasis with the intestinal microbiota, Nat Rev Immunol, vol.10, issue.3, pp.159-169, 2010.

T. Hosoya, Inducibility of cytochrome P450 1A1 and chemical carcinogenesis by benzo[a]pyrene in AhR repressor-deficient mice, Biochemical and Biophysical Research Communications, vol.365, issue.3, pp.562-567, 2008.

C. Hou, Intestinal microbiota succession and immunomodulatory consequences after introduction of lactobacillus reuteri I5007 in neonatal piglets, PLoS ONE, vol.10, issue.3, p.119505, 2015.

C. Hsieh, Strengthening of the intestinal epithelial tight junction by Bifidobacterium bifidum, Physiological Reports, vol.3, issue.3, p.12327, 2015.

M. H. Huang, S. T. Lee, and K. Rajendran, A fresh cadaveric study of the paratubal muscles: implications for eustachian tube function in cleft palate, Plastic and reconstructive surgery, vol.100, issue.4, pp.833-875, 1997.

X. Huang, J. A. Powell-coffman, and Y. Jin, The AHR-1 aryl hydrocarbon receptor and its co-factor the AHA-1 aryl hydrocarbon receptor nuclear translocator specify GABAergic neuron cell fate in C. elegans, Development, vol.131, issue.4, pp.819-828, 2004.

T. D. Hubbard, Adaptation of the human aryl hydrocarbon receptor to sense microbiotaderived indoles, Scientific Reports, vol.5, issue.1, p.12689, 2015.

T. Ikuta, Nuclear localization and export signals of the human aryl hydrocarbon receptor, The Journal of biological chemistry, vol.273, issue.5, pp.2895-2904, 1998.

T. Ikuta, Nucleocytoplasmic shuttling of the aryl hydrocarbon receptor, Journal of biochemistry, vol.127, issue.3, pp.503-509, 2000.

T. Ikuta, Y. Kobayashi, and K. Kawajiri, Phosphorylation of nuclear localization signal inhibits the ligand-dependent nuclear import of aryl hydrocarbon receptor, Biochemical and Biophysical Research Communications, vol.317, issue.2, pp.545-550, 2004.

T. Inagaki, Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor, Proceedings of the National Academy of Sciences, vol.103, issue.10, pp.3920-3925, 2006.

A. S. Ismail, intraepithelial lymphocytes are essential mediators of host-microbial homeostasis at the intestinal mucosal surface, Proceedings of the National Academy of Sciences, vol.108, issue.21, pp.8743-8748, 2011.

A. M. Isomäki, A new cell type (tuft cell) in the gastrointestinal mucosa of the rat. A transmission and scanning electron microscopic study. Acta pathologica et microbiologica Scandinavica. Section A, Pathology, vol.240, pp.1-35, 1973.

T. Ito, A Constitutively Active Arylhydrocarbon Receptor Induces Growth Inhibition of Jurkat T Cells through Changes in the Expression of Genes Related to Apoptosis and Cell Cycle Arrest, Journal of Biological Chemistry, vol.279, issue.24, pp.25204-25210, 2004.

N. Iwabuchi, In vitro Th1 cytokine-independent Th2 suppressive effects of bifidobacteria, Microbiology and immunology, vol.51, issue.7, pp.649-60, 2007.

T. Iwanaga, Cellular expression of monocarboxylate transporters (MCT) in the digestive tract of the mouse, rat, and humans, with special reference to slc5a8, Biomedical research, issue.5, pp.243-54, 2006.

S. G. Jeon, Probiotic Bifidobacterium breve Induces IL-10-Producing Tr1 Cells in the Colon, PLoS Pathogens, vol.8, issue.5, p.1002714, 2012.

U. Jin, Microbiome-Derived Tryptophan Metabolites and Their Aryl Hydrocarbon Receptor-Dependent Agonist and Antagonist Activities, Molecular Pharmacology, vol.85, pp.777-788, 2014.

U. Jin, Short Chain Fatty Acids Enhance Aryl Hydrocarbon (Ah) Responsiveness in Mouse Colonocytes and Caco-2 Human Colon Cancer Cells, Scientific Reports, vol.7, issue.1, p.10163, 2017.

A. Jinno, Induction of cytochrome P450-1A by the equine estrogen equilenin, a new endogenous aryl hydrocarbon receptor ligand, Journal of Steroid Biochemistry and Molecular Biology, vol.98, issue.1, pp.48-55, 2006.

M. E. Johansson, The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria, Proceedings of the National Academy of Science, vol.105, issue.39, pp.15064-15073, 2008.

B. V. Jones, Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome, Proceedings of the National Academy of Sciences, vol.105, issue.36, pp.13580-13585, 2008.

S. Jones, An overview of the basic helix-loop-helix proteins, Genome Biology, vol.5, issue.6, p.226, 2004.

L. Jostins, Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease, Nature, vol.491, issue.7422, pp.119-124, 2012.

G. Kakiyama, A simple and accurate HPLC method for fecal bile acid profile in healthy and cirrhotic subjects: validation by GC-MS and LC-MS, Journal of Lipid Research, vol.55, issue.5, pp.978-990, 2014.

H. Kandori, Histochemical, lectin-histochemical and morphometrical characteristics of intestinal goblet cells of germfree and conventional mice, Experimental animals, vol.45, issue.2, pp.155-60, 1996.

A. Kaser, E. Martínez-naves, and R. S. Blumberg, Endoplasmic reticulum stress: implications for inflammatory bowel disease pathogenesis, Current Opinion in Gastroenterology, vol.26, issue.4, pp.318-326, 2010.

I. Kashiwagi, Smad2 and Smad3 Inversely Regulate TGF-? Autoinduction in Clostridium butyricum-Activated Dendritic Cells, vol.43, pp.65-79, 2015.

S. K. Katiyar, M. S. Matsui, and H. Mukhtar, Ultraviolet-B exposure of human skin induces cytochromes P450 1A1 and 1B1, Journal of Investigative Dermatology, vol.114, issue.2, pp.328-333, 2000.

K. Kawajiri, Aryl hydrocarbon receptor suppresses intestinal carcinogenesis in ApcMin/+ mice with natural ligands, Proceedings of the National Academy of Sciences, vol.106, issue.32, pp.13481-13486, 2009.

K. Kawajiri and Y. Fujii-kuriyama, Cytochrome P450 gene regulation and physiological functions mediated by the aryl hydrocarbon receptor, Archives of Biochemistry and Biophysics, vol.464, issue.2, pp.207-212, 2007.

T. Kawasaki and T. Kawai, Toll-like receptor signaling pathways, Frontiers in immunology, vol.5, p.461, 2014.

A. Kazlauskas, L. Poellinger, and I. Pongratz, Evidence that the co-chaperone p23 regulates ligand responsiveness of the dioxin (Aryl hydrocarbon) receptor, Journal of Biological Chemistry, vol.274, issue.19, pp.13519-13543, 1999.

S. Ke, Mechanism of suppression of cytochrome P-450 1A1 expression by tumor necrosis factor-alpha and lipopolysaccharide, The Journal of biological chemistry, vol.276, issue.43, pp.39638-39682, 2001.

L. Kellingray, Consumption of a diet rich in Brassica vegetables is associated with a reduced abundance of sulphate-reducing bacteria: A randomised crossover study, Mol. Nutr. Food Reyes, vol.61, issue.9, p.1600992, 2017.

N. I. Kerkvliet, AHR-mediated immunomodulation: The role of altered gene transcription, Biochemical Pharmacology, vol.77, pp.746-760, 2009.

R. J. Kewley, M. L. Whitelaw, and A. Chapman-smith, The mammalian basic helix-loop-helix/PAS family of transcriptional regulators, The International Journal of Biochemistry & Cell Biology, vol.36, issue.36, pp.189-204, 2004.

A. Khosravi, Gut microbiota promote hematopoiesis to control bacterial infection, Cell host & microbe, vol.15, issue.3, pp.374-81, 2014.

C. J. Kim, l-Tryptophan exhibits therapeutic function in a porcine model of dextran sodium sulfate (DSS)-induced colitis, The Journal of Nutritional Biochemistry, vol.21, issue.6, pp.468-475, 2010.

D. W. Kim, The RelA NF-kappaB subunit and the aryl hydrocarbon receptor (AhR) cooperate to transactivate the c-myc promoter in mammary cells, Oncogene, vol.19, issue.48, pp.5498-5506, 2000.

A. Kimura, Aryl hydrocarbon receptor in combination with Stat1 regulates LPS-induced inflammatory responses, Journal of Experimental Medicine, vol.206, issue.9, pp.2027-2035, 2009.

A. Kimura, Aryl hydrocarbon receptor regulates Stat1 activation and participates in the development of Th17 cells, Proceedings of the National Academy of Sciences, vol.105, issue.28, pp.9721-9726, 2008.

E. A. Kiss, Natural Aryl Hydrocarbon Receptor Ligands Control Organogenesis of Intestinal Lymphoid Follicles, Science, vol.334, issue.6062, pp.1561-1565, 2011.

S. De-kivit, Intestinal Epithelium-Derived Galectin-9 Is Involved in the Immunomodulating Effects of Nondigestible Oligosaccharides, Journal of Innate Immunity, vol.5, issue.6, pp.625-638, 2013.

E. S. Klaassens, Mixed-Species Genomic Microarray Analysis of Fecal Samples Reveals Differential Transcriptional Responses of Bifidobacteria in Breast-and Formula-Fed Infants, Applied and Environmental Microbiology, vol.75, issue.9, pp.2668-2676, 2009.

H. P. Ko, Transactivation domains facilitate promoter occupancy for the dioxin-inducible CYP1A1 gene in vivo, Molecular and cellular biology, vol.17, issue.7, pp.3497-3507, 1997.

K. S. Kobayashi, Nod2-Dependent Regulation of Innate and Adaptive Immunity in the Intestinal Tract, Advancement Of Science, vol.307, issue.5710, pp.731-734, 2005.

R. Koeth, Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis, Nature Medicine, vol.19, issue.5, pp.576-585, 2013.

M. Korkalainen, J. Tuomisto, and R. Pohjanvirta, The AH receptor of the most dioxin-sensitive species, guinea pig, is highly homologous to the human AH receptor, Biochemical and biophysical research communications, vol.285, issue.5, pp.1121-1130, 2001.

S. Koyasu and K. Moro, Role of innate lymphocytes in infection and inammation, Frontiers in Immunology, vol.3, p.101, 2012.

M. B. Kumar, The Q-rich Subdomain of the Human Ah Receptor Transactivation Domain Is Required for Dioxin-mediated Transcriptional Activity, Journal of Biological Chemistry, vol.276, issue.45, pp.42302-42310, 2001.

K. Kurokawa, Comparative Metagenomics Revealed Commonly Enriched Gene Sets in Human Gut Microbiomes, DNA Research, vol.14, issue.4, pp.169-181, 2007.

J. Lagier, Microbial culturomics: paradigm shift in the human gut microbiome study. Clinical microbiology and infection : the official publication of the European Society of Clinical Microbiology and Infectious Diseases, vol.18, pp.1185-93, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00964466

B. Lamas, CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands, Nature medicine, vol.22, issue.6, pp.598-605, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02785302

B. Lamas, J. M. Natividad, and H. Sokol, Aryl hydrocarbon receptor and intestinal immunity, Mucosal Immunology, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02625696

E. Larsson, Analysis of gut microbial regulation of host gene expression along the length of the gut and regulation of gut microbial ecology through MyD88, Gut, issue.8, pp.1124-1155, 2012.

A. J. Lee, Characterization of the Oxidative Metabolites of 17?-Estradiol and Estrone Formed by 15 Selectively Expressed Human Cytochrome P450 Isoforms, Endocrinology, vol.144, issue.8, pp.3382-3398, 2003.

J. Lee, Maintenance of colonic homeostasis by distinctive apical TLR9 signalling in intestinal epithelial cells, Nature Cell Biology, vol.8, issue.12, pp.1327-1336, 2006.

J. H. Lee, T. K. Wood, and J. Lee, Roles of indole as an interspecies and interkingdom signaling molecule, Trends in Microbiology, vol.23, issue.11, pp.707-718, 2015.

J. S. Lee, AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch, Nat Immunol, vol.13, issue.2, pp.144-151, 2012.

M. J. Lees, D. J. Peet, and M. L. Whitelaw, Defining the role for XAP2 in stabilization of the dioxin receptor, Journal of Biological Chemistry, vol.278, issue.38, pp.35878-35888, 2003.

P. Lepage, Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis, Gastroenterology, vol.141, issue.1, pp.227-236, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01004334

R. E. Ley, Obesity alters gut microbial ecology, Proceedings of the National Academy of Sciences of the United States of America, vol.102, pp.11070-11075, 2005.

H. Li, The outer mucus layer hosts a distinct intestinal microbial niche, Nature Communications, vol.6, issue.1, p.8292, 2015.

J. Li, An integrated catalog of reference genes in the human gut microbiome, Nature Biotechnology, vol.32, issue.8, pp.834-841, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01195478

X. Li, Toll-like receptor 4 increases intestinal permeability through up-regulation of membrane PKC activity in alcoholic steatohepatitis, Alcohol, vol.47, issue.6, pp.459-465, 2013.

Y. Li, Exogenous Stimuli Maintain Intraepithelial Lymphocytes via Aryl Hydrocarbon Receptor Activation, Cell, vol.147, issue.3, pp.629-640, 2011.

Z. Liu, Aryl hydrocarbon receptor activation maintained the intestinal epithelial barrier function through Notch1 dependent signaling pathway, International Journal of Molecular Medicine, vol.41, issue.3, pp.1560-1572, 2018.

M. M. Lowe, Identification of cinnabarinic acid as a novel endogenous aryl hydrocarbon receptor ligand that drives IL-22 production, PLoS ONE, vol.9, issue.2, p.87877, 2014.

C. A. Lozupone, Diversity, stability and resilience of the human gut microbiota, Nature, vol.489, issue.7415, pp.220-230, 2012.

A. E. M'koma, Inflammatory bowel disease: an expanding global health problem, Clinical medicine insights. Gastroenterology, vol.6, pp.33-47, 2013.

Q. Ma and K. T. Baldwin, 8-tetrachlorodibenzo-p-dioxin-induced degradation of aryl hydrocarbon receptor (AhR) by the ubiquitin-proteasome pathway. Role of the transcription activaton and DNA binding of AhR, The Journal of biological chemistry, vol.7, issue.12, pp.8432-8440, 2000.

Q. Ma and J. P. Whitlock, A novel cytoplasmic protein that interacts with the Ah receptor, contains tetratricopeptide repeat motifs, and augments the transcriptional response to 2,3,7,8-tetrachlorodibenzo-p-dioxin, The Journal of biological chemistry, vol.272, issue.14, pp.8878-84, 1997.

Q. Ma and J. P. Whitlock, The aromatic hydrocarbon receptor modulates the Hepa 1c1c7 cell cycle and differentiated state independently of dioxin, Molecular and cellular biology, vol.16, issue.5, pp.2144-50, 1996.

A. J. Macpherson and T. Uhr, Induction of Protective IgA by Intestinal Dendritic Cells Carrying Commensal Bacteria, Science, vol.303, issue.5664, pp.1662-1665, 2004.

L. Macpherson, Aryl hydrocarbon receptor repressor and TiPARP (ARTD14) use similar, but also distinct mechanisms to repress aryl hydrocarbon receptor signaling, International journal of molecular sciences, vol.15, issue.5, pp.7939-57, 2014.

P. Magiatis, Malassezia Yeasts Produce a Collection of Exceptionally Potent Activators of the Ah (Dioxin) Receptor Detected in Diseased Human Skin, Journal of Investigative Dermatology, vol.133, issue.8, pp.2023-2030, 2013.

D. K. Manchester, Ah receptor in human placenta: stabilization by molybdate and characterization of binding of 2,3,7,8-tetrachlorodibenzo-p-dioxin, 3-methylcholanthrene, and benzo(a)pyrene, Cancer research, vol.47, issue.18, pp.4861-4869, 1987.

C. Manichanh, Reduced diversity of faecal microbiota in Crohn's disease revealed by a metagenomic approach, Gut, vol.55, issue.2, pp.205-216, 2006.

A. Marcobal, A refined palate: Bacterial consumption of host glycans in the gut, Glycobiology, vol.23, issue.9, pp.1038-1046, 2013.

C. Martin-gallausiaux, Butyrate from commensal bacteria activates TGF-beta1 expression through the transcription factor SP1 in human intestinal epithelial cells

K. M. Maslowski, Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43, Nature, issue.7268, pp.1282-1286, 2009.

M. J. May and S. Ghosh, Signal transduction through NF-kappa B, Immunology today, vol.19, issue.2, pp.80-88, 1998.

O. Ménard, Gnotobiotic mouse immune response induced by Bifidobacterium sp. strains isolated from infants, Applied and environmental microbiology, vol.74, issue.3, pp.660-666, 2008.

N. Mexia, Pityriazepin and other potent AhR ligands isolated from Malassezia furfur yeast, Archives of Biochemistry and Biophysics, vol.571, pp.16-20, 2015.

B. K. Meyer, Hepatitis B virus X-associated protein 2 is a subunit of the unliganded aryl hydrocarbon receptor core complex and exhibits transcriptional enhancer activity, Molecular and cellular biology, vol.18, issue.2, pp.978-88, 1998.

B. K. Meyer and G. H. Perdew, Characterization of the AhR-hsp90-XAP2 core complex and the role of the immunophilin-related protein XAP2 in AhR stabilization, Biochemistry, vol.38, issue.28, pp.8907-8917, 1999.

C. A. Miller, Expression of the human aryl hydrocarbon receptor complex in yeast. Activation of transcription by indole compounds, The Journal of biological chemistry, vol.272, issue.52, pp.32824-32833, 1997.

J. Mimura, Identification of a novel mechanism of regulation of Ah (dioxin) receptor function, Genes & development, vol.13, issue.1, pp.20-25, 1999.

J. Mimura, Loss of teratogenic response to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in mice lacking the Ah (dioxin) receptor. Genes to cells : devoted to molecular & cellular mechanisms, vol.2, pp.645-654, 2003.

K. A. Mitchell and C. J. Elferink, Timing is everything: Consequences of transient and sustained AhR activity, Biochemical Pharmacology, vol.77, issue.6, pp.947-956, 2009.

I. Monteleone, Aryl hydrocarbon receptor-induced signals up-regulate IL-22 production and inhibit inflammation in the gastrointestinal tract, 2011.

L. A. Monticelli, Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus, Nature immunology, vol.12, issue.11, pp.1045-54, 2011.

P. Moura-alves, AhR sensing of bacterial pigments regulates antibacterial defence, Nature, vol.512, pp.387-392, 2014.

S. Mueller, Differences in Fecal Microbiota in Different European Study Populations in Relation to Age, Gender, and Country: a Cross-Sectional Study, Applied and Environmental Microbiology, vol.72, issue.2, pp.1027-1033, 2006.
URL : https://hal.archives-ouvertes.fr/hal-02667185

I. A. Murray and G. H. Perdew, Ligand activation of the Ah receptor contributes to gastrointestinal homeostasis. Current opinion in toxicology, vol.2, pp.15-23, 2017.

D. W. Nebert, The in vivo and in vitro induction of aryl hydrocarbon hydroxylase in mammalian cells of different species, tissues, strains, and developmental and hormonal states, Archives of biochemistry and biophysics, vol.134, issue.1, pp.76-89, 1969.

D. W. Nebert, K. Wikvall, and W. L. Miller, Human cytochromes P450 in health and disease, Philosophical transactions of the Royal Society of London. Series B, Biological sciences, vol.368, p.20120431, 1612.

D. R. Neill, Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity, Nature, vol.464, issue.7293, pp.1367-1370, 2010.

D. R. Nelson, Comparison of cytochrome P450 (CYP) genes from the mouse and human genomes, including nomenclature recommendations for genes, pseudogenes and alternative-splice variants, Pharmacogenetics, vol.14, issue.14, pp.1-181, 2004.

M. Nepelska, Commensal gut bacteria modulate phosphorylation-dependent PPAR? transcriptional activity in human intestinal epithelial cells, Scientific Reports, vol.7, p.43199, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02628360

M. R. Neutra, A. Frey, and J. P. Kraehenbuhl, Epithelial M cells: gateways for mucosal infection and immunization, Cell, vol.86, issue.3, pp.345-353, 1996.

N. T. Nguyen, Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism, Proceedings of the National Academy of Sciences, vol.107, issue.46, pp.19961-19966, 2010.

L. Nibali and B. Henderson, The Human Microbiota and Chronic Disease, 2016.

G. Nigro, The cytosolic bacterial peptidoglycan sensor Nod2 affords stem cell protection and links microbes to gut epithelial regeneration, Cell host & microbe, vol.15, issue.6, pp.792-800, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01950138

S. Nishiumi, K. Yoshida, and H. Ashida, Curcumin suppresses the transformation of an aryl hydrocarbon receptor through its phosphorylation, Archives of Biochemistry and Biophysics, vol.466, issue.2, pp.267-273, 2007.

M. C. Noverr, Development of allergic airway disease in mice following antibiotic therapy and fungal microbiota increase: role of host genetics, antigen, and interleukin-13, Infection and immunity, vol.73, issue.1, pp.30-38, 2005.

J. Ochoa-reparaz, Central Nervous System Demyelinating Disease Protection by the Human Commensal Bacteroides fragilis Depends on Polysaccharide A Expression, The Journal of Immunology, vol.185, issue.7, pp.4101-4108, 2010.

F. Ohtake, Dioxin receptor is a ligand-dependent E3 ubiquitin ligase, Nature, vol.446, issue.7135, pp.562-566, 2007.

F. Ohtake, Y. Fujii-kuriyama, and S. Kato, AhR acts as an E3 ubiquitin ligase to modulate steroid receptor functions, Biochemical Pharmacology, vol.77, issue.4, pp.474-484, 2009.

M. Oshima, SUMO modification regulates the transcriptional repressor function of aryl hydrocarbon receptor repressor, The Journal of biological chemistry, vol.284, issue.17, pp.11017-11043, 2009.

J. Otte, E. Cario, and D. K. Podolsky, Mechanisms of cross hyporesponsiveness to Toll-like receptor bacterial ligands in intestinal epithelial cells, Gastroenterology, vol.126, issue.4, pp.1054-70, 2004.

R. L. Owen, A. J. Piazza, and T. H. Ermak, Ultrastructural and cytoarchitectural features of lymphoreticular organs in the colon and rectum of adult BALB/c mice, American Journal of Anatomy, vol.190, issue.1, pp.10-18, 1991.

R. J. Pennington, The metabolism of short-chain fatty acids in the sheep. I. Fatty acid utilization and ketone body production by rumen epithelium and other tissues, The Biochemical journal, vol.51, issue.2, pp.251-259, 1952.

G. H. Perdew and C. A. Bradfield, Mapping the 90 kDa heat shock protein binding region of the Ah receptor, Biochemistry and molecular biology international, vol.39, issue.3, pp.589-93, 1996.

J. C. Peters, Tryptophan nutrition and metabolism: an overview. Advances in experimental medicine and biology, vol.294, pp.345-58, 1991.

J. M. Peters and L. M. Wiley, Evidence That Murine Preimplantation Embryos Express Aryl Hydrocarbon Receptor, Toxicology and Applied Pharmacology, vol.134, issue.2, pp.214-221, 1995.

P. I. Petkov, Mechanism-based common reactivity pattern (COREPA) modelling of aryl hydrocarbon receptor binding affinity, SAR and QSAR in environmental research, vol.21, issue.1, pp.187-214, 2010.

J. R. Petrulis, The hsp90 Co-chaperone XAP2 Alters Importin beta Recognition of the Bipartite Nuclear Localization Signal of the Ah Receptor and Represses Transcriptional Activity, Journal of Biological Chemistry, vol.278, issue.4, pp.2677-2685, 2003.

A. Poland and E. Glover, Characterization and strain distribution pattern of the murine Ah receptor specified by the Ahd and Ahb-3 alleles, Molecular pharmacology, vol.38, issue.3, pp.306-318, 1990.

A. Poland and E. Glover, Chlorinated dibenzo-p-dioxins: potent inducers of delta-aminolevulinic acid synthetase and aryl hydrocarbon hydroxylase. II. A study of the structure-activity relationship, Molecular pharmacology, vol.9, issue.6, pp.736-783, 1973.

A. Poland and J. C. Knutson, 8-Tetrachlorodibenzo-p -Dioxin and Related Halogenated Aromatic Hydrocarbons: Examination of the Mechanism of Toxicity, Annual Review of Pharmacology and Toxicology, vol.7, issue.1, pp.517-554, 1982.

A. P. Poland, Genetic expression of aryl hydrocarbon hydroxylase activity. Induction of monooxygenase activities and cytochrome P1-450 formation by 2,3,7,8 tetrachlorodibenzo p dioxin in mice genetically "nonresponsive" to other aromatic hydrocarbons, Journal of Biological Chemistry, vol.249, issue.17, pp.5599-5606, 1974.

R. S. Pollenz, C. A. Sattler, and A. Poland, The aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator protein show distinct subcellular localizations in Hepa 1c1c7 cells by immunofluorescence microscopy, Molecular pharmacology, vol.45, issue.3, pp.428-438, 1994.

I. Pongratz, Role of the PAS domain in regulation of dimerization and DNA binding specificity of the dioxin receptor, Molecular and cellular biology, vol.18, issue.7, pp.4079-88, 1998.

I. Pongratz, G. G. Mason, and L. Poellinger, Dual roles of the 90-kDa heat shock protein hsp90 in modulating functional activities of the dioxin receptor. Evidence that the dioxin receptor functionally belongs to a subclass of nuclear receptors which require hsp90 both for ligand binding activity and repression of intrinsic DNA binding activity, The Journal of biological chemistry, vol.267, issue.19, pp.13728-13762, 1992.

E. M. Porter, The multifaceted Paneth cell. Cellular and molecular life sciences : CMLS, vol.59, pp.156-70, 2002.

L. Poul and E. , Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation, The Journal of biological chemistry, vol.278, issue.28, pp.25481-25490, 2003.

A. Puga, C. Ma, and J. L. Marlowe, The aryl hydrocarbon receptor cross-talks with multiple signal transduction pathways, Biochemical Pharmacology, vol.77, issue.4, pp.713-722, 2009.

J. Purushe, Comparative Genome Analysis of Prevotella ruminicola and Prevotella bryantii: Insights into Their Environmental Niche, Microbial Ecology, vol.60, issue.4, pp.721-729, 2010.

J. Qin, A human gut microbial gene catalog established by metagenomic sequencing, Nature, vol.464, issue.7285, pp.59-65, 2010.

J. Qiu, Group 3 Innate Lymphoid Cells Inhibit T-Cell-Mediated Intestinal Inflammation through Aryl Hydrocarbon Receptor Signaling and Regulation of Microflora, Immunity, vol.39, issue.2, pp.386-399, 2013.

J. Qiu, The Aryl Hydrocarbon Receptor Regulates Gut Immunity through Modulation of Innate Lymphoid Cells, Immunity, vol.36, issue.1, pp.92-104, 2012.

J. Qui, Aryl Hydrocarbon Receptor Promotes RORyt+ ILCs and Controls Intestinal Immunity and Inflammation, Semin Immunopathol, vol.35, issue.6, pp.657-670, 2013.

F. J. Quintana, Control of Treg and TH17 cell differentiation by the aryl hydrocarbon receptor, Nature, vol.453, issue.7191, pp.65-71, 2008.

A. Rannug, Certain photooxidized derivatives of tryptophan bind with very high affinity to the Ah receptor and are likely to be endogenous signal substances, The Journal of biological chemistry, vol.262, issue.32, pp.15422-15429, 1987.

U. Rannug, Structure elucidation of two tryptophan-derived, high affinity Ah receptor ligands, Chemistry & biology, vol.2, issue.12, pp.841-846, 1995.

M. K. Rasmussen, Skatole (3-Methylindole) Is a Partial Aryl Hydrocarbon Receptor Agonist and Induces CYP1A1/2 and CYP1B1 Expression in Primary Human Hepatocytes, PloS one, vol.11, issue.5, p.154629, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01837192

A. Rehman, Nod2 is essential for temporal development of intestinal microbial communities, Gut, vol.60, issue.10, pp.1354-1362, 2011.

M. Rescigno, Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria, Nature immunology, vol.2, issue.4, pp.361-368, 2001.

V. K. Ridaura, Gut Microbiota from Twins Discordant for Obesity Modulate Metabolism in Mice, Science, vol.341, issue.6150, pp.1241214-1241214, 2013.

K. A. Romano, Intestinal Microbiota Composition Modulates Choline Bioavailability from Diet and Accumulation of the Proatherogenic Metabolite Trimethylamine-N-Oxide, mBio, vol.6, issue.2, 2015.

E. Romeo, Luminal kidney and intestine SLC6 amino acid transporters of B0AT-cluster and their tissue distribution in Mus musculus, American journal of physiology. Renal physiology, vol.290, issue.2, pp.376-383, 2006.

J. L. Round, The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota, Science, issue.6032, pp.974-981, 2011.

S. L. Russell, Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic asthma, EMBO reports, vol.13, issue.5, pp.440-447, 2012.

S. Safe, Dibenzofurans (PCDFs), and Related Compounds: Environmental and Mechanistic Considerations Which Support the Development of Toxic Equivalency Factors (TEFs), Dibenzo-p-Dioxins (PCDDs), vol.21, pp.51-88, 1990.

S. Sakurai, T. Shimizu, and U. Ohto, The crystal structure of the AhRR-ARNT heterodimer reveals the structural basis of the repression of AhR-mediated transcription, Journal of Biological Chemistry, vol.292, issue.43, pp.17609-17616, 2017.

O. V. Savinova, A. Hoffmann, and G. Ghosh, The Nf?b1 and Nf?b2 proteins p105 and p100 function as the core of high-molecular-weight heterogeneous complexes, Molecular cell, vol.34, issue.5, pp.591-602, 2009.

C. M. Schaldach, J. Riby, and L. F. Bjeldanes, Lipoxin A 4 : A New Class of Ligand for the Ah Receptor, Biochemistry, vol.38, issue.23, pp.7594-7600, 1999.

C. Schiering, Feedback control of AHR signalling regulates intestinal immunity, Nature, vol.542, issue.7640, pp.242-245, 2017.

R. Schilderink, C. Verseijden, and W. J. De-jonge, Dietary Inhibitors of Histone Deacetylases in Intestinal Immunity and Homeostasis, Frontiers in Immunology, vol.4, p.226, 2013.

J. C. Schroeder, The Uremic Toxin 3-Indoxyl Sulfate Is a Potent Endogenous Agonist for the Human Aryl Hydrocarbon Receptor, Biochemistry, vol.49, issue.2, pp.393-400, 2010.

E. Segura, Human Inflammatory Dendritic Cells Induce Th17 Cell Differentiation, Immunity, vol.38, issue.2, pp.336-348, 2013.

M. Seliskar and D. Rozman, Mammalian cytochromes P450-Importance of tissue specificity, Biochimica et Biophysica Acta (BBA) -General Subjects, vol.1770, issue.3, pp.458-466, 2007.

S. Seok, Structural hierarchy controlling dimerization and target DNA recognition in the AHR transcriptional complex, vol.114, pp.5431-5436, 2017.

F. Shanahan, The host-microbe interface within the gut. Best practice & research, Clinical gastroenterology, vol.16, issue.6, pp.915-946, 2002.

E. S. Shen and J. P. Whitlock, Protein-DNA interactions at a dioxin-responsive enhancer. Mutational analysis of the DNA-binding site for the liganded Ah receptor, Journal of Biological Chemistry, vol.267, issue.10, pp.6815-6819, 1992.

S. Shimba, Transcriptional regulation of the AhR gene during adipose differentiation, Biological & pharmaceutical bulletin, vol.26, issue.9, pp.1266-71, 2003.

J. H. Shin, Modulation of natural killer cell antitumor activity by the aryl hydrocarbon receptor, Proceedings of the National Academy of Sciences, vol.110, issue.30, pp.12391-12396, 2013.

N. Shulzhenko, Crosstalk between B lymphocytes, microbiota and the intestinal epithelium governs immunity versus metabolism in the gut, Nature Medicine, vol.17, issue.12, pp.1585-1593, 2011.

G. M. Simon, J. Cheng, and J. I. Gordon, Quantitative assessment of the impact of the gut microbiota on lysine -acetylation of host proteins using gnotobiotic mice, Proceedings of the National Academy of Sciences, vol.109, issue.28, pp.11133-11138, 2012.

C. J. Sinal and J. R. Bend, Aryl hydrocarbon receptor-dependent induction of cyp1a1 by bilirubin in mouse hepatoma hepa 1c1c7 cells, Molecular pharmacology, vol.52, issue.4, pp.590-599, 1997.

M. Van-der-sluis, Muc2-Deficient Mice Spontaneously Develop Colitis, Indicating That MUC2 Is Critical for Colonic Protection, Gastroenterology, vol.131, issue.1, pp.117-129, 2006.

B. W. Smith, The aryl hydrocarbon receptor directs hematopoietic progenitor cell expansion and differentiation, Blood, vol.122, issue.3, pp.376-85, 2013.

E. A. Smith and G. T. Macfarlane, Formation of Phenolic and Indolic Compounds by Anaerobic Bacteria in the Human Large Intestine, Microbial Ecology, vol.33, issue.3, pp.180-188, 1997.

P. M. Smith, The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis, Science, issue.6145, pp.569-73, 2013.

H. Sokol, Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients, Proceedings of the National Academy of Sciences of the United States of America, vol.105, pp.16731-16737, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00652961

J. Song, A ligand for the aryl hydrocarbon receptor isolated from lung, Proceedings of the National Academy of Sciences of the United States of America, vol.99, pp.14694-14703, 2002.

G. F. Sonnenberg, Innate Lymphoid Cells Promote Anatomical Containment of Lymphoid-Resident Commensal Bacteria, Science, vol.336, issue.6086, pp.1321-1325, 2012.

R. Sonowal, Indoles from commensal bacteria extend healthspan, Proceedings of the National Academy of Sciences, vol.114, pp.7506-7515, 2017.

A. A. Soshilov and M. S. Denison, Ligand promiscuity of aryl hydrocarbon receptor agonists and antagonists revealed by site-directed mutagenesis, Molecular and cellular biology, vol.34, issue.9, pp.1707-1719, 2014.

J. Spencer, Gamma/delta T cells and the diagnosis of coeliac disease, Clinical and experimental immunology, vol.85, issue.1, pp.109-122, 1991.

B. Stockinger, The Aryl Hydrocarbon Receptor: Multitasking in the Immune System, Annual Review of Immunology, vol.32, issue.1, pp.403-432, 2014.

H. I. Swanson, DNA binding and protein interactions of the AHR/ARNT heterodimer that facilitate gene activation, Chemico-biological interactions, vol.141, issue.1-2, pp.63-76, 2002.

H. I. Swanson and J. H. Yang, The aryl hydrocarbon receptor interacts with transcription factor IIB, Molecular pharmacology, vol.54, issue.4, pp.671-678, 1998.

E. D. Tait-wojno and D. Artis, Innate Lymphoid Cells: Balancing Immunity, Inflammation, and Tissue Repair in the Intestine, Cell Host & Microbe, vol.12, issue.4, pp.445-457, 2012.

T. Takamura, Lactobacillus bulgaricus OLL1181 activates the aryl hydrocarbon receptor pathway and inhibits colitis, Immunology and Cell Biology, vol.89, issue.7, pp.817-822, 2011.

M. Tan, Identification of 67 Histone Marks and Histone Lysine Crotonylation as a New Type of Histone Modification, Cell, vol.146, issue.6, pp.1016-1028, 2011.

X. Tan, Indoxyl sulfate, a valuable biomarker in chronic kidney disease and dialysis, Hemodialysis International, vol.21, issue.2, pp.161-167, 2017.

J. Tap, Towards the human intestinal microbiota phylogenetic core, Environmental microbiology, vol.11, issue.10, pp.2574-84, 2009.
URL : https://hal.archives-ouvertes.fr/hal-02667864

M. Thangaraju, Transport by SLC5A8 with subsequent inhibition of histone deacetylase 1 (HDAC1) and HDAC3 underlies the antitumor activity of 3-bromopyruvate, Cancer, vol.115, issue.20, pp.4655-4666, 2009.

K. T. Thia, An Update on the Epidemiology of Inflammatory Bowel Disease in Asia, The American Journal of Gastroenterology, vol.103, issue.12, pp.3167-3182, 2008.

L. V. Thomas and T. Ockhuizen, New insights into the impact of the intestinal microbiota on health and disease: a symposium report, British Journal of Nutrition, vol.107, issue.S1, pp.1-13, 2012.

Y. Tian, Ah receptor and NF-kappaB interactions, a potential mechanism for dioxin toxicity, The Journal of biological chemistry, vol.274, issue.1, pp.510-515, 1999.

N. Tijet, Aryl hydrocarbon receptor regulates distinct dioxin-dependent and dioxinindependent gene batteries, Molecular pharmacology, vol.69, issue.1, pp.140-53, 2006.

A. Tkachenko, The Q-rich/PST domain of the AHR regulates both ligand-induced nuclear transport and nucleocytoplasmic shuttling, Scientific Reports, vol.6, issue.1, p.32009, 2016.

Y. Tsuchiya, Expression of aryl hydrocarbon receptor repressor in normal human tissues and inducibility by polycyclic aromatic hydrocarbons in human tumor-derived cell lines, Toxicological Sciences, vol.72, issue.2, pp.253-259, 2003.

P. J. Turnbaugh, A core gut microbiome in obese and lean twins, Nature, vol.457, issue.7228, pp.480-484, 2009.

M. Veldhoen, Natural agonists for aryl hydrocarbon receptor in culture medium are essential for optimal differentiation of Th17 T cells, The Journal of experimental medicine, vol.206, issue.1, pp.43-49, 2009.

M. Veldhoen, The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins, Nature, vol.453, issue.7191, pp.106-109, 2008.

M. Venkatesh, Symbiotic Bacterial Metabolites Regulate Gastrointestinal Barrier Function via the Xenobiotic Sensor PXR and Toll-like Receptor 4, Immunity, vol.41, issue.2, pp.296-310, 2014.

L. Vikström-bergander, Tryptamine serves as a proligand of the AhR transcriptional pathway whose activation is dependent of monoamine oxidases, Molecular endocrinology, vol.26, issue.9, pp.1542-51, 2012.

M. Villa, Aryl hydrocarbon receptor is required for optimal B-cell proliferation, The EMBO journal, vol.36, issue.1, pp.116-128, 2017.

C. F. Vogel, RelB, a new partner of aryl hydrocarbon receptor-mediated transcription, Molecular endocrinology, vol.21, issue.12, pp.2941-2955, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00268250

R. Vrzal, Activated thyroid hormone receptor modulates dioxin-inducible aryl hydrocarbon receptor-mediated CYP1A1 induction in human hepatocytes but not in human hepatocarcinoma HepG2 cells, Toxicology Letters, vol.275, pp.77-82, 2017.

F. Wang, Gut Microbiota Community and Its Assembly Associated with Age and Diet in Chinese Centenarians, Journal of Microbiology and Biotechnology, vol.25, issue.8, pp.1195-1204, 2015.

I. Wang, The effect of probiotics on serum levels of cytokine and endotoxin in peritoneal dialysis patients: a randomised, double-blind, placebo-controlled trial, Beneficial Microbes, vol.6, issue.4, pp.423-430, 2015.

Z. Wang, Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease, Nature, vol.472, pp.57-63, 2011.

K. D. Wansa, The AF-1 domain of the orphan nuclear receptor NOR-1 mediates transactivation, coactivator recruitment, and activation by the purine anti-metabolite 6-mercaptopurine, The Journal of biological chemistry, vol.278, issue.27, pp.24776-90, 2003.

Y. D. Wei, Rapid and transient induction of CYP1A1 gene expression in human cells by the tryptophan photoproduct 6-formylindolo[3,2-b]carbazole, Chemico-Biological Interactions, vol.110, issue.1-2, pp.39-55, 1998.

K. E. Wellen and G. S. Hotamisligil, Inflammation, stress, and diabetes, The Journal of clinical investigation, vol.115, issue.5, pp.1111-1120, 2005.

M. F. Wernet, Stochastic spineless expression creates the retinal mosaic for colour vision, Nature, vol.440, issue.7081, pp.174-180, 2006.

W. R. Wikoff, Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites, Proceedings of the National Academy of Sciences of the United States of America, vol.106, pp.3698-3703, 2009.

J. M. Wong, Colonic health: fermentation and short chain fatty acids, Journal of clinical gastroenterology, vol.40, issue.3, pp.235-278, 2006.

P. S. Wong, Arylhydrocarbon receptor activation in NCI-H441 cells and C57BL/6 mice: possible mechanisms for lung dysfunction, American journal of respiratory cell and molecular biology, vol.42, issue.2, pp.210-217, 2010.

G. D. Wu, Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes, Science, vol.334, issue.6052, pp.105-108, 2011.

H. Wu, Gut-Residing Segmented Filamentous Bacteria Drive Autoimmune Arthritis via T Helper 17 Cells, Immunity, vol.32, issue.6, pp.815-827, 2010.

Z. Xie, Metabolic Regulation of Gene Expression by Histone Lysine ?-Hydroxybutyrylation, Molecular Cell, vol.62, issue.2, pp.194-206, 2016.

S. Yamaori, Cannabidiol induces expression of human cytochrome P450 1A1 that is possibly mediated through aryl hydrocarbon receptor signaling in HepG2 cells, Life Sciences, vol.136, pp.87-93, 2015.

T. Yatsunenko, Human gut microbiome viewed across age and geography, Nature, vol.486, issue.7402, pp.222-229, 2012.

J. Ye, The Aryl Hydrocarbon Receptor Preferentially Marks and Promotes Gut Regulatory T Cells, Cell reports, vol.21, issue.8, pp.2277-2290, 2017.

V. B. Young and T. M. Schmidt, Antibiotic-associated diarrhea accompanied by large-scale alterations in the composition of the fecal microbiota, Journal of clinical microbiology, vol.42, issue.3, pp.1203-1209, 2004.

M. Yu, Aryl Hydrocarbon Receptor Activation Modulates Intestinal Epithelial Barrier Function by Maintaining Tight Junction Integrity, International Journal of Biological Sciences, vol.14, issue.1, pp.69-77, 2018.

S. H. Zeisel and K. A. Da-costa, Choline: An essential nutrient for public health, Nutrition Reviews, vol.67, pp.615-623, 2009.

T. Zelante, Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22, Immunity, vol.39, issue.6, pp.372-385, 2013.

L. A. Zenewicz, Innate and adaptive interleukin-22 protects mice from inflammatory bowel disease, Immunity, vol.29, issue.6, pp.947-57, 2008.

S. Zhang, C. Qin, and S. H. Safe, Flavonoids as aryl hydrocarbon receptor agonists/antagonists: effects of structure and cell context, Environmental health perspectives, vol.111, issue.16, pp.1877-82, 2003.

L. Zheng, Microbial-Derived Butyrate Promotes Epithelial Barrier Function through IL-10 Receptor-Dependent Repression of Claudin-2, The Journal of Immunology, p.1700105, 2017.

J. Zimmer, A vegan or vegetarian diet substantially alters the human colonic faecal microbiota, European Journal of Clinical Nutrition, vol.66, issue.1, pp.53-60, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00665871

C. L. Zindl, IL-22-producing neutrophils contribute to antimicrobial defense and restitution of colonic epithelial integrity during colitis, Proceedings of the National Academy of Sciences of the United States of America, vol.110, pp.12768-73, 2013.

E. G. Zoetendal, Mucosa-associated bacteria in the human gastrointestinal tract are uniformly distributed along the colon and differ from the community recovered from feces, Applied and environmental microbiology, vol.68, issue.7, pp.3401-3408, 2002.

E. G. Zoetendal, The human small intestinal microbiota is driven by rapid uptake and conversion of simple carbohydrates, The ISME Journal, vol.6, issue.7, pp.1415-1426, 2012.