E. Stolarczyk, Adipose tissue inflammation in obesity: a metabolic or immune response?, Current Opinion in Pharmacology, vol.37, pp.35-40, 2017.

S. M. Reilly and A. R. Saltiel, Adapting to obesity with adipose tissue inflammation, Nature Reviews Endocrinology, vol.13, issue.11, pp.633-643, 2017.

A. K. Loomis, S. Kabadi, D. Preiss, C. Hyde, V. Bonato et al., Body mass index and risk of nonalcoholic fatty liver disease: two electronic health record prospective studies, Journal of Clinical Endocrinology & Metabolism, vol.101, issue.3, pp.945-952, 2016.

M. Monteiro-sepulveda, S. Touch, C. Mendes-sa, S. Andre, C. Poitou et al., Jejunal T cell inflammation in human obesity correlates with decreased enterocyte insulin signaling, Cell Metabolism, vol.22, issue.1, pp.113-124, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02540998

K. Makki, P. Froguel, and I. Wolowczuk, Adipose tissue in obesity-related inflammation and insulin resistance: cells, cytokines, and chemokines, Inflammation, p.139239, 2013.

P. D. Cani, R. Bibiloni, C. Knauf, A. Waget, A. M. Neyrinck et al., Changes in gut microbiota control metabolic endotoxemiainduced inflammation in high-fat diet-induced obesity and diabetes in mice, Diabetes, vol.57, issue.6, pp.1470-1481, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00410066

P. D. Cani, J. Amar, M. A. Iglesias, M. Poggi, C. Knauf et al., Metabolic endotoxemia initiates obesity and insulin resistance, Diabetes, vol.56, issue.7, pp.1761-1772, 2007.

J. R. Araujo, J. Tomas, C. Brenner, and P. J. Sansonetti, Impact of high-fat diet on the intestinal microbiota and small intestinal physiology before and after the onset of obesity, Biochimie, vol.141, pp.97-106, 2017.

A. M. Johnson, A. Costanzo, M. G. Gareau, A. M. Armando, O. Quehenberger et al., High fat diet causes depletion of intestinal eosinophils associated with intestinal permeability, PloS One, vol.10, issue.4, p.122195, 2015.

M. K. Hamilton, G. Boudry, D. G. Lemay, and H. E. Raybould, Changes in intestinal barrier function and gut microbiota in high-fat diet-fed rats are dynamic and region dependent, American Journal of Physiology -Gastrointestinal and Liver Physiology, vol.308, issue.10, pp.840-851, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01409505

S. Ghezzal, B. G. Postal, E. Quevrain, L. Brot, P. Seksik et al., Palmitic acid damages gut epithelium integrity and initiates inflammatory cytokine production, Biochimica et Biophysica Acta (BBA) -Molecular and Cell Biology of Lipids, vol.1865, issue.2, p.158530, 2020.
URL : https://hal.archives-ouvertes.fr/hal-02449651

L. Genser, D. Aguanno, H. A. Soula, L. Dong, L. Trystram et al., Increased jejunal permeability in human obesity is revealed by a lipid challenge and is linked to inflammation and type 2 diabetes, The Journal of Pathology, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02003437

K. Kawajiri and Y. Fujii-kuriyama, The aryl hydrocarbon receptor: a multifunctional chemical sensor for host defense and homeostatic maintenance, Experimental Animals, vol.66, issue.2, pp.75-89, 2017.

P. Ramadoss, C. Marcus, and G. H. Perdew, Role of the aryl hydrocarbon receptor in drug metabolism, Expert Opinion on Drug Metabolism and Toxicology, vol.1, issue.1, pp.9-21, 2005.

V. Rothhammer and F. J. Quintana, The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease, Nature Reviews Immunology, vol.19, issue.3, pp.184-197, 2019.

B. Lamas, M. L. Richard, V. Leducq, H. P. Pham, M. L. Michel et al., 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

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.

K. Yu, Y. Ma, Z. Zhang, X. Fan, T. Li et al., AhR activation protects intestinal epithelial barrier function through regulation of Par-6, Journal of Molecular Histology, vol.49, issue.5, pp.449-458, 2018.

C. X. Xu, C. Wang, Z. M. Zhang, C. D. Jaeger, S. L. Krager et al., Aryl hydrocarbon receptor deficiency protects mice from dietinduced adiposity and metabolic disorders through increased energy expenditure, International Journal of Obesity, vol.39, issue.8, pp.1300-1309, 2015.

B. J. Moyer, I. Y. Rojas, J. S. Kerley-hamilton, K. V. Nemani, H. W. Trask et al., Obesity and fatty liver are prevented by inhibition of the aryl hydrocarbon receptor in both female and male mice, Nutrition Research, vol.44, pp.38-50, 2017.

J. H. Lee, T. Wada, M. Febbraio, J. He, T. Matsubara et al., A novel role for the dioxin receptor in fatty acid metabolism and hepatic steatosis, Gastroenterology, vol.139, issue.2, pp.653-663, 2010.

J. He, B. Hu, X. Shi, E. R. Weidert, P. Lu et al., Activation of the aryl hydrocarbon receptor sensitizes mice to nonalcoholic steatohepatitis by deactivating mitochondrial sirtuin deacetylase Sirt3, Molecular and Cellular Biology, vol.33, issue.10, pp.2047-2055, 2013.

T. Wada, H. Sunaga, K. Miyata, H. Shirasaki, Y. Uchiyama et al., Aryl hydrocarbon receptor plays protective roles against high fat diet (HFD)-induced hepatic steatosis and the subsequent lipotoxicity via direct transcriptional regulation of Socs3 gene expression, Journal of Biological Chemistry, vol.291, issue.13, pp.7004-7016, 2016.

J. M. Natividad, A. Agus, J. Planchais, B. Lamas, A. C. Jarry et al., Impaired aryl hydrocarbon receptor ligand production by the gut microbiota is a key factor in metabolic syndrome, Cell Metabolism, vol.28, issue.5, pp.737-749, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01895943

J. Aron-wisnewsky, E. Prifti, E. Belda, F. Ichou, B. D. Kayser et al., Major microbiota dysbiosis in severe obesity: fate after bariatric surgery, Gut, vol.68, issue.1, pp.70-82, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02629250

E. Dalmas, C. Rouault, M. Abdennour, C. Rovere, S. Rizkalla et al., Variations in circulating inflammatory factors are related to changes in calorie and carbohydrate intakes early in the course of surgeryinduced weight reduction, American Journal of Clinical Nutrition, vol.94, issue.2, pp.450-458, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00858489

I. Chantret, A. Rodolosse, A. Barbat, E. Dussaulx, E. Brot-laroche et al., Differential expression of sucrase-isomaltase in clones isolated from early and late passages of the cell line Caco-2: evidence for glucose-dependent negative regulation, Journal of Cell Science, vol.107, pp.213-225, 1994.

E. Morel, S. Ghezzal, G. Lucchi, C. Truntzer, J. P. Pais-de-barros et al., Cholesterol trafficking and raft-like membrane domain composition mediate scavenger receptor class B type 1-dependent lipid sensing in intestinal epithelial cells, Biochimica et Biophysica Acta, vol.1863, issue.2, pp.199-211, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01661856

O. Beaslas, C. Cueille, F. Delers, D. Chateau, J. Chambaz et al., Sensing of dietary lipids by enterocytes: a new role for SR-BI/CLA-1, PloS One, vol.4, issue.1, p.4278, 2009.

C. S. Petit, F. Barreau, L. Besnier, P. Gandille, B. Riveau et al., Requirement of cellular prion protein for intestinal barrier function and mislocalization in patients with inflammatory bowel disease, Gastroenterology, vol.143, issue.1, pp.122-132, 2012.

J. Ikenouchi, H. Sasaki, S. Tsukita, M. Furuse, and S. Tsukita, Loss of occludin affects tricellular localization of tricellulin, Molecular Biology of the Cell, vol.19, issue.11, pp.4687-4693, 2008.

J. Ikenouchi, M. Furuse, K. Furuse, H. Sasaki, S. Tsukita et al., Tricellulin constitutes a novel barrier at tricellular contacts of epithelial cells, The Journal of Cell Biology, vol.171, issue.6, pp.939-945, 2005.

M. Saitou, Y. Ando-akatsuka, M. Itoh, M. Furuse, J. Inazawa et al., Mammalian occludin in epithelial cells: its expression and subcellular distribution, European Journal of Cell Biology, vol.73, issue.3, pp.222-231, 1997.

O. B. Parks, D. A. Pociask, Z. Hodzic, J. K. Kolls, and M. Good, Interleukin-22 signaling in the regulation of intestinal health and disease, Frontiers in Cellular Developmental Biology, vol.3, p.85, 2015.

C. F. Vogel and T. Haarmann-stemmann, The aryl hydrocarbon receptor repressor -more than a simple feedback inhibitor of AhR signaling: clues for its role in inflammation and cancer, Current Opinion in Toxicology, vol.2, pp.109-119, 2017.

Y. Duan, L. Zeng, C. Zheng, B. Song, F. Li et al., Inflammatory links between high fat diets and diseases, Frontiers in Immunology, vol.9, p.2649, 2018.

B. Rothen-rutishauser, F. K. Riesen, A. Braun, M. Gunthert, and H. Wunderli-allenspach, Dynamics of tight and adherens junctions under EGTA treatment, Journal of Membrane Biology, vol.188, issue.2, pp.151-162, 2002.

A. W. Stadnyk, Intestinal epithelial cells as a source of inflammatory cytokines and chemokines, Canadian Journal of Gastroenterology, vol.16, issue.4, pp.241-246, 2002.

N. Miron and V. Cristea, Enterocytes: active cells in tolerance to food and microbial antigens in the gut, Clinical and Experimental Immunology, vol.167, issue.3, pp.405-412, 2012.

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, 2018.

J. Stein and G. Kottra, Intestinal intercellular tight junctions. I. Structure and molecular mechanisms of regulation, Zeitschrift für Gastroenterologie, vol.35, issue.3, pp.205-220, 1997.

L. L. Mitic and J. M. Anderson, Molecular architecture of tight junctions, Annual Review of Physiology, vol.60, pp.121-142, 1998.

K. Matter and M. S. Balda, Signalling to and from tight junctions, Nature Reviews Molecular Cell Biology, vol.4, issue.3, pp.225-236, 2003.

L. Marinelli, C. Martin-gallausiaux, J. M. Bourhis, F. Beguet-crespel, H. M. Blottiere et al., Identification of the novel role of butyrate as AhR ligand in human intestinal epithelial cells, Scientific Reports, vol.9, issue.1, p.643, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01996381

Y. Kang and Y. Cai, The development of probiotics therapy to obesity: a therapy that has gained considerable momentum, Hormones, vol.17, issue.2, pp.141-151, 2018.

B. A. Peters, J. A. Shapiro, T. R. Church, G. Miller, C. Trinh-shevrin et al., A taxonomic signature of obesity in a large study of American adults, Scientific Reports, vol.8, issue.1, p.9749, 2018.

M. Million, M. Maraninchi, M. Henry, F. Armougom, H. Richet et al., Obesity-associated gut microbiota is enriched in Lactobacillus reuteri and depleted in Bifidobacterium animalis and Methanobrevibacter smithii, International Journal of Obesity, vol.36, issue.6, pp.817-825, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02649498

F. Armougom, M. Henry, B. Vialettes, D. Raccah, and D. Raoult, Monitoring bacterial community of human gut microbiota reveals an increase in Lactobacillus in obese patients and Methanogens in anorexic patients, PloS One, vol.4, issue.9, p.7125, 2009.

C. Andrews, M. H. Mclean, and S. K. Durum, Cytokine tuning of intestinal epithelial function, Frontiers in Immunology, vol.9, p.1270, 2018.

R. Singh, S. Chandrashekharappa, S. R. Bodduluri, B. V. Baby, B. Hegde et al., Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway, Nature Communications, vol.10, issue.1, p.89, 2019.

E. Guyot, A. Chevallier, R. Barouki, and X. Coumoul, The AhR twist: ligand-dependent AhR signaling and pharmaco-toxicological implications, Drug Discovery Today, vol.18, issue.9e10, pp.479-486, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02190808

L. Larigot, L. Juricek, J. Dairou, and X. Coumoul, AhR signaling pathways and regulatory functions, Biochimie Open, vol.7, pp.1-9, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02196328

M. J. Dorfel and O. Huber, Modulation of tight junction structure and function by kinases and phosphatases targeting occludin, Journal of Biomedicine and Biotechnology, vol.807356, 2012.

D. Schuhmacher, J. M. Sontag, and E. Sontag, Protein phosphatase 2A: more than a passenger in the regulation of epithelial cell-cell junctions, Frontiers in Cellular Developmental Biology, vol.7, p.30, 2019.

J. R. Turner, Intestinal mucosal barrier function in health and disease, Nature Reviews Immunology, vol.9, issue.11, pp.799-809, 2009.

C. M. Van-itallie and J. M. Anderson, Phosphorylation of tight junction transmembrane proteins: many sites, much to do, Tissue Barriers, vol.6, issue.1, p.1382671, 2018.