H. Chung, S. J. Pamp, J. A. Hill, N. K. Surana, S. M. Edelman et al., Gut Immune Maturation Depends on Colonization with a Host-Specific Microbiota, Cell, vol.149, issue.7, pp.1578-1593, 2012.

T. Gensollen, S. S. Iyer, D. L. Kasper, and R. S. Blumberg, How colonization by microbiota in early life shapes the immune system, Science, vol.352, issue.6285, pp.539-544, 2016.

L. M. Cox, S. Yamanishi, J. Sohn, A. V. Alekseyenko, J. M. Leung et al., Altering the Intestinal Microbiota during a Critical Developmental Window Has Lasting Metabolic Consequences, Cell, vol.158, issue.4, pp.705-721, 2014.

N. Geva-zatorsky, E. Sefik, L. Kua, L. Pasman, T. G. Tan et al., Mining the Human Gut Microbiota for Immunomodulatory Organisms, Cell, vol.168, issue.5, pp.928-943.e11, 2017.

M. C. Arrieta, L. T. Stiemsma, P. A. Dimitriu, L. Thorson, S. Russell et al., Early infancy microbial and metabolic alterations affect risk of childhood asthma, Science Translational Medicine, vol.7, issue.307, pp.307ra152-307ra152, 2015.

A. Hviid, H. Svanstrom, and M. Frisch, Antibiotic use and inflammatory bowel diseases in childhood, Gut, vol.60, issue.1, pp.49-54, 2010.

J. L. Round and S. K. Mazmanian, The gut microbiota shapes intestinal immune responses during health and disease, Nature Reviews Immunology, vol.9, issue.5, pp.313-323, 2009.

N. Adriaenssens, S. Coenen, A. Versporten, A. Muller, V. Vankerckhoven et al., European Surveillance of Antimicrobial Consumption (ESAC): quality appraisal of antibiotic use in Europe, Journal of Antimicrobial Chemotherapy, vol.66, issue.suppl_6, pp.vi71-vi77, 2011.

L. E. Vaz, K. P. Kleinman, M. A. Raebel, J. D. Nordin, M. D. Lakoma et al., Recent Trends in Outpatient Antibiotic Use in Children, PEDIATRICS, vol.133, issue.3, pp.375-385, 2014.

L. A. Hicks, T. H. Taylor, and R. J. Hunkler, U.S. Outpatient Antibiotic Prescribing, 2010, New England Journal of Medicine, vol.368, issue.15, pp.1461-1462, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01862020

G. Chai, L. Governale, A. W. Mcmahon, J. P. Trinidad, J. Staffa et al., Trends of Outpatient Prescription Drug Utilization in US Children, 2002-2010, PEDIATRICS, vol.130, issue.1, pp.23-31, 2012.

A. L. Hersh, M. A. Jackson, and L. A. Hicks, Principles of Judicious Antibiotic Prescribing for Upper Respiratory Tract Infections in Pediatrics, PEDIATRICS, vol.132, issue.6, pp.1146-1154, 2013.

A. F. Schulfer, T. Battaglia, Y. Alvarez, L. Bijnens, V. E. Ruiz et al., Intergenerational transfer of antibiotic-perturbed microbiota enhances colitis in susceptible mice, Nature Microbiology, vol.3, issue.2, pp.234-242, 2017.

C. Jernberg, S. Löfmark, C. Edlund, and J. K. Jansson, Long-term ecological impacts of antibiotic administration on the human intestinal microbiota, The ISME Journal, vol.1, issue.1, pp.56-66, 2007.

S. Y. Shaw, J. F. Blanchard, and C. N. Bernstein, Association Between the Use of Antibiotics and New Diagnoses of Crohn?s Disease and Ulcerative Colitis, American Journal of Gastroenterology, vol.106, issue.12, pp.2133-2142, 2011.

V. E. Ruiz, T. Battaglia, Z. D. Kurtz, L. Bijnens, A. Ou et al., A single early-in-life macrolide course has lasting effects on murine microbial network topology and immunity, Nature Communications, vol.8, issue.1, p.518, 2017.

B. Chassaing, J. D. Aitken, M. Malleshappa, and M. Vijay?kumar, Dextran Sulfate Sodium (DSS)?Induced Colitis in Mice, Current Protocols in Immunology, vol.104, issue.1, 2014.

Y. R. Nobel, L. M. Cox, F. F. Kirigin, N. A. Bokulich, S. Yamanishi et al., Metabolic and metagenomic outcomes from early-life pulsed antibiotic treatment, Nature Communications, vol.6, issue.1, p.7486, 2015.

B. Chassaing, G. Srinivasan, M. A. Delgado, A. N. Young, A. T. Gewirtz et al., Fecal Lipocalin 2, a Sensitive and Broadly Dynamic Non-Invasive Biomarker for Intestinal Inflammation, PLoS ONE, vol.7, issue.9, p.e44328, 2012.

A. B. Rogers and J. Houghton, Helicobacter -Based Mouse Models of Digestive System Carcinogenesis, Methods in Molecular Biology, vol.511, pp.267-295, 2009.

S. J. Shin, J. H. Lim, S. Chung, D. Y. Youn, H. W. Chung et al., Peroxisome proliferator-activated receptor-? activator fenofibrate prevents high-fat diet-induced renal lipotoxicity in spontaneously hypertensive rats, Hypertension Research, vol.32, issue.10, pp.835-845, 2009.

C. Tang, T. Kamiya, Y. Liu, M. Kadoki, S. Kakuta et al., Inhibition of Dectin-1 Signaling Ameliorates Colitis by Inducing Lactobacillus-Mediated Regulatory T Cell Expansion in the Intestine, Cell Host & Microbe, vol.18, issue.2, pp.183-197, 2015.

C. Gimblet, M. A. Loesche, L. Carvalho, E. M. Carvalho, E. A. Grice et al., IL-22 Protects against Tissue Damage during Cutaneous Leishmaniasis, PLOS ONE, vol.10, issue.8, p.e0134698, 2015.

K. H. Restori, K. L. Mcdaniel, A. E. Wray, M. T. Cantorna, and A. C. Ross, Streptococcus pneumoniae?Induced Pneumonia and Citrobacter rodentium?Induced Gut Infection Differentially Alter Vitamin A Concentrations in the Lung and Liver of Mice, The Journal of Nutrition, vol.144, issue.3, pp.392-398, 2014.

J. Liu, S. N. Cho, B. Akkanti, N. Jin, J. N. Mao et al., ErbB2 Pathway Activation upon Smad4 Loss Promotes Lung Tumor Growth and Metastasis, Cell Reports, vol.10, issue.9, pp.1599-1613, 2015.

K. J. Livak and T. D. Schmittgen, Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2???CT Method, Methods, vol.25, issue.4, pp.402-408, 2001.

J. G. Caporaso, C. L. Lauber, W. A. Walters, D. Berg-lyons, J. Huntley et al., Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms, The ISME Journal, vol.6, issue.8, pp.1621-1624, 2012.

M. J. Anderson, A new method for non-parametric multivariate analysis of variance, Austral Ecology, vol.26, issue.1, pp.32-46, 2001.

N. Segata, J. Izard, L. Waldron, D. Gevers, L. Miropolsky et al., Metagenomic biomarker discovery and explanation, Genome Biology, vol.12, issue.6, p.R60, 2011.

J. De-winter, Small sample sizes, overextraction, and unrealistic expectations: A commentary on M. Mattsson, Accident Analysis & Prevention, vol.50, pp.776-777, 2013.

M. P. Kronman, T. E. Zaoutis, K. Haynes, R. Feng, and S. E. Coffin, Antibiotic Exposure and IBD Development Among Children: A Population-Based Cohort Study, PEDIATRICS, vol.130, issue.4, pp.e794-e803, 2012.

X. S. Zhang, J. Li, K. A. Krautkramer, M. Badri, T. Battaglia et al., Decision letter: Antibiotic-induced acceleration of type 1 diabetes alters maturation of innate intestinal immunity, vol.7, p.37816, 2018.

C. Roubaud-baudron, V. E. Ruiz, A. M. Swan, B. A. Vallance, C. Ozkul et al., Long-Term Effects of Early-Life Antibiotic Exposure on Resistance to Subsequent Bacterial Infection, mBio, vol.10, issue.6, pp.2820-2839, 2019.
URL : https://hal.archives-ouvertes.fr/inserm-03010065

M. Mähler, I. J. Bristol, E. H. Leiter, A. E. Workman, E. H. Birkenmeier et al., Differential susceptibility of inbred mouse strains to dextran sulfate sodium-induced colitis, American Journal of Physiology-Gastrointestinal and Liver Physiology, vol.274, issue.3, pp.G544-G551, 1998.

D. A. Hill, C. Hoffmann, M. C. Abt, Y. Du, D. Kobuley et al., Metagenomic analyses reveal antibiotic-induced temporal and spatial changes in intestinal microbiota with associated alterations in immune cell homeostasis, Mucosal Immunology, vol.3, issue.2, pp.148-158, 2009.

J. H. Yang, P. Bhargava, D. Mccloskey, N. Mao, B. O. Palsson et al., Antibiotic-Induced Changes to the Host Metabolic Environment Inhibit Drug Efficacy and Alter Immune Function, Cell Host & Microbe, vol.22, issue.6, pp.757-765.e3, 2017.

P. Dharmani, P. Leung, and K. Chadee, Tumor Necrosis Factor-? and Muc2 Mucin Play Major Roles in Disease Onset and Progression in Dextran Sodium Sulphate-Induced Colitis, PLoS ONE, vol.6, issue.9, p.e25058, 2011.

K. Sugimoto, A. Ogawa, E. Mizoguchi, Y. Shimomura, A. Andoh et al., IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis, Journal of Clinical Investigation, vol.118, issue.2, pp.534-578, 2008.

M. F. Neurath, Cytokines in inflammatory bowel disease, Nature Reviews Immunology, vol.14, issue.5, pp.329-342, 2014.

L. A. Zenewicz, G. D. Yancopoulos, D. M. Valenzuela, A. J. Murphy, S. Stevens et al., Innate and Adaptive Interleukin-22 Protects Mice from Inflammatory Bowel Disease, Immunity, vol.29, issue.6, pp.947-957, 2008.

C. Hoebler, E. Gaudier, P. De-coppet, M. Rival, and C. Cherbut, MUC Genes Are Differently Expressed During Onset and Maintenance of Inflammation in Dextran Sodium Sulfate-Treated Mice, Digestive Diseases and Sciences, vol.51, issue.2, pp.381-389, 2006.

C. C. Booijink, S. El-aidy, M. Rajili?-stojanovi?, H. G. Heilig, F. J. Troost et al., High temporal and inter-individual variation detected in the human ileal microbiota, Environmental Microbiology, vol.12, issue.12, pp.3213-3227, 2010.

H. Hayashi, R. Takahashi, T. Nishi, M. Sakamoto, and Y. Benno, Molecular analysis of jejunal, ileal, caecal and recto-sigmoidal human colonic microbiota using 16S rRNA gene libraries and terminal restriction fragment length polymorphism, Journal of Medical Microbiology, vol.54, issue.11, pp.1093-1101, 2005.

K. Korpela, A. Salonen, L. J. Virta, R. A. Kekkonen, K. Forslund et al., Intestinal microbiome is related to lifetime antibiotic use in Finnish pre-school children, Nature Communications, vol.7, issue.1, p.10410, 2016.

S. Melgar, A. Karlsson, and E. Michaëlsson, Acute colitis induced by dextran sulfate sodium progresses to chronicity in C57BL/6 but not in BALB/c mice: correlation between symptoms and inflammation, American Journal of Physiology-Gastrointestinal and Liver Physiology, vol.288, issue.6, pp.G1328-G1338, 2005.

L. Virta, A. Auvinen, H. Helenius, P. Huovinen, and K. L. Kolho, Association of Repeated Exposure to Antibiotics With the Development of Pediatric Crohn's Disease--A Nationwide, Register-based Finnish Case-Control Study, American Journal of Epidemiology, vol.175, issue.8, pp.775-784, 2012.

A. K. Örtqvist, C. Lundholm, J. Halfvarson, J. F. Ludvigsson, and C. Almqvist, Fetal and early life antibiotics exposure and very early onset inflammatory bowel disease: a population-based study, Gut, vol.68, issue.2, pp.218-225, 2018.

I. Brook, H. M. Wexler, and E. J. Goldstein, Antianaerobic Antimicrobials: Spectrum and Susceptibility Testing, Clinical Microbiology Reviews, vol.26, issue.3, pp.526-546, 2013.

R. Leclercq and P. Courvalin, Intrinsic and unusual resistance to macrolide, lincosamide, and streptogramin antibiotics in bacteria., Antimicrobial Agents and Chemotherapy, vol.35, issue.7, pp.1273-1276, 1991.

S. Tanaka, T. Kobayashi, P. Songjinda, A. Tateyama, M. Tsubouchi et al., Influence of antibiotic exposure in the early postnatal period on the development of intestinal microbiota, FEMS Immunology & Medical Microbiology, vol.56, issue.1, pp.80-87, 2009.

C. Hagiwara, M. Tanaka, and H. Kudo, Increase in colorectal epithelial apoptotic cells in patients with ulcerative colitis ultimately requiring surgery, Journal of Gastroenterology and Hepatology, vol.17, issue.7, pp.758-764, 2002.

S. Zeissig, C. Bojarski, N. Buergel, J. Mankertz, M. Zeitz et al., Downregulation of epithelial apoptosis and barrier repair in active Crohn's disease by tumour necrosis factor antibody treatment, Gut, vol.53, issue.9, pp.1295-1302, 2004.

R. M. Jones, C. Desai, T. M. Darby, L. Luo, A. A. Wolfarth et al., Lactobacilli Modulate Epithelial Cytoprotection through the Nrf2 Pathway, Cell Reports, vol.12, issue.8, pp.1217-1225, 2015.

M. M. Prata, A. Havt, D. T. Bolick, R. Pinkerton, A. Lima et al., Comparisons between myeloperoxidase, lactoferrin, calprotectin and lipocalin-2, as fecal biomarkers of intestinal inflammation in malnourished children, J Transl Sci, vol.2, issue.2, pp.134-143, 2016.

M. Vieira, S. Hiltensperger, M. Kumar, V. Zegarra-ruiz, D. Dehner et al., Translocation of a gut pathobiont drives autoimmunity in mice and humans, Science, vol.359, issue.6380, pp.1156-61, 2018.

N. W. Palm, M. R. De zoete, T. W. Cullen, N. A. Barry, J. Stefanowski et al., Immunoglobulin A Coating Identifies Colitogenic Bacteria in Inflammatory Bowel Disease, Cell, vol.158, issue.5, pp.1000-1010, 2014.

I. I. Ivanov, K. Atarashi, N. Manel, E. L. Brodie, T. Shima et al., Induction of Intestinal Th17 Cells by Segmented Filamentous Bacteria, Cell, vol.139, issue.3, pp.485-498, 2009.

J. B. Grigg and G. F. Sonnenberg, Host-Microbiota Interactions Shape Local and Systemic Inflammatory Diseases, The Journal of Immunology, vol.198, issue.2, pp.564-571, 2017.

N. Kamada, S. U. Seo, G. Y. Chen, and G. Núñez, Role of the gut microbiota in immunity and inflammatory disease, Nature Reviews Immunology, vol.13, issue.5, pp.321-335, 2013.

C. P. Harkins, H. H. Kong, and J. A. Segre, Manipulating the Human Microbiome to Manage Disease, JAMA, vol.323, issue.4, p.303, 2020.

B. Sadlack, H. Merz, H. Schorle, A. Schimpl, A. C. Feller et al., Ulcerative colitis-like disease in mice with a disrupted interleukin-2 gene, Cell, vol.75, issue.2, pp.253-261, 1993.

R. K. Sellon, S. Tonkonogy, M. Schultz, L. A. Dieleman, W. Grenther et al., Resident Enteric Bacteria Are Necessary for Development of Spontaneous Colitis and Immune System Activation in Interleukin-10-Deficient Mice, Infection and Immunity, vol.66, issue.11, pp.5224-5231, 1998.

L. A. Zenewicz, A. Antov, and R. A. Flavell, CD4 T-cell differentiation and inflammatory bowel disease, Trends in Molecular Medicine, vol.15, issue.5, pp.199-207, 2009.

E. K. Boden and S. B. Snapper, Regulatory T cells in inflammatory bowel disease, Current Opinion in Gastroenterology, vol.24, issue.6, pp.733-741, 2008.

S. Jin, D. Zhao, C. Cai, D. Song, J. Shen et al., Low-dose penicillin exposure in early life decreases Th17 and the susceptibility to DSS colitis in mice through gut microbiota modification, Scientific Reports, vol.7, issue.1, p.43662, 2017.

Z. Al-nabhani, S. Dulauroy, R. Marques, C. Cousu, S. Al-bounny et al., A Weaning Reaction to Microbiota Is Required for Resistance to Immunopathologies in the Adult, Immunity, vol.50, issue.5, pp.1276-1288.e5, 2019.

K. Atarashi, T. Tanoue, T. Shima, A. Imaoka, T. Kuwahara et al., Induction of Colonic Regulatory T Cells by Indigenous Clostridium Species, Science, vol.331, issue.6015, pp.337-341, 2010.

M. B. Geuking, J. Cahenzli, M. A. Lawson, D. C. Ng, E. Slack et al., Intestinal Bacterial Colonization Induces Mutualistic Regulatory T Cell Responses, Immunity, vol.34, issue.5, pp.794-806, 2011.

A. Bergström, M. B. Kristensen, M. I. Bahl, S. B. Metzdorff, L. N. Fink et al., Nature of bacterial colonization influences transcription of mucin genes in mice during the first week of life, BMC Research Notes, vol.5, issue.1, p.402, 2012.

S. Cornick, A. Tawiah, and K. Chadee, Roles and regulation of the mucus barrier in the gut, Tissue Barriers, vol.3, issue.1-2, p.e982426, 2015.

G. Lennon, Á. Balfe, H. Earley, L. A. Devane, A. Lavelle et al., Influences of the colonic microbiome on the mucous gel layer in ulcerative colitis, Gut Microbes, vol.5, issue.3, pp.277-476, 2014.

M. Wlodarska, B. Willing, K. M. Keeney, A. Menendez, K. S. Bergstrom et al., Antibiotic Treatment Alters the Colonic Mucus Layer and Predisposes the Host to ExacerbatedCitrobacter rodentium-Induced Colitis, Infection and Immunity, vol.79, issue.4, pp.1536-1545, 2011.

J. M. Wells, R. J. Brummer, M. Derrien, T. T. Macdonald, F. Troost et al., Homeostasis of the gut barrier and potential biomarkers, American Journal of Physiology-Gastrointestinal and Liver Physiology, vol.312, issue.3, pp.G171-G193, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01602268

N. L. Ward, C. D. Phillips, D. D. Nguyen, N. K. Shanmugam, Y. Song et al., Antibiotic Treatment Induces Long-lasting Changes in the Fecal Microbiota that Protect Against Colitis, Inflammatory Bowel Diseases, vol.22, issue.10, pp.2328-2340, 2016.

P. M. Munyaka, N. Eissa, C. N. Bernstein, E. Khafipour, and J. E. Ghia, Antepartum Antibiotic Treatment Increases Offspring Susceptibility to Experimental Colitis: A Role of the Gut Microbiota, PLOS ONE, vol.10, issue.11, p.e0142536, 2015.

A. Gonzalez, J. A. Navas-molina, T. Kosciolek, D. Mcdonald, Y. Vázquez-baeza et al., Qiita: rapid, web-enabled microbiome meta-analysis, Nature Methods, vol.15, issue.10, pp.796-798, 2018.

, Jurisdictional immunity of foreign States with regard to claims relating to infringements of obligations under peremptory norms