S. T. Shulman, H. C. Friedmann, and R. H. Sims, Theodor Escherich: The First Pediatric Infectious Diseases Physician? Clinical Infectious Diseases, vol.45, pp.1025-1034, 2007.

K. Chung, B. M. Robert, and . Hungate, Pioneer of Anaerobic Microbial Ecology, Anaerobe. août, vol.3, issue.4, pp.213-220, 1997.

R. E. Hungate, Chapter IV A Roll Tube Method for Cultivation of Strict Anaerobes, Methods in Microbiology, pp.117-149, 1969.

A. Suau, G. R. Gibson, M. D. Collins, and J. Dore, Direct Analysis of Genes Encoding 16S rRNA from Complex Communities Reveals Many Novel Molecular Species within the Human Gut, APPL ENVIRON MICROBIOL, vol.65, p.9, 1999.

P. Marteau and J. Doré, Le microbiote intestinal, un organe à part entière, vol.338, 2017.

P. D. Schloss and J. Handelsman, Introducing DOTUR, a Computer Program for Defining Operational Taxonomic Units and Estimating Species Richness, Appl Environ Microbiol. mars, vol.71, issue.3, pp.1501-1507, 2005.

S. R. Gill, M. Pop, R. T. Deboy, P. B. Eckburg, P. J. Turnbaugh et al., Metagenomic Analysis of the Human Distal Gut Microbiome, Science. 2 juin, vol.312, issue.5778, pp.1355-1364, 2006.

P. J. Turnbaugh, M. Hamady, T. Yatsunenko, B. L. Cantarel, A. Duncan et al., A core gut microbiome in obese and lean twins, Nature. 22 janv, vol.457, issue.7228, pp.480-484, 2009.

M. Consortium, J. Qin, R. Li, J. Raes, M. Arumugam et al., A human gut microbial gene catalogue established by metagenomic sequencing, Nature. mars, vol.464, issue.7285, pp.59-65, 2010.

M. Consortium, J. Li, H. Jia, X. Cai, H. Zhong et al., An integrated catalog of reference genes in the human gut microbiome, Nature Biotechnology. août, vol.32, issue.8, pp.834-875, 2014.

P. Y. Lee, S. Chin, H. Neoh, and R. Jamal, Metaproteomic analysis of human gut microbiota: where are we heading, Journal of Biomedical Science [Internet]. déc, vol.24, issue.1, 2017.

D. Sur,

C. Juste, D. P. Kreil, C. Beauvallet, A. Guillot, S. Vaca et al., Bacterial protein signals are associated with Crohn's disease. Gut, vol.63, pp.1566-77, 2014.

J. Lagier, F. Armougom, M. Million, P. Hugon, I. Pagnier et al., Microbial culturomics: paradigm shift in the human gut microbiome study, Clinical Microbiology and Infection. déc, vol.18, issue.12, pp.1185-93, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01478655

P. J. Turnbaugh and J. I. Gordon, An Invitation to the Marriage of Metagenomics and Metabolomics, Cell. sept, vol.134, issue.5, pp.708-721, 2008.

S. Coudeyras and C. Forestier, Microbiote et probiotiques : impact en santé humaine, Canadian Journal of Microbiology. août, vol.56, issue.8, pp.611-50, 2010.

J. Tap, S. Mondot, F. Levenez, E. Pelletier, C. Caron et al., 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

J. Cheng, A. M. Palva, W. M. De-vos, and R. Satokari, Contribution of the Intestinal Microbiota to Human Health: From Birth to 100 Years of Age, Dobrindt U, Hacker JH, Svanborg C, éditeurs. Between Pathogenicity and Commensalism

H. Berlin, Disponible sur, Current Topics in Microbiology and Immunology), pp.323-369, 2013.

A. Sghir, G. Gramet, A. Suau, V. Rochet, P. Pochart et al., Quantification of Bacterial Groups within Human Fecal Flora by Oligonucleotide Probe Hybridization, Appl Environ Microbiol. mai, vol.66, issue.5, pp.2263-2269, 2000.
URL : https://hal.archives-ouvertes.fr/hal-02698941

L. Rigottier-gois, A. Bourhis, G. Gramet, V. Rochet, and J. Doré, Fluorescent hybridisation combined with flow cytometry and hybridisation of total RNA to analyse the composition of microbial communities in human faeces using 16S rRNA probes, FEMS Microbiology Ecology. mars, vol.43, issue.2, pp.237-282, 2003.
URL : https://hal.archives-ouvertes.fr/hal-02675094

C. Lay, L. Rigottier-gois, K. Holmstrøm, M. Rajilic, E. E. Vaughan et al., Colonic Microbiota Signatures across Five Northern European Countries, Appl Environ Microbiol. juill, vol.71, issue.7, pp.4153-4158, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02682963

B. Dridi, M. Henry, A. El-khéchine, D. Raoult, and M. Drancourt, High Prevalence of Methanobrevibacter smithii and Methanosphaera stadtmanae Detected in the Human Gut Using an Improved DNA Detection Protocol, PLoS One, vol.4, issue.9, 2009.

D. Sur,

C. Hoffmann, S. Dollive, S. Grunberg, J. Chen, H. Li et al., Archaea and Fungi of the Human Gut Microbiome: Correlations with Diet and Bacterial Residents, Disponible sur, vol.8, 2013.

M. Breitbart, I. Hewson, B. Felts, J. M. Mahaffy, J. Nulton et al., Metagenomic Analyses of an Uncultured Viral Community from Human Feces, J Bacteriol, vol.185, issue.20, pp.6220-6223, 2003.

J. A. Stewart, Investigations into the influence of host genetics on the predominant eubacteria in the faecal microflora of children, Journal of Medical Microbiology. 1 déc, vol.54, issue.12, pp.1239-1281, 2005.

I. Adlerberth, B. Carlsson, P. D. Man, F. Jalil, S. R. Khan et al., Intestinal Colonization with Enterobacteriaceae in Pakistani and Swedish Hospital-Delivered Infants, Acta Paediatrica, vol.80, issue.6-7, pp.602-612, 1991.

F. Campeotto, A. Waligora-dupriet, F. Doucet-populaire, N. Kalach, C. Dupont et al., Mise en place de la flore intestinale du nouveau-né, Gastroentérologie Clinique et Biologique. mai, vol.31, issue.5, pp.533-575, 2007.

G. Biasucci, M. Rubini, S. Riboni, L. Morelli, E. Bessi et al., Mode of delivery affects the bacterial community in the newborn gut, Early Human Development. 1 juill, vol.86, issue.1, pp.13-18, 2010.

M. G. Dominguez-bello, E. K. Costello, M. Contreras, M. Magris, G. Hidalgo et al., Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns, Proc Natl Acad Sci U S A. 29 juin, vol.107, issue.26, pp.11971-11976, 2010.

M. M. Grönlund, O. P. Lehtonen, E. Eerola, and P. Kero, Fecal microflora in healthy infants born by different methods of delivery: permanent changes in intestinal flora after cesarean delivery, J Pediatr Gastroenterol Nutr. janv, vol.28, issue.1, pp.19-25, 1999.

M. B. Azad, T. Konya, H. Maughan, D. S. Guttman, C. J. Field et al., Gut microbiota of healthy Canadian infants: profiles by mode of delivery and infant diet at 4 months, CMAJ. 19 mars, vol.185, issue.5, pp.385-94, 2013.

L. Bode and E. Jantscher-krenn, Structure-Function Relationships of Human Milk Oligosaccharides123, Adv Nutr. 4 mai, vol.3, issue.3, pp.383-391, 2012.

J. Langhendries, T. Paquay, M. Hannon, and J. Darimont, Acquisition de la flore intestinale néonatale: rôle sur la morbidité et perspectives thérapeutiques, Archives de Pédiatrie. juin, vol.5, issue.6, pp.644-53, 1998.

P. S. Pannaraj, F. Li, C. Cerini, J. M. Bender, S. Yang et al., Association Between Breast Milk Bacterial Communities and Establishment and Development of the Infant Gut Microbiome, JAMA Pediatr. 1 juill, vol.171, issue.7, pp.647-54, 2017.

P. L. Stark and A. Lee, The microbial ecology of the large bowel of breast-fed and formula-fed infants during the first year of life, J. Med Microbiol, p.p, 1982.

C. Haro, M. Montes-borrego, O. A. Rangel-zúñiga, J. F. Alcalá-díaz, F. Gómez-delgado et al., Two Healthy Diets Modulate Gut Microbial Community Improving Insulin Sensitivity in a Human Obese Population, The Journal of Clinical Endocrinology & Metabolism. janv, vol.101, issue.1, pp.233-275, 2016.

C. Haro, S. Garcia-carpintero, J. F. Alcala-diaz, F. Gomez-delgado, J. Delgado-lista et al., The gut microbial community in metabolic syndrome patients is modified by diet, The Journal of Nutritional Biochemistry. janv, vol.27, pp.27-31, 2016.

L. Metahit, E. Chatelier, T. Nielsen, J. Qin, E. Prifti et al., Richness of human gut microbiome correlates with metabolic markers, Nature. août, vol.500, issue.7464, pp.541-547, 2013.

. Anr-microobes-consortium, . Microobes-consortium-members, A. Cotillard, S. P. Kennedy, L. C. Kong et al., Dietary intervention impact on gut microbial gene richness, Nature. août, vol.500, issue.7464, pp.585-593, 2013.

G. D. Wu, J. Chen, C. Hoffmann, K. Bittinger, Y. Chen et al., Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes, Science, vol.334, issue.6052, pp.105-113, 2011.

A. Tomova, I. Bukovsky, E. Rembert, W. Yonas, J. Alwarith et al., The Effects of Vegetarian and Vegan Diets on Gut Microbiota

H. Hayashi, M. Sakamoto, and Y. Benno, Fecal Microbial Diversity in a Strict Vegetarian as Determined by Molecular Analysis and Cultivation, Microbiology and Immunology. déc, vol.46, issue.12, pp.819-850, 2002.

D. Palma, G. Nadal, I. Collado, M. C. Sanz, and Y. , Effects of a gluten-free diet on gut microbiota and immune function in healthy adult human subjects, British Journal of Nutrition, vol.102, issue.08, p.1154, 2009.

R. E. Ley, F. Bäckhed, P. Turnbaugh, C. A. Lozupone, R. D. Knight et al., Obesity alters gut microbial ecology, Proc Natl Acad Sci, vol.102, issue.31, pp.11070-11075, 2005.

S. A. Bingham, N. E. Day, R. Luben, P. Ferrari, N. Slimani et al., Dietary fibre in food and protection against colorectal cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC): an observational study, The Lancet. mai, vol.361, issue.9368, pp.1496-501, 2003.
URL : https://hal.archives-ouvertes.fr/inserm-00110132

L. Dethlefsen and D. A. Relman, Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation, Proc Natl Acad Sci U S A. 15 mars, vol.108, issue.1, pp.4554-61, 2011.

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. mai, vol.1, issue.1, pp.56-66, 2007.

V. B. Young and T. M. Schmidt, Antibiotic-Associated Diarrhea Accompanied by Large-Scale Alterations in the Composition of the Fecal Microbiota, J Clin Microbiol. mars, vol.42, issue.3, pp.1203-1209, 2004.

F. Sommer, J. M. Anderson, R. Bharti, J. Raes, and P. Rosenstiel, The resilience of the intestinal microbiota influences health and disease, Nature Reviews Microbiology. 19 juin, vol.15, issue.10, pp.630-638, 2017.

C. A. Thaiss, D. Zeevi, M. Levy, G. Zilberman-schapira, J. Suez et al., Transkingdom Control of Microbiota Diurnal Oscillations Promotes Metabolic Homeostasis, Cell, vol.159, issue.3, pp.514-543, 2014.

H. Gordon, E. Bruckner-kardoss, S. Wagner, and B. Wostmann, Characteristics of the germfree rat, Acta Anatomica, pp.367-89, 1966.

J. Tomas, J. Reygner, C. Mayeur, R. Ducroc, S. Bouet et al., Early colonizing Esche richia coli elicits remodeling of rat colonic epithelium shifting toward a new homeostatic state, ISME J. janv, vol.9, issue.1, pp.46-58, 2015.

A. Marcobal, A. M. Southwick, K. A. Earle, and J. L. Sonnenburg, A refined palate: Bacterial consumption of host glycans in the gut, Glycobiology. 1 sept, vol.23, issue.9, pp.1038-1084, 2013.

C. K. Heazlewood, M. C. Cook, R. Eri, G. R. Price, S. B. Tauro et al., Aberrant Mucin Assembly in Mice Causes Endoplasmic Reticulum Stress and Spontaneous Inflammation Resembling Ulcerative Colitis, PLoS Medicine, vol.5, issue.3, p.21, 2008.

C. V. Srikanth and B. A. Mccormick, Interactions of the Intestinal Epithelium with the Pathogen and the Indigenous Microbiota: A Three-Way Crosstalk, Interdiscip Perspect Infect Dis, 2008.

D. Gerding, T. Meyer, and C. Lee, Administration of spores of nontoxigenic of Clostridium difficile strain M3 for prevention of recurrent C. difficile infection, JAMA, 2015.

J. Zheng, M. G. Gänzle, X. B. Lin, L. Ruan, and M. Sun, Diversity and dynamics of bacteriocins from human microbiome: Bacteriocins of human microbiome, Environ Microbiol. juin, vol.17, issue.6, pp.2133-2176, 2015.

C. G. Buffie, V. Bucci, R. R. Stein, P. T. Mckenney, L. Ling et al., Precision microbiome restoration of bile acid-mediated resistance to Clostridium difficile, Nature. 8 janv, vol.517, issue.7533, pp.205-213, 2015.

D. Ulluwishewa, R. C. Anderson, W. C. Mcnabb, P. J. Moughan, J. M. Wells et al., Regulation of Tight Junction Permeability by Intestinal Bacteria and Dietary Components, The Journal of Nutrition. 1 mai, vol.141, issue.5, pp.769-76, 2011.

A. Bernalier-donadille, Fermentative metabolism by the human gut microbiota, Gastroentérologie Clinique et Biologique. sept, vol.34, pp.16-22, 2010.

L. Willemsen, M. A. Koetsier, S. Van-deventer, and E. Van-tol, Short chain fatty acids stimulate epithelial mucin 2 expression through differential effects on prostaglandin E1 and E2 production by intestinal myofibroblasts. Gut, vol.52, pp.1442-1449, 2003.

M. H. Kim, S. G. Kang, J. H. Park, M. Yanagisawa, and C. H. Kim, Short-Chain Fatty Acids Activate GPR41 and GPR43 on Intestinal Epithelial Cells to Promote Inflammatory Responses in Mice, Gastroenterology. août, vol.145, issue.2, pp.396-406, 2013.

M. Vinolo, H. G. Rodrigues, E. Hatanaka, F. T. Sato, S. C. Sampaio et al., Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils, The Journal of Nutritional Biochemistry. sept, vol.22, issue.9, pp.849-55, 2011.

G. T. Macfarlane and J. H. Cummings, The colonic flora, fermentation and large bowel digestive function, The large intestine: physiology, pathophysiology and disease, 1991.

C. Robert and A. Bernalier-donadille, The cellulolytic microflora of the human colon: evidence of microcrystalline cellulose-degrading bacteria in methane-excreting subjects, FEMS Microbiology Ecology, vol.46, issue.1, pp.81-90, 2003.
URL : https://hal.archives-ouvertes.fr/hal-02681974

C. Chassard, V. Goumy, M. Leclerc, C. Del'homme, and A. Bernalier-donadille, Characterization of the xylan-degrading microbial community from human faeces: Xylanolytic microbiota from human faeces, FEMS Microbiology Ecology. juill, vol.61, issue.1, pp.121-152, 2007.

H. Flint, K. Scott, P. Louis, and S. Duncan, The role of the gut microbiota in nutrition and health, Nature Reviews Gastroenterolgy & Hepatology, vol.9, 2012.

Y. B. David, B. Dassa, I. Borovok, R. Lamed, N. M. Koropatkin et al., Ruminococcal cellulosome systems from rumen to human, Environmental Microbiology, vol.17, issue.9, pp.3407-3433, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02641296

K. Jaeger, S. Ransac, B. W. Dijkstra, C. Colson, M. Heuvel et al., Bacterial lipases, FEMS Microbiology Reviews. sept, vol.15, issue.1, pp.29-63, 1994.

S. Baron and P. Hylemon, Biotransformation of bile acis, cholesterol and steroid hormones, Gastrointestinal microbiology, gastrointestinal ecosystems and fermentations, 1997.

O. Gillor, A. Etzion, and M. A. Riley, The dual role of bacteriocins as anti-and probiotics, Appl Microbiol Biotechnol. déc, vol.81, issue.4, pp.591-606, 2008.

V. Liévin-le-moal and A. L. Servin, The Front Line of Enteric Host Defense against Unwelcome Intrusion of Harmful Microorganisms: Mucins, Antimicrobial Peptides, and Microbiota, Clin Microbiol Rev. avr, vol.19, issue.2, pp.315-352, 2006.

F. Sommer and F. Bäckhed, The gut microbiota -masters of host development and physiology, Nat Rev Microbiol. avr, vol.11, issue.4, pp.227-265, 2013.

A. Macpherson and N. Harris, Interactions between commensal intestinal bacteria and the immune system, Nature Reviews Immunology, vol.4, pp.478-85, 2004.

S. Y. Salim and J. D. Söderholm, Importance of disrupted intestinal barrier in inflammatory bowel diseases: Inflammatory Bowel Diseases, janv, vol.17, issue.1, pp.362-81, 2011.

H. Tremel, B. Kienle, L. Weilemann, P. Stehle, and P. Furst, Glutamine Dipeptide-Supplemented Parenteral Nutrition Maintains Intestinal Function in the Critically III, Gastroenterology, 1994.

. Hulst-rrwj-van-der, . Meyenfeldt-mf-von, N. Deutz, P. B. Soeters, and R. Brummer, Glutamine and the preservation of gut integrity. The Lancet, vol.29, pp.1363-1368, 1993.

N. Li, P. Lewis, D. Samuelson, K. Liboni, and J. Neu, Glutamine regulates Caco-2 cell tight junction proteins, American Journal of Physiology-Gastrointestinal and Liver Physiology. sept, vol.287, issue.3, pp.726-759, 2004.

A. Finamore, M. Massimi, C. Devirgiliis, L. Mengheri, and E. , Zinc Deficiency Induces Membrane Barrier Damage and Increases Neutrophil Transmigration in Caco-2 Cells, J Nutr. 1 sept, vol.138, issue.9, pp.1664-70, 2008.

J. L. Watson, S. Ansari, H. Cameron, A. Wang, M. Akhtar et al., Green tea polyphenol (?)-epigallocatechin gallate blocks epithelial barrier dysfunction provoked by IFN-? but not by IL-4, American Journal of Physiology-Gastrointestinal and Liver Physiology, vol.287, issue.5, pp.954-61, 2004.

T. Suzuki and H. Hara, Quercetin Enhances Intestinal Barrier Function through the Assembly of Zonnula Occludens-2, Occludin, and Claudin-1 and the Expression of Claudin-4 in Caco-2 Cells, The Journal of Nutrition. 1 mai, vol.139, issue.5, pp.965-74, 2009.

M. Osanai, N. Nishikiori, M. Murata, H. Chiba, T. Hansen et al., Cellular retinoic acid bioavailability determines epithelial integrity : Role of retinoic acid receptor alpha agonists in colitis, Mol Pharmacol, 2007.

D. I. Thurnham, C. A. Northrop-clewes, F. Mccullough, B. S. Das, and P. G. Lunn, Innate Immunity, Gut Integrity, and Vitamin A in Gambian and Indian Infants, J INFECT DIS. sept, vol.182, issue.s1, pp.23-31, 2000.

H. Wang, P. Wang, W. X. Wan, Y. Liu, and Y. , Butyrate Enhances Intestinal Epithelial Barrier Function via Up-Regulation of Tight Junction Protein Claudin-1 Transcription, Dig Dis Sci. déc, vol.57, issue.12, pp.3126-3161, 2012.

L. Peng, Z. Li, R. S. Green, I. R. Holzman, and J. Lin, Butyrate Enhances the Intestinal Barrier by Facilitating Tight Junction Assembly via Activation of AMP-Activated Protein Kinase in Caco-2 Cell Monolayers, J Nutr. sept, vol.139, issue.9, pp.1619-1644, 2009.

M. Nébot-vivinus, C. Harkat, H. Bzioueche, C. Cartier, R. Plichon-dainese et al., Multispecies probiotic protects gut barrier function in experimental models, World J Gastroenterol. 14 juin, vol.20, issue.22, pp.6832-6875, 2014.

S. Holowacz, C. Guigné, and G. Chêne, Probiotic supplementation reduced obesity, insulin resistance and gene expression modulating inflammation in gut and adipose tissus of mice fed a high-fat diet, Pharmanutrition, 2015.

H. Szajewska, M. Kotowska, and J. Mrukowicz, Efficacy of Lactobacillus GG in prevention of nosocomial diarrhea in infants, Journal of Pediatrics, vol.138, pp.361-366, 2001.

V. Rosenfeldt, K. Michaelsen, M. Jakobsen, C. Larsen, and P. Moller, Effect of probiotic Lactobacillus strains on acute diarrhea in a cohort of nonhospitalized children attending day-care centers, Pediatric Infectious Disease Journal, vol.21, pp.417-426, 2002.

S. Siitonen, H. Vapaatalo, S. Salminen, A. Gordin, M. Saxelin et al., Effect of Lactobacillus GG Yoghurt in Prevention of Antibiotic Associated Diarrhoea, Annals of Medicine. 1 janv, vol.22, issue.1, pp.57-66, 1990.

H. Szajewska and J. Z. Mrukowicz, Probiotics in the Treatment and Prevention of Acute Infectious Diarrhea in Infants and Children: A Systematic Review of Published Randomized, Double-Blind, Placebo-Controlled Trials, Journal of Pediatric Gastroenterology and Nutrition, vol.33, pp.17-25, 2001.

K. Sindhu, T. V. Sowmyanarayanan, A. Paul, S. Babji, S. Ajjampur et al., Immune Response and Intestinal Permeability in Children With Acute Gastroenteritis Treated With Lactobacillus rhamnosus GG: A Randomized, Double-Blind, Placebo-Controlled Trial, Clin Infect Dis. 15 avr, vol.58, issue.8, pp.1107-1122, 2014.

E. Dinleyici, A. Kara, M. Ozen, and Y. Vandenplas, Saccharomyces boulardii CNCM I-745 in diffrent clinical conditions, Expert Opin Biol Ther, 2014.

G. Villarruel, D. M. Rubio, F. Lopez, J. Cintioni, R. Gurevech et al., Saccharomyces boulardii in acute childhood diarrhoea: a randomized, placebo-controlled study, Acta Paediatrica, vol.96, issue.4, pp.538-579, 2007.

M. F. De-la-cochetiere, T. Durand, P. Lepage, A. Bourreille, J. P. Galmiche et al., Resilience of the Dominant Human Fecal Microbiota upon Short-Course Antibiotic Challenge, Journal of Clinical Microbiology, vol.43, issue.11, pp.5588-92, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02682740

I. Mangin, A. Suau, M. Gotteland, O. Brunser, and P. Pochart, Amoxicillin treatment modifies the composition of Bifidobacterium species in infant intestinal microbiota, Anaerobe. août, vol.16, issue.4, pp.433-441, 2010.

H. Szajewska and M. Ko?odziej, Systematic review with meta-analysis: Saccharomyces boulardii in the prevention of antibiotic-associated diarrhoea, Alimentary Pharmacology & Therapeutics, vol.42, issue.7, pp.793-801, 2015.

H. Szajewska and J. Mrukowicz, Meta-analysis: non-pathogenic yeast Saccharomyces boulardii in the prevention of antibiotic-associated diarrhoea, Alimentary Pharmacology & Therapeutics, vol.22, issue.5, pp.365-72, 2005.

R. Pattani, V. A. Palda, S. W. Hwang, and P. S. Shah, Probiotics for the prevention of antibioticassociated diarrhea and Clostridium difficile infection among hospitalized patients: systematic review and meta-analysis, Open Med. 28 mai, vol.7, issue.2, pp.56-67, 2013.

C. Neut, S. Mahieux, and L. J. Dubreuil, Antibiotic susceptibility of probiotic strains: Is it reasonable to combine probiotics with antibiotics? Médecine et Maladies Infectieuses, vol.47, pp.477-83, 2017.

S. Plummer, M. A. Weaver, J. C. Harris, P. Dee, and J. Hunter, INTERNATIONAL MICROBIOLOGY, vol.7, p.4, 2004.

I. L. Khalif, E. Quigley, E. A. Konovitch, and I. D. Maximova, Alterations in the colonic flora and intestinal permeability and evidence of immune activation in chronic constipation, Digestive and Liver Disease, vol.37, issue.11, pp.838-887, 2005.

E. Husebye, P. M. Hellström, F. Sundler, J. Chen, and T. Midtvedt, Influence of microbial species on small intestinal myoelectric activity and transit in germ-free rats, American Journal of Physiology-Gastrointestinal and Liver Physiology. 1 mars, vol.280, issue.3, pp.368-80, 2001.

, The Effect of Lactobacillus reuteri Supplementation in Adults with Chronic Functional Constipation: a Randomized, Double-Blind, Placebo-Controlled Trial, J Gastrointestin Liver Dis [Internet]. 1 déc, vol.23, issue.4, 2014.

D. Piano, M. Carmagnola, S. Anderloni, A. Andorno, S. Ballare et al., The use of probiotics in healthy volunteers with evacuation disorders and hard stools: a double-blind, randomized, placebo-controlled study, J Clin Gastroenterol, 2010.

M. M. Mazlyn, L. Nagarajah, F. A. Norimah, A. K. Goh, and K. , Effects of a probiotic fermented milk on functional constipation: A randomized, double-blind, placebocontrolled study: Probiotics for functional constipation, J Gastroenterol Hepatol. juill, vol.28, issue.7, pp.1141-1148, 2013.

G. Riezzo, A. Orlando, D. 'attoma, B. Guerra, V. Valerio et al., Randomised clinical trial: efficacy of Lactobacillus paracasei-enriched artichokes in the treatment of patients with functional constipation -a double-blind, controlled, crossover study, Aliment Pharmacol Ther. févr, vol.35, issue.4, pp.441-50, 2012.

L. Bu, M. Chang, Y. Ni, H. Chen, and C. Cheng, Lactobacillus casei rhamnosus Lcr35 in children with chronic constipation, Pediatr Int. août, vol.49, issue.4, pp.485-90, 2007.

T. Li, X. Lu, and X. Yang, Stachyose-Enriched ?-Galacto-oligosaccharides Regulate Gut Microbiota and Relieve Constipation in Mice, J Agric Food Chem. 4 déc, vol.61, issue.48, pp.11825-11856, 2013.

K. Miki and D. Moore, The sugar permability test reflects disease activity in children and adolescents with inflammatory bowel disease, The Journal of Pediatrics, pp.750-754, 1998.

D. N. Frank, . St, A. L. Amand, R. A. Feldman, E. C. Boedeker et al., Molecularphylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases, Proc Natl Acad Sci, vol.104, issue.34, pp.13780-13785, 2007.

C. Manichanh, Reduced diversity of faecal microbiota in Crohn's disease revealed by a metagenomic approach, Gut. 1 févr, vol.55, issue.2, pp.205-216, 2006.

H. Sokol, B. Pigneur, L. Watterlot, O. Lakhdari, L. G. Bermudez-humaran et al., Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients, Proceedings of the National Academy of Sciences, vol.105, pp.16731-16737, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00652961

E. Quévrain, M. A. Maubert, C. Michon, F. Chain, R. Marquant et al., Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn's disease, Gut. mars, vol.65, issue.3, pp.415-440, 2016.

S. Rajca, V. Grondin, E. Louis, G. Vernier-massouille, J. Grimaud et al., Alterations in the Intestinal Microbiome (Dysbiosis) as a Predictor of Relapse After Infliximab Withdrawal in Crohn?s Disease, Inflammatory Bowel Diseases. avr, vol.1, 2014.

A. Darfeuille-michaud, J. Boudeau, P. Bulois, C. Neut, A. Glasser et al., High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn's disease, Gastroenterology. août, vol.127, issue.2, pp.412-433, 2004.

N. Barnich and A. Darfeuille-michaud, Adherent-invasive Escherichia coli and Crohn's disease. Current Opinion in Gastroenterology, vol.23, pp.16-20, 2007.
URL : https://hal.archives-ouvertes.fr/hal-02539567

A. Bourreille, G. Cadiot, L. Dreau, G. Laharie, D. Beaugerie et al., Saccharomyces boulardii Does Not Prevent Relapse of Crohn's Disease, Clinical Gastroenterology and Hepatology. août, vol.11, issue.8, pp.982-989, 2013.

P. Marteau, M. Lémann, P. Seksik, D. Laharie, J. F. Colombel et al., Ineffectiveness of Lactobacillus johnsonii LA1 for prophylaxis of postoperative recurrence in Crohn's disease: a randomised, double blind, placebo controlled GETAID trial, Gut. juin, vol.55, issue.6, pp.842-849, 2006.

R. Rahimi, S. Nikfar, F. Rahimi, B. Elahi, S. Derakhshani et al., A Meta-Analysis on the Efficacy of Probiotics for Maintenance of Remission and Prevention of Clinical and Endoscopic Relapse in Crohn's Disease, Dig Dis Sci. sept, vol.53, issue.9, pp.2524-2555, 2008.

J. O. Lindsay, Clinical, microbiological, and immunological effects of fructooligosaccharide in patients with Crohn's disease, Gut. 1 mars, vol.55, issue.3, pp.348-55, 2006.

K. Gecse, R. Roka, and T. Sera, Leaky gut in patients with diarrhea-predominant irritable syndrome and inactive ulcerative colitis, Digestion, vol.85, pp.40-46, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02649940

E. Varela, C. Manichanh, M. Gallart, A. Torrejón, N. Borruel et al., Colonisation by Faecalibacterium prausnitzii and maintenance of clinical remission in patients with ulcerative colitis, Aliment Pharmacol Ther. juill, vol.38, issue.2, pp.151-61, 2013.

A. Tursi, G. Brandimarte, A. Papa, A. Giglio, W. Elisei et al., Treatment of Relapsing Mild-to-Moderate Ulcerative Colitis With the Probiotic VSL#3 as Adjunctive to a Standard Pharmaceutical Treatment: A Double-Blind, Randomized, Placebo-Controlled Study, American Journal of Gastroenterology, vol.105, issue.10, pp.2218-2245, 2010.

A. Sood, V. Midha, G. K. Makharia, V. Ahuja, D. Singal et al., The Probiotic Preparation, VSL#3 Induces Remission in Patients With Mild-to-Moderately Active Ulcerative Colitis, Clinical Gastroenterology and Hepatology, vol.7, issue.11, pp.1202-1209, 2009.

E. Miele, F. Pascarella, E. Giannetti, L. Quaglietta, R. Baldassano et al., Effect of a Probiotic Preparation (VSL# 3) on Induction and Maintenance of Remission in Children With Ulcerative Colitis, The American journal of gastroenterology. 1 févr, vol.104, pp.437-480, 2009.

W. Kruis, E. Schutz, P. Fric, B. Fixa, G. Judmaier et al., Double-blind comparison of an oral Escherichia coli preparation and mesalazine in maintaining remission of ulcerative colitis, Aliment Pharmacol Ther, vol.11, issue.5, pp.853-861, 1997.

B. Rembacken, A. Snelling, P. Hawkey, D. Chalmers, and A. Axon, Non-pathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis: a randomised trial. The Lancet, août, vol.21, issue.9179, pp.635-644, 1999.

W. Kruis, Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine, Gut, vol.53, issue.11, pp.1617-1640, 2004.

K. Kato, S. Mizuno, Y. Umesaki, Y. Ishii, M. Sugitani et al., Randomized placebo-controlled trial assessing the effect of bifidobacteria-fermented milk on active ulcerative colitis, Aliment Pharmacol Ther, vol.20, issue.10, pp.1133-1174, 2004.

M. A. Zocco, L. Z. Verme, F. Cremonini, A. C. Piscaglia, E. C. Nista et al., Efficacy of Lactobacillus GG in maintaining remission of ulcerative colitis, Aliment Pharmacol Ther. juin, vol.23, issue.11, pp.1567-74, 2006.

S. Oliva, D. Nardo, G. Ferrari, F. Mallardo, S. Rossi et al., Randomised clinical trial: the effectiveness of Lactobacillus reuteri ATCC 55730 rectal enema in children with active distal ulcerative colitis: Randomised clinical trial: rectal Lactobacillus reuteri in ulcerative colitis, Alimentary Pharmacology & Therapeutics. févr, vol.35, issue.3, pp.327-361, 2012.

L. Sang, Remission induction and maintenance effect of probiotics on ulcerative colitis: A meta-analysis, WJG, vol.16, issue.15, p.1908, 2010.

F. Casellas, N. Borruel, A. Torrejón, E. Varela, M. Antolin et al., Oral oligofructose-enriched inulin supplementation in acute ulcerative colitis is well tolerated and associated with lowered faecal calprotectin: Inulin supplementation in acute ulcerative colitis, Alimentary Pharmacology & Therapeutics. 16 févr, vol.25, issue.9, pp.1061-1068, 2007.

N. Bertiaux-vandaele, S. Youmba, and L. Belmonte, The expression and the cellular distribution of the tight junction proteins are altered in irritable bowel syndrome patients with differences according to the disease subtype, American Journal of Gastroenterology, vol.106, issue.12, pp.2165-73, 2011.

J. Bertrand, I. Ghouzali, and C. Guerin, Glutamine restores tight junction protein Claudin-1 expression in colonic mucosa of patients with diarrhea-predominant irritable bowel syndrome, Journal of Parenteral and Enteral Nutrition, vol.40, pp.1170-1176, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02288963

S. Miquel, R. Martín, A. Lashermes, M. Gillet, M. Meleine et al., Anti-nociceptive effect of Faecalibacterium prausnitzii in non-inflammatory IBS-like models, Sci Rep. mai, vol.6, issue.1, p.19399, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01263911

K. N. Lee and O. Y. Lee, Intestinal microbiota in pathophysiology and management of irritable bowel syndrome, World J Gastroenterol. 21 juill, vol.20, issue.27, pp.8886-97, 2014.

A. Balsari, A. Ceccarelli, F. Dubini, E. Fesce, and G. Poli, The fecal microbial population in the irritable bowel syndrome, Microbiologica, 1982.

S. Drouault-holowacz, S. Bieuvelet, A. Burckel, M. Cazaubiel, X. Dray et al., A double blind randomized controlled trial of a probiotic combination in 100 patients with irritable bowel syndrome, Gastroentérologie Clinique et Biologique. févr, vol.32, issue.2, pp.147-52, 2008.

F. Yuan, H. Ni, C. V. Asche, M. Kim, S. Walayat et al., Efficacy of Bifidobacterium infantis 35624 in patients with irritable bowel syndrome: a meta-analysis, Current Medical Research and Opinion. 3 juill, vol.33, issue.7, pp.1191-1198, 2017.

S. Nobaek, M. Johansson, G. Molin, S. Ahrne, and B. Jeppsson, Alteration of intestinal microflora is associated with reduction in abdominal bloating and pain in patients with irritable bowel syndrome, American Journal of Gastroenterology, vol.95, pp.1231-1239, 2000.

K. Niedzielin, H. Kordecki, and B. Birkenfeld, A controlled, double-lind, randomized study on the efficacy of Lactobacillus plantarum 299V in patients with irritable bowel syndrome, European Journal of Gastroenterology & Hepatology, vol.13, pp.1143-1150, 2001.

M. Johansson, S. Nobaek, A. Berggren, M. Nyman, I. Björck et al., Survival of Lactobacillus plantarum DSM 9843 (299v), and effect on the short-chain fatty acid content of faeces after ingestion of a rose-hip drink with fermented oats, International Journal of Food Microbiology. juin, vol.42, issue.1-2, pp.29-38, 1998.

S. Xiao, D. Zhang, S. Jiang, H. Liu, G. Wang et al., Multicenter, randomized, controlled trial of heat-killed Lactobacillus acidophilus LB in patients with chronic diarrhea, Adv Ther, 2003.

C. Rousseaux, X. Thuru, A. Gelot, N. Barnich, C. Neut et al., Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors, Nature medicine. 1 févr, vol.13, pp.35-42, 2007.
URL : https://hal.archives-ouvertes.fr/hal-02667010

D. Silk, A. Davis, J. Vulevic, G. Tzortzis, and G. R. Gibson, Clinical trial: the effects of a trans-galactooligosaccharide prebiotic on faecal microbiota and symptoms in irritable bowel syndrome, Alimentary Pharmacology & Therapeutics. mars, vol.29, issue.5, pp.508-526, 2009.

F. Joly, C. Mayeur, A. Bruneau, M. Noordine, T. Meylheuc et al., Drastic changes in fecal and mucosa-associated microbiota in adult patients with short bowel syndrome, Biochimie. juill, vol.92, issue.7, pp.753-61, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02662544

Z. H. Davidovics, B. A. Carter, R. A. Luna, E. B. Hollister, R. J. Shulman et al., The Fecal Microbiome in Pediatric Patients With Short Bowel Syndrome, JPEN J Parenter Enteral Nutr, vol.40, issue.8, pp.1106-1119, 2016.

E. Lilja, H. Wefer, H. Nyström, N. Finkel, Y. Engstrand et al., Intestinal dysbiosis in children with short bowel syndrome is associated with impaired outcome, vol.3, 2015.

A. Zaouche-cl-p-de-lagausie, M. Peuchmaur, . Macry, P. Fitoussi, . Bernasconi et al., Effects of Oral Saccharomyces boulardii on Bacterial Overgrowth, Translocation, and Intestinal Adaptation after Small-Bowel Resection in Rats, Scandinavian Journal of Gastroenterology. 1 janv, vol.35, issue.2, pp.160-165, 2000.

T. Sentongo, V. Cohran, S. Korff, K. Iyer, and X. Zheng, Intestinal permeability and effets of Lactobacillus rhamnosus therapy in children with short bowel syndrome, J Pediatric Gastroenterology Nutr, 2008.

E. Sadoun-journo, J. Gaillard, H. Blehaut, O. Goulet, P. Bernasconi et al., Grêle court dysfonctionnel compliqué de pullulation bactérienne chez l'enfant : effet de Saccharomyces boulardii, Gastroenterology Clin Biol, 1994.

M. C. Collado, E. Donat, C. Ribes-koninckx, M. Calabuig, and Y. Sanz, Specific duodenal and faecal bacterial groups associated with paediatric coeliac disease, Journal of Clinical Pathology. 1 mars, vol.62, issue.3, pp.264-273, 2009.

D. Cagno, R. , D. Angelis, M. , D. Pasquale et al., Duodenal and faecal microbiota of celiac children: molecular, phenotype and metabolome characterization, BMC Microbiol, vol.11, p.219, 2011.

H. J. Galipeau, J. L. Mccarville, S. Huebener, O. Litwin, M. Meisel et al., Intestinal Microbiota Modulates Gluten-Induced Immunopathology in Humanized Mice, Am J Pathol, vol.185, issue.11, pp.2969-82, 2015.

J. Cinova, D. Palma, G. Stepankova, R. Kofronova, O. Kverka et al., Role of Intestinal Bacteria in Gliadin-Induced Changes in Intestinal Mucosa: Study in Germ-Free Rats, Disponible sur, vol.6, 2011.

A. Orlando, M. Linsalata, M. Notarnicola, V. Tutino, and F. Russo, Lactobacillus GG restoration of the gliadin induced epithelial barrier disruption: the role of cellular polyamines, BMC Microbiol. 31 janv, vol.14, p.19, 2014.

C. Papista, V. Gerakopoulos, A. Kourelis, M. Sounidaki, A. Kontana et al., Gluten induces coeliac-like disease in sensitised mice involving IgA, CD71 and transglutaminase 2 interactions that are prevented by probiotics, Lab Invest. avr, vol.92, issue.4, pp.625-660, 2012.

R. D'arienzo, R. Stefanile, F. Maurano, G. Mazzarella, R. E. Troncone et al., Immunomodulatory Effects of Lactobacillus casei Administration in a Mouse Model of Gliadin-Sensitive Enteropathy: L. casei Modulates Gliadin-Sensitive Enteropathy, Scandinavian Journal of Immunology, vol.74, issue.4, pp.335-376, 2011.

K. Lindfors, T. Blomqvist, K. Juuti-uusitalo, S. Stenman, J. Venäläinen et al., Live probiotic Bifidobacterium lactis bacteria inhibit the toxic effects induced by wheat gliadin in epithelial cell culture, Clinical & Experimental Immunology, vol.152, issue.3, pp.552-560, 2008.

M. Klemenak, J. Dolin?ek, T. Langerholc, D. Gioia, D. Mi?eti?-turk et al., Administration of Bifidobacterium breve decreases the production of TNF-? in children with celiac disease, vol.60, pp.3386-92, 2015.

M. Medina, D. Palma, G. Ribes-koninckx, C. Calabuig, M. Sanz et al., Bifidobacterium strains suppress in vitro the pro-inflammatory milieu triggered by the large intestinal microbiota of coeliac patients, J Inflamm, vol.5, p.19, 2008.

P. Marteau and F. Shanahan, Basic aspects and pharmacology of probiotics: an overview of pharmacokinetics, mechanisms of action and side-effects. Best Practice & Research Clinical Gastroenterology, vol.17, pp.725-765, 2003.

M. Liong, Safety of probiotics: translocation and infection, Nutrition Reviews. 26 mars, vol.66, issue.4, pp.192-202, 2008.

P. Marteau, M. F. Gerhardt, A. Myara, E. Bouvier, F. Trivin et al., Metabolism of Bile Salts by Alimentary Bacteria During Transit in the Human Small Intestine. Microbial Ecology in Health and Disease, janv, vol.8, issue.4, pp.151-158, 1995.

E. Connolly, T. Abrahamsson, and B. Björksten, Safety of D(-)-lactic acid producing bacteria in the human infant, J Pediatr Gastroenterol Nutr, 2005.

R. B. Sartor, T. M. Bond, and J. H. Schwab, Systemic uptake and intestinal inflammatory effects of luminal bacterial cell wall polymers in rats with acute colonic injury, Infect Immun. août, vol.56, issue.8, pp.2101-2109, 1988.

. Butel, Les probiotiques et leur place en médecine humaine, J Anti-Infect, 2014.