, Malgré un profil de glycosylation apparemment médiocre (344), les parasites des paludismes murin (P. berghei, P. yoelii) et humain (P. falciparum) présentent des glycanes ?-Gal à la surface des sporozoïtes (324)(345) ainsi qu'au stade érythrocytaire de l'infection (324)(346)(347)(348)

, Pourtant, aucun gène orthologue ?1,3GT n'a encore été identifié chez Plasmodium spp

, Des niveaux résiduels d'?-Gal ont été détectés dans les glandes salivaires de moustiques non infectés, p.3

, origine des glycanes ?-Gal détectés dans les sporozoïtes de Plasmodium spp

, Ces derniers comprenennent des membres Gram (-) de Klebsiella, Serratia, et E. coli ainsi que des Gram (+) telles que Lactobacillus casei (345). L'expression d'?-Gal par ces entérobactéries est associée aux glycoprotéines de la capsule et de la paroi cellulaire, Contrairement aux humains, les souris de type sauvage possèdent un gène GGTA1 fonctionnel et expriment l'?-Gal à la surface de leurs cellules, inhibant le développement de l'immunité anti-?-Gal (351)

, La colonisation de l'intestin par le pathobionte humain Escherichia coli O86:B7 exprimant des taux élevés d'?-Gal (357) résume l'étiologie de la production d'AC anti-?-Gal chez la souris (358) et chez les primates (359), ainsi que la production d'AC dirigés contre le glycane du groupe sanguin B apparenté à ?-Gal

, Cela suggère que la colonisation de l'intestin par E. coli O86:B7 pourrait être particulièrement utile pour déclencher la production d'AC spécifiques de l'?-Gal, contribuant probablement aux titres élevés de ces AC circulants chez l'adulte en bonne santé (345)

. Oms and . Paludisme,

O. Romoli and M. Gendrin, The tripartite interactions between the mosquito, its microbiota and Plasmodium, Parasites & Vectors. 20 mars, vol.11, 0200.

C. Landman and E. Quévrain,

, Rev Med Interne. juin, vol.37, issue.6, pp.418-441, 2016.

F. Berrilli, D. Cave, D. Cavallero, S. D'amelio, and S. , Interactions between parasites and microbial communities in the human gut, Front Cell Infect Microbiol, vol.2, p.141, 2012.

, Institut Pasteur. Paludisme, 2013.

, Muséum national d'Histoire naturelle

, Campus de Parasitologie-Mycologie -Association Française des Enseignants de Parasitologie et Mycologie (ANOFEL). Paludisme, 2016.

E. P. Ecn, ALINEA Plus, vol.5, 2018.

. Inserm and . Paludisme, Le parasite P. vivax infecte des populations considérées comme résistantes

R. Ross, On some Peculiar Pigmented Cells Found in Two Mosquitos Fed on Malarial Blood, Br Med J. 18 déc, vol.1897, pp.1786-1794, 1929.

B. Grassi and A. Bignami, Ulteriori ricerche sul ciclo dei parassiti malarici umani nel corpo del zanzarone, 1899.

P. Carnevale and V. Robert, Les anophèles : biologie, transmission du Plasmodium et lutte antivectorielle

R. E. Harbach, The classification of genus Anopheles (Diptera: Culicidae): a working hypothesis of phylogenetic relationships, Bull Entomol Res. déc, vol.94, issue.6, pp.537-53, 2004.

. Oms and . Paludisme, Alerte vectorielle: invasion et propagation d'Anopheles stephensi

, Médecine tropicale. Paludisme actualités, 2018.

C. López, C. Saravia, A. Gomez, J. Hoebeke, and M. A. Patarroyo, Mechanisms of geneticallybased resistance to malaria, Gene, vol.467, issue.1-2, pp.1-12, 2010.

F. B. Piel, The Present and Future Global Burden of the Inherited Disorders of Hemoglobin, Hematol Oncol Clin North Am. avr, vol.30, issue.2, pp.327-368, 2016.

, OMS. Paludisme -Groupes à haut risque

P. Bourée and S. Delaigue, Paludisme : avantages et inconvénients des tests de diagnostic rapide, Médecine thérapeutique / Pédiatrie. 1 avr, vol.18, issue.2, pp.109-121, 2015.

F. Bourdillon, BEH -Recommandations sanitaires pour les voyageurs, 2019.

, OMS. Paludisme -Résistance aux insecticides

, OMS. Paludisme -Résistance aux médicaments antipaludiques

, Avis relatif aux recommandations de prévention du paludisme pour les voyageurs, HCSP, vol.17, 2015.

. Oms and . Paludisme, Retrait des monothérapies à base d'artémisinine par voie orale

J. G. Kublin, S. A. Mikolajczak, B. K. Sack, M. E. Fishbaugher, A. Seilie et al., Complete attenuation of genetically engineered Plasmodium falciparum sporozoites in human subjects, Sci Transl Med, vol.04, issue.371, 2017.

Y. Dong, F. Manfredini, and G. Dimopoulos, Implication of the mosquito midgut microbiota in the defense against malaria parasites, PLoS Pathogens. 1 mai, vol.5, issue.5, p.1000423, 2009.

F. H. Rodgers, M. Gendrin, C. Wyer, and G. K. Christophides, Microbiota-induced peritrophic matrix regulates midgut homeostasis and prevents systemic infection of malaria vector mosquitoes, PLoS Pathogens. 1 mai, vol.13, issue.5, p.1006391, 2017.

R. E. Sinden and P. F. Billingsley, Plasmodium invasion of mosquito cells: hawk or dove?, Trends Parasitol. mai, vol.17, issue.5, pp.209-221, 2001.

H. Alout, N. T. Ndam, M. M. Sandeu, I. Djégbe, F. Chandre et al., Insecticide Resistance Alleles Affect Vector Competence of Anopheles gambiae s.s. for Plasmodium falciparum Field Isolates, PLoS One. 21 mai, vol.8, issue.5, 2013.

, The Journey of the Malaria Parasite in the Mosquito: Hopes for the New Century, Parasitology Today. 1 mai, vol.16, issue.5, pp.196-201, 2000.

K. Michel and F. C. Kafatos, Mosquito immunity against Plasmodium, Insect Biochem Mol Biol. juill, vol.35, issue.7, pp.677-89, 2005.

D. Vlachou, T. Schlegelmilch, G. K. Christophides, and F. C. Kafatos, Functional genomic analysis of midgut epithelial responses in Anopheles during Plasmodium invasion, Curr Biol. 12 juill, vol.15, issue.13, pp.1185-95, 2005.

S. Kumar, A. Molina-cruz, L. Gupta, J. Rodrigues, and C. Barillas-mury, A peroxidase/dual oxidase system modulates midgut epithelial immunity in Anopheles gambiae, Science. 26 mars, vol.327, issue.5973, pp.1644-1652, 2010.

C. B. Pumpuni, J. Demaio, M. Kent, J. R. Davis, and J. C. Beier, Bacterial population dynamics in three anopheline species: the impact on Plasmodium sporogonic development, Am J Trop Med Hyg. févr, vol.54, issue.2, pp.214-222, 1996.

A. C. Bahia, Y. Dong, B. J. Blumberg, G. Mlambo, A. Tripathi et al., Exploring Anopheles gut bacteria for Plasmodium blocking activity, Environmental Microbiology. 15 janv, vol.16, issue.9, pp.2980-94, 2014.

J. L. Ramirez, S. M. Short, A. C. Bahia, R. G. Saraiva, Y. Dong et al., Chromobacterium Csp_P reduces malaria and dengue infection in vector mosquitoes and has entomopathogenic and in vitro anti-pathogen activities, PLoS Pathogens, vol.10, issue.10, p.1004398, 2014.

C. M. Cirimotich, Y. Dong, A. M. Clayton, S. L. Sandiford, J. A. Souza-neto et al., Natural Microbe-Mediated Refractoriness to Plasmodium Infection in Anopheles gambiae, Science. 13 mai, vol.332, issue.6031, pp.855-863, 2011.

N. J. Dennison, R. G. Saraiva, C. M. Cirimotich, G. Mlambo, E. F. Mongodin et al., Functional genomic analyses of Enterobacter, Anopheles and Plasmodium reciprocal interactions that impact vector competence, Malar J, vol.22, issue.1, p.425, 2016.

S. Meister, B. Agianian, F. Turlure, A. Relógio, I. Morlais et al., Anopheles gambiae PGRPLC-mediated defense against bacteria modulates infections with malaria parasites, PLoS Pathogens. 1 août, vol.5, issue.8, p.1000542, 2009.

M. Huber, E. Cabib, and L. H. Miller, Malaria parasite chitinase and penetration of the mosquito peritrophic membrane, PNAS. 1 avr, vol.88, issue.7, pp.2807-2817, 1991.

Y. Tsai, R. E. Hayward, R. C. Langer, D. A. Fidock, and J. M. Vinetz, Disruption of Plasmodium falciparumChitinase Markedly Impairs Parasite Invasion of Mosquito Midgut, Infection and Immunity. 1 juin, vol.69, issue.6, pp.4048-54, 2001.

C. M. Cirimotich, J. L. Ramirez, and G. Dimopoulos, Native Microbiota Shape Insect Vector Competence for Human Pathogens, Cell Host & Microbe, vol.10, issue.4, pp.307-317, 2011.

L. Gonzalez-ceron, F. Santillan, M. H. Rodriguez, D. Mendez, and J. E. Hernandez-avila, Bacteria in midguts of field-collected Anopheles albimanus block Plasmodium vivax sporogonic development, J Med Entomol. mai, vol.40, issue.3, pp.371-375, 2003.

S. Wang, A. K. Ghosh, N. Bongio, K. A. Stebbings, D. J. Lampe et al., Fighting malaria with engineered symbiotic bacteria from vector mosquitoes, PNAS. 31 juill, vol.109, issue.31, pp.12734-12743, 2012.

, Microbiote intestinal (flore intestinale)

D. Gevers, R. Knight, J. F. Petrosino, K. Huang, A. L. Mcguire et al., The Human Microbiome Project: A Community Resource for the Healthy Human Microbiome, PLoS Biol. 14 août, vol.10, issue.8, 2012.

F. Bäckhed, R. E. Ley, J. L. Sonnenburg, D. A. Peterson, and J. I. Gordon, Host-bacterial mutualism in the human intestine, Science. 25 mars, vol.307, issue.5717, pp.1915-1935, 2005.

A. S. Neish, Microbes in gastrointestinal health and disease, Gastroenterology. janv, vol.136, issue.1, pp.65-80, 2009.

. Bengmark-s, Ecological control of the gastrointestinal tract. The role of probiotic flora, Gut. janv, vol.42, issue.1, pp.2-7, 1998.

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.

R. Sender, S. Fuchs, and R. Milo, Revised Estimates for the Number of Human and Bacteria Cells in the Body, PLoS Biol, vol.14, issue.8, p.1002533, 2016.

T. D. Luckey, Introduction to intestinal microecology, Am J Clin Nutr. déc, vol.25, issue.12, pp.1292-1296, 1972.

A. A. Fodor, T. Z. Desantis, K. M. Wylie, J. H. Badger, Y. Ye et al., The « most wanted » taxa from the human microbiome for whole genome sequencing, PLoS ONE, vol.7, issue.7, p.41294, 2012.

J. Qin, R. Li, J. Raes, M. Arumugam, K. S. Burgdorf et al., A human gut microbial gene catalogue established by metagenomic sequencing, Nature. 4 mars, vol.464, issue.7285, pp.59-65, 2010.
URL : https://hal.archives-ouvertes.fr/cea-00908974

M. Dave, P. D. Higgins, S. Middha, and K. P. Rioux, The human gut microbiome: current knowledge, challenges, and future directions, Transl Res, vol.160, issue.4, pp.246-57, 2012.

, Encyclopédie de l'environnement. Les microbiotes humains : des alliés pour notre santé

H. B. Nielsen, M. Almeida, A. S. Juncker, S. Rasmussen, J. Li et al., Identification and assembly of genomes and genetic elements in complex metagenomic samples without using reference genomes, Nat Biotechnol. août, vol.32, issue.8, pp.822-830, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01195477

J. Schluter and K. R. Foster, The evolution of mutualism in gut microbiota via host epithelial selection, PLoS Biol, vol.10, issue.11, p.1001424, 2012.

E. K. Costello, C. L. Lauber, M. Hamady, N. Fierer, J. I. Gordon et al., Bacterial community variation in human body habitats across space and time, Science. 18 déc, vol.326, issue.5960, pp.1694-1701, 2009.

A. E. Pérez-cobas, M. J. Gosalbes, A. Friedrichs, H. Knecht, A. Artacho et al., Gut microbiota disturbance during antibiotic therapy: a multi-omic approach, Gut, vol.62, issue.11, pp.1591-601, 2013.

J. Li, H. Jia, X. Cai, H. Zhong, Q. Feng et al., An integrated catalog of reference genes in the human gut microbiome, Nat Biotechnol. août, vol.32, issue.8, pp.834-875, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01195478

A. Moya and M. Ferrer, Functional Redundancy-Induced Stability of Gut Microbiota Subjected to Disturbance, Trends Microbiol. mai, vol.24, issue.5, pp.402-415, 2016.

A. J. Macpherson and K. D. Mccoy, Stratification and compartmentalisation of immunoglobulin responses to commensal intestinal microbes, Semin Immunol, vol.25, issue.5, pp.358-63, 2013.

G. P. Donaldson, S. M. Lee, and S. K. Mazmanian, Gut biogeography of the bacterial microbiota, Nat Rev Microbiol. janv, vol.14, issue.1, pp.20-32, 2016.

A. Lavelle, G. Lennon, O. O'sullivan, N. Docherty, A. Balfe et al., Spatial variation of the colonic microbiota in patients with ulcerative colitis and control volunteers, Gut. oct, vol.64, issue.10, pp.1553-61, 2015.

H. Li, J. P. Limenitakis, T. Fuhrer, M. B. Geuking, M. A. Lawson et al., The outer mucus layer hosts a distinct intestinal microbial niche, Nat Commun. 22 sept, vol.6, p.8292, 2015.

C. L. Bevins and N. H. Salzman, Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis, Nat Rev Microbiol. mai, vol.9, issue.5, pp.356-68, 2011.

D. A. Peterson, N. P. Mcnulty, J. L. Guruge, and J. I. Gordon, IgA response to symbiotic bacteria as a mediator of gut homeostasis, Cell Host Microbe, vol.2, issue.5, pp.328-367, 2007.

J. L. Round and S. K. Mazmanian, The gut microbiota shapes intestinal immune responses during health and disease, Nat Rev Immunol. mai, vol.9, issue.5, pp.313-336, 2009.

P. B. Eckburg, E. M. Bik, C. N. Bernstein, E. Purdom, L. Dethlefsen et al., Diversity of the human intestinal microbial flora, Science. 10 juin, vol.308, issue.5728, pp.1635-1643, 2005.

P. Van-den-abbeele, C. Belzer, M. Goossens, M. Kleerebezem, D. Vos et al., Butyrate-producing Clostridium cluster XIVa species specifically colonize mucins in an in vitro gut model, ISME J. mai, vol.7, issue.5, pp.949-61, 2013.

J. M. Rodríguez, K. Murphy, C. Stanton, R. P. Ross, O. I. Kober et al., The composition of the gut microbiota throughout life, with an emphasis on early life, Microb Ecol Health Dis, vol.26, p.26050, 2015.

M. P. Francino, Early development of the gut microbiota and immune health, Pathogens. 24 sept, vol.3, issue.3, pp.769-90, 2014.

F. Bäckhed, Programming of host metabolism by the gut microbiota, Ann Nutr Metab, vol.58, issue.2, pp.44-52, 2011.

C. Palmer, E. M. Bik, D. B. Digiulio, D. A. Relman, and P. O. Brown, Development of the human infant intestinal microbiota, PLoS Biol. juill, vol.5, issue.7, p.177, 2007.

F. Bäckhed, J. Roswall, Y. Peng, Q. Feng, H. Jia et al., Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life, Cell Host Microbe. 10 juin, vol.17, issue.6, p.852, 2015.

A. Bergström, T. H. Skov, M. I. Bahl, H. M. Roager, L. B. Christensen et al., Establishment of Intestinal Microbiota during Early Life: a Longitudinal, Explorative Study of a Large Cohort of Danish Infants, Appl Environ Microbiol. 1 mai, vol.80, issue.9, pp.2889-900, 2014.

T. Mitsuoka, Development of Functional Foods. Bioscience of Microbiota, Food and Health, vol.33, pp.117-145, 2014.

S. Dogra, O. Sakwinska, S. Soh, C. Ngom-bru, W. M. Brück et al., Rate of establishing the gut microbiota in infancy has consequences for future health, Gut Microbes. 3 sept, vol.6, issue.5, pp.321-326, 2015.

S. Dogra, O. Sakwinska, S. Soh, C. Ngom-bru, W. M. Brück et al., Dynamics of infant gut microbiota are influenced by delivery mode and gestational duration and are associated with subsequent adiposity, MBio. 3 févr, vol.6, issue.1, 2015.

T. Yatsunenko, F. E. Rey, M. J. Manary, I. Trehan, M. G. Dominguez-bello et al., Human gut microbiome viewed across age and geography, Nature. 9 mai, vol.486, issue.7402, pp.222-229, 2012.

F. Purchiaroni, A. Tortora, M. Gabrielli, F. Bertucci, G. Gigante et al., The role of intestinal microbiota and the immune system, Eur Rev Med Pharmacol Sci. févr, vol.17, issue.3, pp.323-356, 2013.

L. T. Stiemsma and S. E. Turvey, Asthma and the microbiome: defining the critical window in early life, Allergy Asthma Clin Immunol, vol.13, p.3, 2017.

M. Tanaka and J. Nakayama, Development of the gut microbiota in infancy and its impact on health in later life, Allergol Int, vol.66, issue.4, pp.515-537, 2017.

J. Penders, C. Thijs, C. Vink, F. F. Stelma, B. Snijders et al., Factors influencing the composition of the intestinal microbiota in early infancy, Pediatrics. août, vol.118, issue.2, pp.511-532, 2006.

R. I. Mackie, A. Sghir, and H. R. Gaskins, Developmental microbial ecology of the neonatal gastrointestinal tract, Am J Clin Nutr, vol.69, issue.5, pp.1035-1045, 1999.

D. B. Digiulio, R. Romero, H. P. Amogan, J. P. Kusanovic, E. M. Bik et al., Microbial prevalence, diversity and abundance in amniotic fluid during preterm labor: a molecular and culture-based investigation, PLoS ONE. 26 août, vol.3, issue.8, p.3056, 2008.

D. B. Digiulio, R. Romero, J. P. Kusanovic, R. Gómez, C. J. Kim et al., Prevalence and diversity of microbes in the amniotic fluid, the fetal inflammatory response, and pregnancy outcome in women with preterm pre-labor rupture of membranes, Am J Reprod Immunol. 1 juill, vol.64, issue.1, pp.38-57, 2010.

R. Romero, J. Miranda, J. P. Kusanovic, T. Chaiworapongsa, P. Chaemsaithong et al., Clinical chorioamnionitis at term I: microbiology of the amniotic cavity using cultivation and molecular techniques, J Perinat Med. janv, vol.43, issue.1, pp.19-36, 2015.

E. Jiménez, L. Fernández, M. L. Marín, R. Martín, J. M. Odriozola et al., Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by cesarean section, Curr Microbiol, vol.51, issue.4, pp.270-274, 2005.

K. Aagaard, J. Ma, K. M. Antony, R. Ganu, J. Petrosino et al., The placenta harbors a unique microbiome, Sci Transl Med. 21 mai, vol.6, issue.237, pp.237-65, 2014.

E. Jiménez, M. L. Marín, R. Martín, J. M. Odriozola, M. Olivares et al., Is meconium from healthy newborns actually sterile?, Res Microbiol. avr, vol.159, issue.3, pp.187-93, 2008.

L. Moles, M. Gómez, H. Heilig, G. Bustos, S. Fuentes et al., Bacterial diversity in meconium of preterm neonates and evolution of their fecal microbiota during the first month of life, PLoS ONE, vol.8, issue.6, p.66986, 2013.

M. J. Gosalbes, S. Llop, Y. Vallès, A. Moya, F. Ballester et al., Meconium microbiota types dominated by lactic acid or enteric bacteria are differentially associated with maternal eczema and respiratory problems in infants, Clin Exp Allergy. févr, vol.43, issue.2, pp.198-211, 2013.

J. C. Madan, R. C. Salari, D. Saxena, L. Davidson, G. A. O'toole et al., Gut microbial colonisation in premature neonates predicts neonatal sepsis. Arch Dis Child Fetal Neonatal Ed, vol.97, pp.456-462, 2012.

S. Matamoros, C. Gras-leguen, L. Vacon, F. Potel, G. De-la-cochetiere et al., Development of intestinal microbiota in infants and its impact on health, Trends Microbiol. avr, vol.21, issue.4, pp.167-73, 2013.

R. Mändar and M. Mikelsaar, Transmission of mother's microflora to the newborn at birth, Biol Neonate, vol.69, issue.1, pp.30-35, 1996.

G. W. Tannock, R. Fuller, S. L. Smith, and M. A. Hall, Plasmid profiling of members of the family Enterobacteriaceae, lactobacilli, and bifidobacteria to study the transmission of bacteria from mother to infant, J Clin Microbiol. juin, vol.28, issue.6, pp.1225-1233, 1990.

. Nowrouzian, Escherichia coli in infants' intestinal microflora: colonization rate, strain turnover, and virulence gene carriage

L. V. Hooper, M. H. Wong, A. Thelin, L. Hansson, P. G. Falk et al., Molecular analysis of commensal host-microbial relationships in the intestine, Science. 2 févr, vol.291, issue.5505, pp.881-885, 2001.

R. Ducluzeau, Development, equilibrium and role of microbial flora in the newborn

, Ann Pediatr (Paris). janv, vol.40, issue.1, pp.13-22, 1993.

H. J. Harmsen, A. C. Wildeboer-veloo, G. C. Raangs, A. A. Wagendorp, N. Klijn et al., Analysis of intestinal flora development in breast-fed and formula-fed infants by using molecular identification and detection methods, J Pediatr Gastroenterol Nutr. janv, vol.30, issue.1, pp.61-68, 2000.

F. Turroni, C. Peano, D. A. Pass, E. Foroni, M. Severgnini et al., Diversity of bifidobacteria within the infant gut microbiota, PLoS ONE, vol.7, issue.5, p.36957, 2012.

B. Kleessen and J. Bezirtzoglou, Culture-Based Knowledge on Biodiversity, Development and Stability of Human Gastrointestinal Microflora, Microbial Ecology in Health and Disease. 1 janv, vol.12, issue.2, pp.53-63, 2000.

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 USA. 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.

G. Biasucci, B. Benenati, L. Morelli, E. Bessi, and G. Boehm, Cesarean delivery may affect the early biodiversity of intestinal bacteria, J Nutr. sept, vol.138, issue.9, pp.1796-1800, 2008.

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

A. Huurre, M. Kalliomäki, S. Rautava, M. Rinne, S. Salminen et al., Mode of delivery -effects on gut microbiota and humoral immunity, Neonatology, vol.93, issue.4, pp.236-276, 2008.

H. E. Jakobsson, T. R. Abrahamsson, M. C. Jenmalm, K. Harris, C. Quince et al., Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by caesarean section, Gut. avr, vol.63, issue.4, pp.559-66, 2014.

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.

S. Salminen, G. R. Gibson, A. L. Mccartney, and E. Isolauri, Influence of mode of delivery on gut microbiota composition in seven year old children, Gut. sept, vol.53, issue.9, pp.1388-1397, 2004.

E. Decker, M. Hornef, and S. Stockinger, Cesarean delivery is associated with celiac disease but not inflammatory bowel disease in children, Gut Microbes. avr, vol.2, issue.2, pp.91-99, 2011.

H. Renz-polster, M. R. David, A. S. Buist, W. M. Vollmer, E. A. O'connor et al., Caesarean section delivery and the risk of allergic disorders in childhood, Clin Exp Allergy, vol.35, issue.11, pp.1466-72, 2005.

S. Arboleya, L. Ang, A. Margolles, L. Yiyuan, Z. Dongya et al., Deep 16S rRNA metagenomics and quantitative PCR analyses of the premature infant fecal microbiota, Anaerobe. juin, vol.18, issue.3, pp.378-80, 2012.

S. Arboleya, A. Binetti, N. Salazar, N. Fernández, G. Solís et al., Establishment and development of intestinal microbiota in preterm neonates, FEMS Microbiol Ecol. mars, vol.79, issue.3, pp.763-72, 2012.

F. Magne, A. Suau, P. Pochart, and J. Desjeux, Fecal Microbial Community in Preterm Infants, Journal of Pediatric Gastroenterology and Nutrition, vol.41, issue.4, p.386, 2005.

E. Westerbeek, A. Van-den-berg, H. N. Lafeber, J. Knol, W. Fetter et al., The intestinal bacterial colonisation in preterm infants: a review of the literature, Clin Nutr. juin, vol.25, issue.3, pp.361-369, 2006.

H. Sakata, H. Yoshioka, and K. Fujita, Development of the intestinal flora in very low birth weight infants compared to normal full-term newborns, Eur J Pediatr. juill, vol.144, issue.2, pp.186-90, 1985.

A. Schwiertz, B. Gruhl, M. Löbnitz, P. Michel, M. Radke et al., Development of the intestinal bacterial composition in hospitalized preterm infants in comparison with breastfed, full-term infants, Pediatr Res. sept, vol.54, issue.3, pp.393-402, 2003.

J. E. Koenig, A. Spor, N. Scalfone, A. D. Fricker, J. Stombaugh et al., Succession of microbial consortia in the developing infant gut microbiome, Proc Natl Acad Sci, vol.108, issue.1, pp.4578-85, 2011.

N. J. Andreas, B. Kampmann, M. Le-doare, and K. , Human breast milk: A review on its composition and bioactivity. Early Hum Dev, vol.91, pp.629-664, 2015.

C. Boquien, Human Milk: An Ideal Food for Nutrition of Preterm Newborn. Front Pediatr, vol.6, 2018.

L. Bode, Human milk oligosaccharides: prebiotics and beyond, Nutr Rev, vol.67, issue.suppl_2, pp.183-91, 2009.

A. Marcobal, M. Barboza, J. W. Froehlich, D. E. Block, J. B. German et al., Consumption of Human Milk Oligosaccharides by Gut-Related Microbes, J Agric Food Chem. 12 mai, vol.58, issue.9, pp.5334-5374, 2010.

L. Huërou-luron, I. Blat, S. Boudry, and G. , Breast-v. formula-feeding: impacts on the digestive tract and immediate and long-term health effects, Nutr Res Rev. juin, vol.23, issue.1, pp.23-36, 2010.

R. González, E. S. Klaassens, E. Malinen, W. M. De-vos, and E. E. Vaughan, Differential Transcriptional Response of Bifidobacterium longum to Human Milk, Formula Milk, and Galactooligosaccharide, Appl Environ Microbiol. août, vol.74, issue.15, pp.4686-94, 2008.

D. A. Sela and D. A. Mills, Nursing our microbiota: molecular linkages between bifidobacteria and milk oligosaccharides, Trends Microbiol. juill, vol.18, issue.7, pp.298-307, 2010.

H. Sakurama, M. Kiyohara, J. Wada, Y. Honda, M. Yamaguchi et al., Lacto-Nbiosidase Encoded by a Novel Gene of Bifidobacterium longum Subspecies longum Shows Unique Substrate Specificity and Requires a Designated Chaperone for Its Active Expression, J Biol Chem. 30 août, vol.288, issue.35, pp.25194-206, 2013.

T. Matsuki, K. Yahagi, H. Mori, H. Matsumoto, T. Hara et al., A key genetic factor for fucosyllactose utilization affects infant gut microbiota development, Nature Communications. 24 juin, vol.7, p.11939, 2016.

Y. M. Sjögren, S. Tomicic, A. Lundberg, M. F. Böttcher, B. Björkstén et al., Influence of early gut microbiota on the maturation of childhood mucosal and systemic immune responses, Clinical & Experimental Allergy, vol.39, issue.12, pp.1842-51, 2009.

M. Chichlowski, G. De-lartigue, J. B. German, H. E. Raybould, and D. A. Mills, Bifidobacteria isolated from infants and cultured on human milk oligosaccharides affect intestinal epithelial function, J Pediatr Gastroenterol Nutr. sept, vol.55, issue.3, pp.321-328, 2012.

S. R. Iyengar and W. A. Walker, Immune factors in breast milk and the development of atopic disease, J Pediatr Gastroenterol Nutr. déc, vol.55, issue.6, pp.641-648, 2012.

C. J. Field, The immunological components of human milk and their effect on immune development in infants, J Nutr. janv, vol.135, issue.1, pp.1-4, 2005.

P. Palmeira and M. Carneiro-sampaio, Immunology of breast milk, Rev Assoc Med Bras, vol.62, issue.6, pp.584-93, 1992.

B. Levast, Z. Li, and J. Madrenas, The role of IL-10 in microbiome-associated immune modulation and disease tolerance, Cytokine. oct, vol.75, issue.2, pp.291-301, 2015.

P. Brandtzaeg, Mucosal immunity: integration between mother and the breast-fed infant, Vaccine. 28 juill, vol.21, issue.24, pp.3382-3390, 2003.

. Jeurink-p-v, J. Van-bergenhenegouwen, E. Jiménez, . Knippels-l-m.-j, L. Fernández et al., Human milk: a source of more life than we imagine, Beneficial Microbes. 27 déc, vol.4, issue.1, pp.17-30, 2012.

N. R. Klatt, N. T. Funderburg, and J. M. Brenchley, Microbial translocation, immune activation and HIV disease, Trends Microbiol. janv, vol.21, issue.1, pp.6-13, 2013.

M. C. Collado, S. Delgado, A. Maldonado, and J. M. Rodríguez, Assessment of the bacterial diversity of breast milk of healthy women by quantitative real-time PCR, Lett Appl Microbiol. mai, vol.48, issue.5, pp.523-531, 2009.

J. W. Sahl, M. N. Matalka, and D. A. Rasko, Phylomark, a tool to identify conserved phylogenetic markers from whole-genome alignments, Appl Environ Microbiol. juill, vol.78, issue.14, pp.4884-92, 2012.

L. Fernández, S. Langa, V. Martín, A. Maldonado, E. Jiménez et al., The human milk microbiota: origin and potential roles in health and disease, Pharmacol Res. mars, vol.69, issue.1, pp.1-10, 2013.

R. Cabrera-rubio, M. C. Collado, K. Laitinen, S. Salminen, E. Isolauri et al., The human milk microbiome changes over lactation and is shaped by maternal weight and mode of delivery, Am J Clin Nutr. 1 sept, vol.96, issue.3, pp.544-51, 2012.

V. Martín, A. Maldonado-barragán, L. Moles, M. Rodriguez-baños, R. D. Campo et al., Sharing of bacterial strains between breast milk and infant feces, J Hum Lact. févr, vol.28, issue.1, pp.36-44, 2012.

R. Ducluzeau, Development, equilibrium and role of microbial flora in the newborn

, Ann Pediatr (Paris). janv, vol.40, issue.1, pp.13-22, 1993.

K. Orrhage and C. E. Nord, Factors controlling the bacterial colonization of the intestine in breastfed infants, Acta Paediatr Suppl. août, vol.88, issue.430, pp.47-57, 1999.

E. Bezirtzoglou, A. Tsiotsias, and G. W. Welling, Microbiota profile in feces of breast-and formula-fed newborns by using fluorescence in situ hybridization (FISH), Anaerobe. déc, vol.17, issue.6, pp.478-82, 2011.

M. Fallani, D. Young, J. Scott, E. Norin, S. Amarri et al., Intestinal microbiota of 6-week-old infants across Europe: geographic influence beyond delivery mode, breastfeeding, and antibiotics, J Pediatr Gastroenterol Nutr. juill, vol.51, issue.1, pp.77-84, 2010.

M. Rinne, M. Kalliomaki, H. Arvilommi, S. Salminen, and E. Isolauri, Effect of probiotics and breastfeeding on the bifidobacterium and lactobacillus/enterococcus microbiota and humoral immune responses, J Pediatr. août, vol.147, issue.2, pp.186-91, 2005.

B. Lundequist, C. E. Nord, and J. Winberg, The composition of the faecal microflora in breastfed and bottle fed infants from birth to eight weeks, Acta Paediatr Scand. janv, vol.74, issue.1, pp.45-51, 1985.

P. L. Stark and A. Lee, Clostridia isolated from the feces of infants during the first year of life, J Pediatr. mars, vol.100, issue.3, pp.362-367, 1982.

A. Marcobal, M. Barboza, E. D. Sonnenburg, N. Pudlo, E. C. Martens et al., Bacteroides in the infant gut consume milk oligosaccharides via mucus-utilization pathways, Cell Host Microbe, vol.10, issue.5, pp.507-521, 2011.

F. Campeotto, A. Waligora-dupriet, F. Doucet-populaire, N. Kalach, C. Dupont et al.,

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. 1 juin, vol.5, issue.6, pp.644-53, 1998.

G. V. Coppa, S. Bruni, L. Morelli, S. Soldi, and O. Gabrielli, The first prebiotics in humans: human milk oligosaccharides, J Clin Gastroenterol. juill, vol.38, issue.6, pp.80-83, 2004.

G. Veereman-wauters, S. Staelens, H. Van-de-broek, K. Plaskie, F. Wesling et al., Physiological and bifidogenic effects of prebiotic supplements in infant formulae, J Pediatr Gastroenterol Nutr. juin, vol.52, issue.6, pp.763-71, 2011.

K. E. Gregory, B. S. Samuel, P. Houghteling, G. Shan, F. M. Ausubel et al., Influence of maternal breast milk ingestion on acquisition of the intestinal microbiome in preterm infants, Microbiome, vol.30, issue.1, p.68, 2016.

Y. Vallès, A. Artacho, A. Pascual-garcía, M. L. Ferrús, M. J. Gosalbes et al., Microbial succession in the gut: directional trends of taxonomic and functional change in a birth cohort of Spanish infants, PLoS Genet. juin, vol.10, issue.6, p.1004406, 2014.

M. Fallani, S. Amarri, A. Uusijarvi, R. Adam, S. Khanna et al., Determinants of the human infant intestinal microbiota after the introduction of first complementary foods in infant samples from five European centres, Microbiology (Reading, Engl). mai, vol.157, pp.1385-92, 2011.

P. A. Vaishampayan, J. V. Kuehl, J. L. Froula, J. L. Morgan, H. Ochman et al., Comparative Metagenomics and Population Dynamics of the Gut Microbiota in Mother and Infant, Genome Biol Evol. 1 janv, vol.2, pp.53-66, 2010.

L. Dethlefsen, S. Huse, M. L. Sogin, and D. A. Relman, The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing, PLoS Biol, vol.6, issue.11, p.280, 2008.

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 Immunol Med Microbiol. juin, vol.56, issue.1, pp.80-87, 2009.

F. Fouhy, C. M. Guinane, S. Hussey, R. Wall, C. A. Ryan et al., High-throughput sequencing reveals the incomplete, short-term recovery of infant gut microbiota following parenteral antibiotic treatment with ampicillin and gentamicin, Antimicrob Agents Chemother, vol.56, issue.11, pp.5811-5831, 2012.

C. Greenwood, A. L. Morrow, A. J. Lagomarcino, M. Altaye, D. H. Taft et al., Early empiric antibiotic use in preterm infants is associated with lower bacterial diversity and higher relative abundance of Enterobacter, J Pediatr. juill, vol.165, issue.1, pp.23-32, 2014.

A. M. Moore, S. Ahmadi, S. Patel, M. K. Gibson, B. Wang et al., Gut resistome development in healthy twin pairs in the first year of life, Microbiome, vol.3, p.27, 2015.

R. Bennet, M. Eriksson, C. E. Nord, and R. Zetterström, Fecal bacterial microflora of newborn infants during intensive care management and treatment with five antibiotic regimens, Pediatr Infect Dis. oct, vol.5, issue.5, pp.533-542, 1986.

A. C. White, R. L. Atmar, J. Wilson, T. R. Cate, C. E. Stager et al., Effects of requiring prior authorization for selected antimicrobials: expenditures, susceptibilities, and clinical outcomes, Clin Infect Dis. août, vol.25, issue.2, pp.230-239, 1997.

B. J. Stoll, N. Hansen, A. A. Fanaroff, L. L. Wright, W. A. Carlo et al., Changes in pathogens causing early-onset sepsis in very-low-birth-weight infants, N Engl J Med. 25 juill, vol.347, issue.4, pp.240-247, 2002.

F. Jauréguy, M. Carton, P. Panel, P. Foucaud, M. Butel et al., Effects of intrapartum penicillin prophylaxis on intestinal bacterial colonization in infants, J Clin Microbiol, vol.42, issue.11, pp.5184-5192, 2004.

L. A. Hanson, M. Korotkova, and E. Telemo, Breast-feeding, infant formulas, and the immune system, Ann Allergy Asthma Immunol. juin, vol.90, issue.6, pp.59-63, 2003.

A. Langdon, N. Crook, and G. Dantas, The effects of antibiotics on the microbiome throughout development and alternative approaches for therapeutic modulation, Genome Medicine. déc, vol.8, issue.1, p.39, 2016.

J. Nakayama, T. Kobayashi, S. Tanaka, Y. Korenori, A. Tateyama et al., Aberrant structures of fecal bacterial community in allergic infants profiled by 16S rRNA gene pyrosequencing, FEMS Immunol Med Microbiol. 1 déc, vol.63, issue.3, pp.397-406, 2011.

T. Vatanen, A. D. Kostic, E. Hennezel, H. Siljander, E. A. Franzosa et al., Variation in Microbiome LPS Immunogenicity Contributes to, Autoimmunity in Humans. Cell. 5 mai, vol.165, issue.4, pp.842-53, 2016.

E. G. Zoetendal, A. D. Akkermans, D. Vos, and W. M. , Temperature gradient gel electrophoresis analysis of 16S rRNA from human fecal samples reveals stable and host-specific communities of active bacteria, Appl Environ Microbiol, vol.64, issue.10, pp.3854-3863, 1998.

, Science for health. Evolution du microbiote avec l'âge

M. J. Claesson, S. Cusack, O. O'sullivan, R. Greene-diniz, H. De-weerd et al., Composition, variability, and temporal stability of the intestinal microbiota of the elderly, Proc Natl Acad Sci, vol.108, issue.1, pp.4586-91, 2011.

E. J. Woodmansey, Intestinal bacteria and ageing, J Appl Microbiol. mai, vol.102, issue.5, pp.1178-86, 2007.

E. J. Woodmansey, M. Mcmurdo, G. T. Macfarlane, and S. Macfarlane, Comparison of compositions and metabolic activities of fecal microbiotas in young adults and in antibiotic-treated and non-antibiotic-treated elderly subjects, Appl Environ Microbiol, vol.70, issue.10, pp.6113-6135, 2004.

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

J. K. Goodrich, J. L. Waters, A. C. Poole, J. L. Sutter, O. Koren et al., Human genetics shape the gut microbiome, Cell, vol.159, issue.4, pp.789-99, 2014.

J. K. Goodrich, E. R. Davenport, M. Beaumont, M. A. Jackson, R. Knight et al., Genetic Determinants of the Gut Microbiome in UK Twins, Cell Host & Microbe. 11 mai, vol.19, issue.5, pp.731-774, 2016.

L. Maier, M. Pruteanu, M. Kuhn, G. Zeller, A. Telzerow et al., Extensive impact of non-antibiotic drugs on human gut bacteria, Nature, vol.29, issue.7698, pp.623-631, 2018.

L. Biedermann, J. Zeitz, J. Mwinyi, E. Sutter-minder, A. Rehman et al., Smoking cessation induces profound changes in the composition of the intestinal microbiota in humans, PLoS ONE, vol.8, issue.3, p.59260, 2013.

H. Jiang, Z. Ling, Y. Zhang, H. Mao, Z. Ma et al., Altered fecal microbiota composition in patients with major depressive disorder, Brain Behav Immun. août, vol.48, pp.186-94, 2015.

M. Lyte, L. Vulchanova, and D. R. Brown, Stress at the intestinal surface: catecholamines and mucosa-bacteria interactions, Cell Tissue Res. janv, vol.343, issue.1, pp.23-32, 2011.

M. G. Dominguez-bello, F. Godoy-vitorino, R. Knight, and M. J. Blaser, Role of the microbiome in human development, Gut, vol.68, issue.6, pp.1108-1122, 2019.

S. Chen, C. Tsai, Y. Lee, C. Lin, K. Huang et al., Intestinal microbiome in children with severe and complicated acute viral gastroenteritis, Sci Rep, vol.11, p.46130, 2017.

F. E. Garrett-bakelman, M. Darshi, S. J. Green, R. C. Gur, L. Lin et al., The NASA Twins Study: A multidimensional analysis of a year-long human spaceflight, Science, vol.12, issue.6436, 2019.

C. De-filippo, D. Cavalieri, D. Paola, M. Ramazzotti, M. Poullet et al., Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa, Proc Natl Acad Sci, vol.107, issue.33, pp.14691-14697, 2010.

A. Lin, E. M. Bik, E. K. Costello, L. Dethlefsen, R. Haque et al., Distinct Distal Gut Microbiome Diversity and Composition in Healthy Children from Bangladesh and the United States, PLOS ONE. 22 janv, vol.8, issue.1, p.53838, 2013.

J. Nakayama, K. Watanabe, J. Jiang, K. Matsuda, S. Chao et al., Diversity in gut bacterial community of school-age children in Asia, Scientific Reports. 23 févr, vol.5, p.8397, 2015.

S. Ruengsomwong, O. La-ongkham, J. Jiang, B. Wannissorn, J. Nakayama et al., Microbial Community of Healthy Thai Vegetarians and Non-Vegetarians, Their Core Gut Microbiota, and Pathogen Risk, Journal of Microbiology and Biotechnology, vol.26, issue.10, pp.1723-1758, 2016.

G. D. Wu, J. Chen, C. Hoffmann, K. Bittinger, Y. Chen et al., Linking longterm dietary patterns with gut microbial enterotypes, Science, vol.334, issue.6052, pp.105-113, 2011.

J. Nakayama, A. Yamamoto, L. A. Palermo-conde, K. Higashi, K. Sonomoto et al., Impact of Westernized Diet on, Gut Microbiota in Children on Leyte Island. Front Microbiol, vol.8, p.197, 2017.

M. J. Claesson, I. B. Jeffery, S. Conde, S. E. Power, E. M. O'connor et al., Gut microbiota composition correlates with diet and health in the elderly, Nature. 9 août, vol.488, issue.7410, pp.178-84, 2012.

C. T. Kåhrström, N. Pariente, and U. Weiss, Intestinal microbiota in health and disease, Nature, vol.07, issue.7610, p.47, 2016.

K. Forslund, F. Hildebrand, T. Nielsen, G. Falony, L. Chatelier et al., Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota, Nature. 10 déc, vol.528, issue.7581, pp.262-268, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01535324

F. Imhann, M. J. Bonder, V. Vila, A. Fu, J. Mujagic et al., Proton pump inhibitors affect the gut microbiome, Gut. mai, vol.65, issue.5, pp.740-748, 2016.

M. A. Jackson, J. K. Goodrich, M. Maxan, D. E. Freedberg, J. A. Abrams et al., Proton pump inhibitors alter the composition of the gut microbiota, Gut. mai, vol.65, issue.5, pp.749-56, 2016.

K. R. Reveles, C. N. Ryan, L. Chan, R. A. Cosimi, and W. L. Haynes, Proton pump inhibitor use associated with changes in gut microbiota composition, Gut, vol.67, issue.7, pp.1369-70, 2018.

. Rogers-m-a.-m and D. M. Aronoff, The influence of non-steroidal anti-inflammatory drugs on the gut microbiome, Clin Microbiol Infect. févr, vol.22, issue.2, pp.178-179, 2016.

S. A. Flowers, S. J. Evans, K. M. Ward, M. G. Mcinnis, and V. L. Ellingrod, Interaction Between Atypical Antipsychotics and the Gut Microbiome in a Bipolar Disease Cohort, Pharmacotherapy, vol.37, issue.3, pp.261-268, 2017.

L. Maier, M. Pruteanu, M. Kuhn, G. Zeller, A. Telzerow et al., Extensive impact of non-antibiotic drugs on human gut bacteria, Nature. 29 mars, vol.555, issue.7698, pp.623-631, 2018.

C. F. Maurice, H. J. Haiser, and P. J. Turnbaugh, Xenobiotics shape the physiology and gene expression of the active human gut microbiome, Cell. 17 janv, vol.152, issue.1-2, pp.39-50, 2013.

X. Lescure, Gestion des effets secondaires des antibiotiques, p.87

C. Högenauer, H. F. Hammer, G. J. Krejs, and E. C. Reisinger, Mechanisms and management of antibiotic-associated diarrhea, Clin Infect Dis, vol.27, issue.4, pp.702-712, 1998.

A. Gabaron-kerleguer, C. P. Soler, C. Désidéri-vaillant, E. Garrabé, and J. D. Cavallo, Diarrhées post-antibiothérapie, Médecine et Maladies Infectieuses. 1 janv, vol.31, issue.11, pp.650-655, 2001.

T. L. Peeters, Erythromycin and other macrolides as prokinetic agents, Gastroenterology. déc, vol.105, issue.6, pp.1886-99, 1993.

F. Caron, P. Ducrotte, E. Lerebours, C. R. Humbert, G. Denis et al., Effects of amoxicillinclavulanate combination on the motility of the small intestine in human beings, Antimicrob Agents Chemother. juin, vol.35, issue.6, pp.1085-1093, 1991.

W. O. Dobbins, B. A. Herrero, and C. M. Mansbach, Morphologic alterations associated with neomycin induced malabsorption, Am J Med Sci. janv, vol.255, pp.63-77, 1968.

G. Kaltenbach and D. Heitz,

, Rev Med Interne. janv, vol.25, issue.1, pp.46-53, 2004.

E. Masson, . Colite-postantibiotique, and . Em-consulte,

D. F. Newton, S. Macfarlane, and G. T. Macfarlane, Effects of antibiotics on bacterial species composition and metabolic activities in chemostats containing defined populations of human gut microorganisms, Antimicrob Agents Chemother. mai, vol.57, issue.5, pp.2016-2041, 2013.

C. Jernberg, S. Löfmark, C. Edlund, and J. K. Jansson, Long-term impacts of antibiotic exposure on the human intestinal microbiota, Microbiology, vol.156, issue.11, pp.3216-3239, 2010.

G. E. Bignardi, Risk factors for Clostridium difficile infection, J Hosp Infect. sept, vol.40, issue.1, pp.1-15, 1998.

L. V. Mcfarland, C. M. Surawicz, and W. E. Stamm, Risk factors for Clostridium difficile carriage and C. difficile-associated diarrhea in a cohort of hospitalized patients, J Infect Dis. sept, vol.162, issue.3, pp.678-84, 1990.

F. Barbut and J. C. Petit, Epidemiology of Clostridium difficile-associated infections, Clin Microbiol Infect. août, vol.7, issue.8, pp.405-415, 2001.

M. F. De-la-cochetière, T. Durand, P. Lepage, A. Bourreille, J. P. Galmiche et al., Resilience of the dominant human fecal microbiota upon short-course antibiotic challenge, J Clin Microbiol, vol.43, issue.11, pp.5588-92, 2005.

J. M. Lankelma, M. Nieuwdorp, W. M. De-vos, and W. J. Wiersinga, The gut microbiota in internal medicine: implications for health and disease, Neth J Med. févr, vol.73, issue.2, pp.61-69, 2015.

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 USA. 15 mars, vol.108, pp.4554-61, 2011.

H. E. Jakobsson, C. Jernberg, A. F. Andersson, M. Sjölund-karlsson, J. K. Jansson et al., Short-term antibiotic treatment has differing long-term impacts on the human throat and gut microbiome, PLoS ONE. 24 mars, vol.5, issue.3, p.9836, 2010.

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

M. Mcfall-ngai, M. G. Hadfield, T. Bosch, H. V. Carey, T. Domazet-lo?o et al., Animals in a bacterial world, a new imperative for the life sciences, Proc Natl Acad Sci, vol.110, issue.9, pp.3229-3265, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00972300

Y. Belkaid and T. W. Hand, Role of the microbiota in immunity and inflammation, Cell. 27 mars, vol.157, issue.1, pp.121-162, 2014.

A. Bernalier-donadille, Fermentative metabolism by the human gut microbiota, Gastroentérologie Clinique et Biologique. 1 sept, vol.34, pp.16-22, 2010.

C. Chang and H. Lin, Dysbiosis in gastrointestinal disorders, Best Pract Res Clin Gastroenterol. févr, vol.30, issue.1, pp.3-15, 2016.

B. O. Schroeder and F. Bäckhed, Signals from the gut microbiota to distant organs in physiology and disease, Nat Med, vol.22, issue.10, pp.1079-89, 2016.

M. Lyte, Microbial Endocrinology: The Microbiota-Gut-Brain Axis in Health and Disease, Cryan JF, éditeur, pp.3-24, 2014.

V. Gaboriau-routhiau and N. Cerf-bensussan, Gut microbiota and development of the immune system, Med Sci, vol.32, issue.11, pp.961-968, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01606693

J. Natividad and E. F. Verdu, Modulation of intestinal barrier by intestinal microbiota: pathological and therapeutic implications, Pharmacol Res. mars, vol.69, issue.1, pp.42-51, 2013.

K. Smith, K. D. Mccoy, and A. J. Macpherson, Use of axenic animals in studying the adaptation of mammals to their commensal intestinal microbiota, Semin Immunol. avr, vol.19, issue.2, pp.59-69, 2007.

P. A. Swanson, A. Kumar, S. Samarin, M. Vijay-kumar, K. Kundu et al., Enteric commensal bacteria potentiate epithelial restitution via reactive oxygen species-mediated inactivation of focal adhesion kinase phosphatases, Proc Natl Acad Sci USA. 24 mai, vol.108, issue.21, pp.8803-8811, 2011.

J. Petersson, O. Schreiber, G. C. Hansson, S. J. Gendler, A. Velcich et al., Importance and regulation of the colonic mucus barrier in a mouse model of colitis, Am J Physiol Gastrointest Liver Physiol. févr, vol.300, issue.2, pp.327-333, 2011.

L. Wrzosek, S. Miquel, M. Noordine, S. Bouet, J. Chevalier-curt et al., Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent, BMC Biol. 21 mai, vol.11, p.61, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01001072

L. V. Hooper, Bacterial contributions to mammalian gut development, Trends Microbiol. mars, vol.12, issue.3, pp.129-163, 2004.

A. J. Bäumler and V. Sperandio, Interactions between the microbiota and pathogenic bacteria in the gut, Nature, vol.07, issue.7610, pp.85-93, 2016.

S. Fukuda, H. Toh, K. Hase, K. Oshima, Y. Nakanishi et al., Bifidobacteria can protect from enteropathogenic infection through production of acetate, Nature. 27 janv, vol.469, issue.7331, pp.543-550, 2011.

L. V. Hooper and A. J. Macpherson, Immune adaptations that maintain homeostasis with the intestinal microbiota, Nat Rev Immunol. mars, vol.10, issue.3, pp.159-69, 2010.

A. Mathias, B. Pais, L. Favre, J. Benyacoub, and B. Corthésy, Role of secretory IgA in the mucosal sensing of commensal bacteria, Gut Microbes, vol.5, issue.6, pp.688-95, 2014.

E. W. Rogier, A. L. Frantz, M. Bruno, and C. S. Kaetzel, Secretory IgA is Concentrated in the Outer Layer of Colonic Mucus along with Gut Bacteria, Pathogens. 29 avr, vol.3, issue.2, pp.390-403, 2014.

M. P. Francino, Early Development of the Gut Microbiota and Immune Health, Pathogens. sept, vol.3, issue.3, pp.769-90, 2014.

Q. N. Nguyen, J. E. Himes, D. R. Martinez, and S. R. Permar, The Impact of the Gut Microbiota on Humoral Immunity to Pathogens and Vaccination in Early Infancy, PLOS Pathogens. 22 déc, vol.12, issue.12, p.1005997, 2016.

A. Hevia, S. Delgado, B. Sánchez, and A. Margolles, Molecular Players Involved in the Interaction Between Beneficial Bacteria and the Immune System, Front Microbiol, vol.6, p.1285, 2015.

P. Gérard and A. Bernalier-donadille, Les fonctions majeures du microbiote intestinal, Cahiers de Nutrition et de Diététique. 1 avr, vol.42, pp.28-36, 2007.

S. U. Christl, P. R. Murgatroyd, G. R. Gibson, and J. H. Cummings, Production, metabolism, and excretion of hydrogen in the large intestine, Gastroenterology. 1 avr, vol.102, issue.4, pp.1269-77, 1992.

A. H. Lichtenstein, Intestinal cholesterol metabolism, Ann Med. févr, vol.22, issue.1, pp.49-52, 1990.

J. M. Ridlon, D. Kang, and P. B. Hylemon, Bile salt biotransformations by human intestinal bacteria, J Lipid Res. 2 janv, vol.47, issue.2, pp.241-59, 2006.

I. A. Macdonald, V. D. Bokkenheuser, J. Winter, A. M. Mclernon, and E. H. Mosbach, Degradation of steroids in the human gut, J Lipid Res. 6 janv, vol.24, issue.6, pp.675-700, 1983.

P. J. Turnbaugh, R. E. Ley, M. A. Mahowald, V. Magrini, E. R. Mardis et al., An obesityassociated gut microbiome with increased capacity for energy harvest, Nature. 21 déc, vol.444, issue.7122, pp.1027-1058, 2006.

J. H. Cummings and G. T. Macfarlane, The control and consequences of bacterial fermentation in the human colon, Journal of Applied Bacteriology, vol.70, issue.6, pp.443-59, 1991.

H. N. Englyst, S. Hay, and G. T. Macfarlane, Polysaccharide breakdown by mixed populations of human faecal bacteria, FEMS Microbiology Letters. 1 juin, vol.45, issue.3, pp.163-71, 1987.

T. L. Miller and M. J. Wolin, Fermentations by saccharolytic intestinal bacteria, Am J Clin Nutr. janv, vol.32, issue.1, pp.164-72, 1979.

N. Reichardt, S. H. Duncan, P. Young, A. Belenguer, M. Leitch et al., Phylogenetic distribution of three pathways for propionate production within the human gut microbiota, ISME J. juin, vol.8, issue.6, pp.1323-1358, 2014.

S. E. Pryde, S. H. Duncan, G. L. Hold, C. S. Stewart, and H. J. Flint, The microbiology of butyrate formation in the human colon, FEMS Microbiol Lett. 17 déc, vol.217, issue.2, pp.133-142, 2002.

G. Den-besten, K. Van-eunen, A. K. Groen, K. Venema, D. Reijngoud et al., The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism, J Lipid Res. sept, vol.54, issue.9, pp.2325-2365, 2013.

M. S. Ahmad, S. Krishnan, B. S. Ramakrishna, M. Mathan, A. B. Pulimood et al., Butyrate and glucose metabolism by colonocytes in experimental colitis in mice, Gut. avr, vol.46, issue.4, pp.493-502, 2000.

B. S. Ramakrishna and V. I. Mathan, Colonic dysfunction in acute diarrhoea: the role of luminal short chain fatty acids, Gut. sept, vol.34, issue.9, pp.1215-1223, 1993.

Y. Sun and M. O'riordan, Regulation of bacterial pathogenesis by intestinal short-chain Fatty acids, Adv Appl Microbiol, vol.85, pp.93-118, 2013.

L. B. Bindels, P. Porporato, E. M. Dewulf, J. Verrax, A. M. Neyrinck et al., Gut microbiota-derived propionate reduces cancer cell proliferation in the liver, Br J Cancer, vol.107, issue.8, pp.1337-1381, 2012.

R. Corrêa-oliveira, J. L. Fachi, A. Vieira, F. T. Sato, and M. Vinolo, Regulation of immune cell function by short-chain fatty acids, Clin Transl Immunology. 22 avr, vol.5, issue.4, p.73, 2016.

S. F. Phillips, J. H. Pemberton, and R. Shorter, The large intestine Physiology, pathophysiology and disease, Acta Endosc. 1 mars, vol.21, issue.2, pp.XVIII-XVIII, 1991.

G. T. Macfarlane, Metabolic Activities of the Normal Colonic Flora. In: Gibson SAW, éditeur. Human Health: The Contribution of Microorganisms, pp.17-52, 1994.

N. Koppel, V. M. Rekdal, and E. P. Balskus, Chemical transformation of xenobiotics by the human gut microbiota, Science, vol.356, issue.6344, 2018.

H. Li, J. He, and W. Jia, The influence of gut microbiota on drug metabolism and toxicity, Expert Opin Drug Metab Toxicol, vol.12, issue.1, pp.31-40, 2016.

J. G. Leblanc, J. E. Laiño, M. J. Del-valle, V. Vannini, D. Van-sinderen et al., Bgroup vitamin production by lactic acid bacteria--current knowledge and potential applications, J Appl Microbiol. déc, vol.111, issue.6, pp.1297-309, 2011.

J. G. Leblanc, C. Milani, G. S. De-giori, F. Sesma, D. Van-sinderen et al., Bacteria as vitamin suppliers to their host: a gut microbiota perspective, Curr Opin Biotechnol. avr, vol.24, issue.2, pp.160-168, 2013.

H. M. Said and Z. M. Mohammed, Intestinal absorption of water-soluble vitamins: an update, Curr Opin Gastroenterol. mars, vol.22, issue.2, pp.140-146, 2006.

T. Ichihashi, Y. Takagishi, K. Uchida, and H. Yamada, Colonic absorption of menaquinone-4 and menaquinone-9 in rats, J Nutr. mars, vol.122, issue.3, pp.506-518, 1992.

J. H. Martens, H. Barg, M. J. Warren, and D. Jahn, Microbial production of vitamin B12, Appl Microbiol Biotechnol. mars, vol.58, issue.3, pp.275-85, 2002.

A. Pompei, L. Cordisco, A. Amaretti, S. Zanoni, D. Matteuzzi et al., Folate production by bifidobacteria as a potential probiotic property, Appl Environ Microbiol. janv, vol.73, issue.1, pp.179-85, 2007.

M. J. Hill, Intestinal flora and endogenous vitamin synthesis, Eur J Cancer Prev. mars, vol.6, issue.1, pp.43-45, 1997.

P. Lepage, The human gut microbiota: Interactions with the host and dysfunctions, Rev Mal Respir. déc, vol.34, issue.10, pp.1085-90, 2017.

H. Sokol, P. Seksik, J. P. Furet, O. Firmesse, I. Nion-larmurier et al., Low counts of Faecalibacterium prausnitzii in colitis microbiota, Inflamm Bowel Dis. août, vol.15, issue.8, pp.1183-1192, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00657435

J. Kabeerdoss, V. Sankaran, S. Pugazhendhi, and B. S. Ramakrishna, Clostridium leptum group bacteria abundance and diversity in the fecal microbiota of patients with inflammatory bowel disease: a case-control study in India, BMC Gastroenterol. 26 janv, vol.13, p.20, 2013.

J. R. Marchesi, B. E. Dutilh, N. Hall, W. Peters, R. Roelofs et al., Towards the Human Colorectal Cancer Microbiome, PLOS ONE. 24 mai, vol.6, issue.5, p.20447, 2011.

M. Castellarin, R. L. Warren, J. D. Freeman, L. Dreolini, M. Krzywinski et al., Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma, Genome Res. févr, vol.22, issue.2, pp.299-306, 2012.

L. Flanagan, J. Schmid, M. Ebert, P. Soucek, T. Kunicka et al., Fusobacterium nucleatum associates with stages of colorectal neoplasia development, colorectal cancer and disease outcome, Eur J Clin Microbiol Infect Dis. août, vol.33, issue.8, pp.1381-90, 2014.

J. G. Bartlett, Clostridium difficile Infection, Infect Dis Clin North Am, vol.31, issue.3, pp.489-95, 2017.

C. Vael, V. Nelen, S. L. Verhulst, H. Goossens, and K. N. Desager, Early intestinal Bacteroides fragilis colonisation and development of asthma, BMC Pulm Med. 26 sept, vol.8, p.19, 2008.

J. Penders, C. Thijs, P. A. Van-den-brandt, I. Kummeling, B. Snijders et al., Gut microbiota composition and development of atopic manifestations in infancy: the KOALA Birth Cohort Study, Gut. mai, vol.56, issue.5, pp.661-668, 2007.

R. E. Ley, P. J. Turnbaugh, S. Klein, and J. I. Gordon, Human gut microbes associated with obesity, Nature. déc, vol.444, issue.7122, p.1022, 2006.

J. Qin, Y. Li, Z. Cai, S. Li, J. Zhu et al., A metagenome-wide association study of gut microbiota in type 2 diabetes, Nature, vol.490, issue.7418, pp.55-60, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01204262

F. H. Karlsson, V. Tremaroli, I. Nookaew, G. Bergström, C. J. Behre et al., Gut metagenome in European women with normal, impaired and diabetic glucose control, Nature. 6 juin, vol.498, issue.7452, pp.99-103, 2013.

J. Sato, A. Kanazawa, F. Ikeda, T. Yoshihara, H. Goto et al., Gut dysbiosis and detection of « live gut bacteria » in blood of Japanese patients with type 2 diabetes, Diabetes Care. août, vol.37, issue.8, pp.2343-50, 2014.

F. Scheperjans, V. Aho, P. Pereira, K. Koskinen, L. Paulin et al., Gut microbiota are related to Parkinson's disease and clinical phenotype, Mov Disord. mars, vol.30, issue.3, pp.350-358, 2015.

L. Wang, C. T. Christophersen, M. J. Sorich, J. P. Gerber, M. T. Angley et al., Low Relative Abundances of the Mucolytic Bacterium Akkermansia muciniphila and Bifidobacterium spp. in Feces of Children with Autism, Appl Environ Microbiol. 15 sept, vol.77, issue.18, pp.6718-6739, 2011.

B. L. Williams, M. Hornig, T. Parekh, and W. I. Lipkin, Application of novel PCR-based methods for detection, quantitation, and phylogenetic characterization of Sutterella species in intestinal biopsy samples from children with autism and gastrointestinal disturbances, MBio, vol.3, issue.1, 2012.

W. Tang, Z. Wang, B. S. Levison, R. A. Koeth, E. B. Britt et al., Intestinal Microbial Metabolism of Phosphatidylcholine and Cardiovascular Risk, New England Journal of Medicine. 25 avr, vol.368, issue.17, pp.1575-84, 2013.

A. Cotillard, S. P. Kennedy, L. C. Kong, E. Prifti, N. Pons et al., Dietary intervention impact on gut microbial gene richness, Nature. 29 août, vol.500, issue.7464, pp.585-593, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01001543

. Vidal, Greffe de flore intestinale : les recommandations de l'ANSM pour améliorer la sécurité

A. Dinh, F. Bouchand, and A. M. Le, Current treatment and epidemiology of Clostridium difficile infections, Rev Med Interne. sept, vol.36, issue.9, pp.596-602, 2015.

N. G. Rossen, J. K. Macdonald, E. M. Vries, D. De, G. R. Haens et al., Fecal microbiota transplantation as novel therapy in gastroenterology: A systematic review, World Journal of Gastroenterology. 7 mai, vol.21, issue.17, pp.5359-71, 2015.

J. Boclé, ANSES -Effets des probiotiques et prébiotiques sur la flore et l'immunité de l'Homme adulte, vol.128, 2005.

P. Markowiak and K. ?li?ewska, Effects of Probiotics, Prebiotics, and Synbiotics on Human Health, Nutrients. 15 sept, vol.9, issue.9, 2017.

S. Bengmark, Bioecological control of inflammatory bowel disease, Clin Nutr. avr, vol.26, issue.2, pp.169-81, 2007.

M. S. Geier, R. N. Butler, and G. S. Howarth, Inflammatory bowel disease: current insights into pathogenesis and new therapeutic options; probiotics, prebiotics and synbiotics, Int J Food Microbiol. 1 avr, vol.115, issue.1, pp.1-11, 2007.

L. V. Mcfarland, Meta-analysis of probiotics for the prevention of antibiotic associated diarrhea and the treatment of Clostridium difficile disease, Am J Gastroenterol. avr, vol.101, issue.4, pp.812-834, 2006.

P. A. Vandenbergh, Lactic acid bacteria, their metabolic products and interference with microbial growth, FEMS Microbiol Rev. 1 sept, vol.12, issue.1-3, pp.221-258, 1993.

J. F. Guillot, Probiotic feed additives, J Vet Pharmacol Ther, 2003.

R. L. Brandão, I. M. Castro, E. A. Bambirra, S. C. Amaral, L. G. Fietto et al., Intracellular signal triggered by cholera toxin in Saccharomyces boulardii and Saccharomyces cerevisiae, Appl Environ Microbiol. févr, vol.64, issue.2, pp.564-572, 1998.

E. Isolauri, Y. Sütas, P. Kankaanpää, H. Arvilommi, and S. Salminen, Probiotics: effects on immunity, Am J Clin Nutr, vol.73, issue.2, pp.444-450, 2001.

A. T. Borchers, C. Selmi, F. J. Meyers, C. L. Keen, and M. E. Gershwin, Probiotics and immunity, J Gastroenterol, vol.44, issue.1, pp.26-46, 2009.

S. Doron and D. R. Snydman, Risk and safety of probiotics, Clin Infect Dis. 15 mai, vol.60, issue.2, pp.129-134, 2015.

R. J. Boyle, R. M. Robins-browne, and M. Tang, Probiotic use in clinical practice: what are the risks?, Am J Clin Nutr. juin, vol.83, issue.6, pp.1446-1453, 2006.

G. R. Gibson and M. B. Roberfroid, Dietary Modulation of the Human Colonic Microbiota: Introducing the Concept of Prebiotics, J Nutr. 1 juin, vol.125, issue.6, pp.1401-1413, 1995.

J. Schrezenmeir and M. De-vrese, Probiotics, prebiotics, and synbiotics-approaching a definition, Am J Clin Nutr, vol.73, issue.2, pp.361-364, 2001.

A. Foxx-orenstein and W. Chey, Manipulation of the Gut Microbiota as a Novel Treatment Strategy for Gastrointestinal Disorders, The American Journal of Gastroenterology Supplements. 1 juill, vol.1, pp.41-47, 2012.

A. A. Aachary and S. G. Prapulla, Xylooligosaccharides (XOS) as an Emerging Prebiotic: Microbial Synthesis, Utilization, Structural Characterization, Bioactive Properties, and Applications, Comprehensive Reviews in Food Science and Food Safety, vol.10, issue.1, pp.2-16, 2011.

D. Ríos-covián, P. Ruas-madiedo, A. Margolles, M. Gueimonde, C. G. De-los-reyes-gavilán et al., Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health, Front Microbiol, vol.7, p.185, 2016.

Y. Tsai, T. Lin, C. Chang, T. Wu, W. Lai et al., Probiotics, prebiotics and amelioration of diseases, J Biomed Sci, vol.4, issue.1, p.3, 2019.

G. R. Gibson, H. M. Probert, J. V. Loo, R. A. Rastall, and M. B. Roberfroid, Dietary modulation of the human colonic microbiota: updating the concept of prebiotics, Nutr Res Rev. déc, vol.17, issue.2, pp.259-75, 2004.

A. J. Waldman and E. P. Balskus, The Human Microbiota, Infectious Disease, and Global Health: Challenges and Opportunities, ACS Infect Dis, vol.12, issue.1, pp.14-26, 2018.

K. R. Pandey, S. R. Naik, and B. V. Vakil, Probiotics, prebiotics and synbiotics-a review, J Food Sci Technol. déc, vol.52, issue.12, pp.7577-87, 2015.

J. Ma, Q. Zhou, and H. Li, Gut Microbiota and Nonalcoholic Fatty Liver Disease: Insights on Mechanisms and Therapy, Nutrients, vol.9, issue.10, 2017.

I. Figueroa-gonzález, G. Quijano, G. Ramírez, and A. Cruz-guerrero, Probiotics and prebiotics -perspectives and challenges, J Sci Food Agric. juin, vol.91, issue.8, pp.1341-1349, 2011.

P. Panigrahi, S. Parida, L. Pradhan, S. S. Mohapatra, P. R. Misra et al., Long-term colonization of a Lactobacillus plantarum synbiotic preparation in the neonatal gut, J Pediatr Gastroenterol Nutr. juill, vol.47, issue.1, pp.45-53, 2008.

N. J. Dennison, N. Jupatanakul, and G. Dimopoulos, The mosquito microbiota influences vector competence for human pathogens, Curr Opin Insect Sci. 1 sept, vol.3, pp.6-13, 2014.

A. Boissière, M. T. Tchioffo, D. Bachar, L. Abate, M. A. Nsango et al., Midgut microbiota of the malaria mosquito vector Anopheles gambiae and interactions with Plasmodium falciparum infection, PLoS Pathog, vol.8, issue.5, p.1002742, 2012.

M. Gendrin, F. H. Rodgers, R. S. Yerbanga, J. B. Ouédraogo, M. Basáñez et al., Antibiotics in ingested human blood affect the mosquito microbiota and capacity to transmit malaria, Nat Commun. 6 janv, vol.6, p.5921, 2015.

, Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome, Nature. 13 juin, vol.486, issue.7402, pp.207-221, 2012.

F. Hildebrand, T. Nguyen, B. Brinkman, R. G. Yunta, B. Cauwe et al., Inflammation-associated enterotypes, host genotype, cage and inter-individual effects drive gut microbiota variation in common laboratory mice, Genome Biol. 24 janv, vol.14, issue.1, p.4, 2013.

L. A. David, C. F. Maurice, R. N. Carmody, D. B. Gootenberg, J. E. Button et al., Diet rapidly and reproducibly alters the human gut microbiome, Nature. 23 janv, vol.505, issue.7484, pp.559-63, 2014.

B. Yilmaz, S. Portugal, T. M. Tran, R. Gozzelino, S. Ramos et al., Gut Microbiota Elicits a Protective Immune Response against Malaria Transmission, Cell. 4 déc, vol.159, issue.6, pp.1277-89, 2014.

S. Yooseph, E. F. Kirkness, T. M. Tran, D. M. Harkins, M. B. Jones et al., Stool microbiota composition is associated with the prospective risk of Plasmodium falciparum infection, BMC Genomics. 22 août, vol.16, issue.1, 2015.

N. F. Villarino, G. R. Lecleir, J. E. Denny, S. P. Dearth, C. L. Harding et al., Composition of the gut microbiota modulates the severity of malaria, PNAS. 23 févr, vol.113, issue.8, pp.2235-2275, 2016.

J. P. Mooney, K. L. Lokken, M. X. Byndloss, M. D. George, E. M. Velazquez et al., Inflammation-associated alterations to the intestinal microbiota reduce colonization resistance against non-typhoidal Salmonella during concurrent malaria parasite infection, Scientific Reports, vol.5, p.14603, 2015.

T. Taniguchi, E. Miyauchi, S. Nakamura, M. Hirai, K. Suzue et al., Plasmodium berghei ANKA causes intestinal malaria associated with dysbiosis. Sci Rep, vol.5, p.15699, 2015.

M. P. Soares and B. Yilmaz, Microbiota Control of Malaria Transmission, Trends Parasitol. févr, vol.32, issue.2, pp.120-150, 2016.

A. Cabezas-cruz and J. De-la-fuente, Immunity to ?-Gal: The Opportunity for Malaria and Tuberculosis Control, Front Immunol, vol.8, 2017.

R. Ménard, J. Tavares, I. Cockburn, M. Markus, F. Zavala et al., Looking under the skin: the first steps in malarial infection and immunity, Nat Rev Microbiol, vol.11, issue.10, pp.701-713, 2013.

L. S. Rickman, T. R. Jones, G. W. Long, S. Paparello, I. Schneider et al., Plasmodium falciparum-infected Anopheles stephensi inconsistently transmit malaria to humans, Am J Trop Med Hyg, vol.43, issue.5, pp.441-446, 1990.

R. W. Sauerwein, M. Roestenberg, and V. S. Moorthy, Experimental human challenge infections can accelerate clinical malaria vaccine development, Nat Rev Immunol, vol.11, issue.1, pp.57-64, 2011.

D. F. Verhage, D. Telgt, J. T. Bousema, C. C. Hermsen, G. Van-gemert et al., Clinical outcome of experimental human malaria induced by Plasmodium falciparum-infected mosquitoes, Neth J Med. févr, vol.63, issue.2, pp.52-60, 2005.

O. Oyelaran, L. M. Mcshane, L. Dodd, and J. C. Gildersleeve, Profiling human serum antibodies with a carbohydrate antigen microarray, J Proteome Res. sept, vol.8, issue.9, pp.4301-4311, 2009.

S. Avrameas, Natural autoantibodies: from « horror autotoxicus » to « gnothi seauton, Immunol Today. mai, vol.12, issue.5, pp.154-163, 1991.

J. R. Bishop and P. Gagneux, Evolution of carbohydrate antigens--microbial forces shaping host glycomes?, Glycobiology. mai, vol.17, issue.5, pp.23-34, 2007.

T. Hayakawa, Y. Satta, P. Gagneux, A. Varki, and N. Takahata, Alu-mediated inactivation of the human CMP-N-acetylneuraminic acid hydroxylase gene, Proc Natl Acad Sci U S A. 25 sept, vol.98, issue.20, pp.11399-404, 2001.

P. Tangvoranuntakul, P. Gagneux, S. Diaz, M. Bardor, N. Varki et al., Human uptake and incorporation of an immunogenic nonhuman dietary sialic acid, Proc Natl Acad Sci, vol.100, issue.21, pp.12045-50, 2003.

U. Galili and K. Swanson, Gene sequences suggest inactivation of alpha-1,3-galactosyltransferase in catarrhines after the divergence of apes from monkeys, Proceedings of the National Academy of Sciences of the United States of America. 15 août, vol.88, issue.16, p.7401, 1991.

U. Galili, E. A. Rachmilewitz, A. Peleg, and I. Flechner, A unique natural human IgG antibody with anti-alpha-galactosyl specificity, J Exp Med, vol.160, issue.5, pp.1519-1550, 1984.

C. Cywes-bentley, D. Skurnik, T. Zaidi, D. Roux, R. B. Deoliveira et al., Antibody to a conserved antigenic target is protective against diverse prokaryotic and eukaryotic pathogens, Proc Natl Acad Sci, vol.110, issue.24, pp.2209-2218, 2013.

A. Varki and P. Gagneux, Human-specific evolution of sialic acid targets: explaining the malignant malaria mystery?, Proc Natl Acad Sci USA. 1 sept, vol.106, issue.35, pp.14739-14779, 2009.

C. S. Macedo, . De, R. T. Schwarz, A. R. Todeschini, J. O. Previato et al., Overlooked post-translational modifications of proteins in Plasmodium falciparum: Nand O-glycosylation --a review, Mem Inst Oswaldo Cruz. déc, vol.105, issue.8, pp.949-56, 2010.

U. Galili, R. E. Mandrell, R. M. Hamadeh, S. B. Shohet, and J. M. Griffiss, Interaction between human natural anti-alpha-galactosyl immunoglobulin G and bacteria of the human flora, Infect Immun. juill, vol.56, issue.7, pp.1730-1737, 1988.

R. Ramasamy and R. T. Reese, A role for carbohydrate moieties in the immune response to malaria, J Immunol. mars, vol.134, issue.3, pp.1952-1957, 1985.

R. Ramasamy and R. T. Reese, Terminal galactose residues and the antigenicity of Plasmodium falciparum glycoproteins, Mol Biochem Parasitol. mai, vol.19, issue.2, pp.91-101, 1986.

R. Ramasamy, M. Ramasamy, and S. Yasawardena, Antibodies and Plasmodium falciparum merozoites, Trends in Parasitology. 1 avr, vol.17, issue.4, pp.194-201, 2001.

R. Ramasamy and M. C. Field, Terminal galactosylation of glycoconjugates in Plasmodium falciparum asexual blood stages and Trypanosoma brucei bloodstream trypomastigotes, Exp Parasitol. avr, vol.130, issue.4, pp.314-334, 2012.

E. H. Nardin, V. Nussenzweig, R. S. Nussenzweig, W. E. Collins, K. T. Harinasuta et al., Circumsporozoite proteins of human malaria parasites Plasmodium falciparum and Plasmodium vivax, J Exp Med. 1 juill, vol.156, issue.1, pp.20-30, 1982.

Y. Yang, E. Ohdan, H. Bracy, J. L. Xu, Y. Iacomini et al., Tolerization of Anti-Gal?1-3Gal Natural Antibody-forming B Cells by Induction of Mixed Chimerism, J Exp Med. 20 avr, vol.187, issue.8, pp.1335-1377, 1998.

T. R. Chiang, L. Fanget, R. Gregory, Y. Tang, D. L. Ardiet et al., Anti-Gal antibodies in humans and 1, 3alpha-galactosyltransferase knock-out mice, Transplantation. 27 juin, vol.69, issue.12, pp.2593-600, 2000.

R. G. Tearle, M. J. Tange, Z. L. Zannettino, M. Katerelos, T. A. Shinkel et al., The alpha-1,3-galactosyltransferase knockout mouse. Implications for xenotransplantation, Transplantation. 15 janv, vol.61, issue.1, pp.13-22, 1996.

B. A. Macher and U. Galili, The Galalpha1,3Galbeta1,4GlcNAc-R (alpha-Gal) epitope: a carbohydrate of unique evolution and clinical relevance, Biochim Biophys Acta. févr, vol.1780, issue.2, pp.75-88, 2008.

S. Avrameas, Natural autoantibodies: from « horror autotoxicus » to « gnothi seauton, Immunol Today. mai, vol.12, issue.5, pp.154-163, 1991.

L. Wang, M. Z. Radic, and U. Galili, Human anti-Gal heavy chain genes. Preferential use of VH3 and the presence of somatic mutations, The Journal of Immunology. 1 août, vol.155, issue.3, pp.1276-85, 1995.

S. C. Pal, K. Rao, C. Kereselidze, T. Krishnaswami, A. K. Murty et al., An extensive community outbreak of enteropathogenic Escherichia coli O86: B7 gastroenteritis, Bull World Health Organ, vol.41, issue.6, pp.851-859, 1969.

K. J. Posekany, H. K. Pittman, J. F. Bradfield, C. E. Haisch, and K. M. Verbanac, Induction of Cytolytic Anti-Gal Antibodies in ?-1,3-Galactosyltransferase Gene Knockout Mice by Oral Inoculation with Escherichia coli O86:B7 Bacteria, Infection and Immunity, vol.70, issue.11, pp.6215-6237, 2002.

R. Mañez, F. J. Blanco, I. Díaz, A. Centeno, E. Lopez-pelaez et al., Removal of bowel aerobic gram-negative bacteria is more effective than immunosuppression with cyclophosphamide and steroids to decrease natural alpha-galactosyl IgG antibodies, Xenotransplantation. févr, vol.8, issue.1, pp.15-23, 2001.

G. F. Springer, R. E. Horton, and M. Forbes, Origin of anti-human blood group B agglutinins in white Leghorn chicks, J Exp Med. 1 août, vol.110, issue.2, pp.221-265, 1959.

G. F. Springer and R. E. Horton, Blood group isoantibody stimulation in man by feeding blood group-active bacteria, J Clin Invest. juill, vol.48, issue.7, pp.1280-91, 1969.

J. L. Avila, M. Rojas, and U. Galili, Immunogenic Gal alpha 1----3Gal carbohydrate epitopes are present on pathogenic American Trypanosoma and Leishmania, J Immunol. 15 avr, vol.142, issue.8, pp.2828-2862, 1989.

U. Galili, B. A. Macher, J. Buehler, and S. B. Shohet, Human natural anti-alpha-galactosyl IgG. II. The specific recognition of alpha (1----3)-linked galactose residues, J Exp Med. 1 août, vol.162, issue.2, pp.573-82, 1985.

U. Galili, J. Buehler, S. B. Shohet, and B. A. Macher, The human natural anti-Gal IgG. III. The subtlety of immune tolerance in man as demonstrated by crossreactivity between natural anti-Gal and anti-B antibodies, J Exp Med. 1 mars, vol.165, issue.3, pp.693-704, 1987.

U. Doenz, U. E. Nydegger, A. Kueng, T. Carrel, and P. Mohacsi, Anti-Galalpha1-3Gal IgM/IgG antibody levels in infants: do they have a clinical relevance in pediatric xenotransplantation?, J Heart Lung Transplant, vol.19, issue.11, pp.1108-1121, 2000.

D. C. Latemple and U. Galili, Adult and neonatal anti-Gal response in knock-out mice for alpha1,3galactosyltransferase, Xenotransplantation. août, vol.5, issue.3, pp.191-197, 1998.

J. F. Kearney, P. Patel, E. K. Stefanov, and R. G. King, Natural antibody repertoires: development and functional role in inhibiting allergic airway disease, Annu Rev Immunol, vol.33, pp.475-504, 2015.

A. Lundell, V. Björnsson, A. Ljung, M. Ceder, S. Johansen et al., Infant B Cell Memory Differentiation and Early Gut Bacterial Colonization, The Journal of Immunology. 1 mai, vol.188, issue.9, pp.4315-4337, 2012.

C. G. Buffie and E. G. Pamer, Microbiota-mediated colonization resistance against intestinal pathogens, Nat Rev Immunol, vol.13, issue.11, pp.790-801, 2013.

X. Fan, A. Ang, S. M. Pollock-barziv, A. I. Dipchand, P. Ruiz et al., Donor-specific B-cell tolerance after ABO-incompatible infant heart transplantation, Nature Medicine, vol.10, issue.11, pp.1227-1260, 2004.

D. P. Kwiatkowski, How Malaria Has Affected the Human Genome and What Human Genetics Can Teach Us about Malaria, Am J Hum Genet. août, vol.77, issue.2, pp.171-92, 2005.

L. Turner, T. Lavstsen, S. S. Berger, C. W. Wang, J. Petersen et al., Severe malaria is associated with parasite binding to endothelial protein C receptor, Nature. 27 juin, vol.498, issue.7455, pp.502-507, 2013.

R. J. Pleass, S. C. Moore, L. Stevenson, and L. Hviid, Immunoglobulin M: Restrainer of Inflammation and Mediator of Immune Evasion by Plasmodium falciparum Malaria, Trends Parasitol. févr, vol.32, issue.2, pp.108-127, 2016.

J. Aakko, A. Endo, C. Mangani, K. Maleta, P. Ashorn et al., Distinctive Intestinal Lactobacillus Communities in 6-Month-Old Infants From Rural Malawi and Southwestern Finland, J Pediatr Gastroenterol Nutr. déc, vol.61, issue.6, pp.641-649, 2015.

J. Stough, S. P. Dearth, J. E. Denny, G. R. Lecleir, N. W. Schmidt et al., Functional Characteristics of the Gut Microbiome in C57BL/6 Mice Differentially Susceptible to Plasmodium yoelii, Front Microbiol, vol.7, p.1520, 2016.

P. Cabrales, G. M. Zanini, D. Meays, J. A. Frangos, and L. Carvalho, Nitric oxide protection against murine cerebral malaria is associated with improved cerebral microcirculatory physiology, J Infect Dis. 15 mai, vol.203, issue.10, pp.1454-63, 2011.

L. Serghides, H. Kim, Z. Lu, D. C. Kain, C. Miller et al., Inhaled Nitric Oxide Reduces Endothelial Activation and Parasite Accumulation in the Brain, and Enhances Survival in Experimental Cerebral Malaria, PLOS ONE, vol.6, issue.11, p.27714, 2011.

Y. C. Martins, G. M. Zanini, J. A. Frangos, and L. Carvalho, Efficacy of different nitric oxidebased strategies in preventing experimental cerebral malaria by Plasmodium berghei ANKA, PLoS ONE, vol.7, issue.2, p.32048, 2012.

D. Burgner, W. Xu, K. Rockett, M. Gravenor, I. G. Charles et al., Inducible nitric oxide synthase polymorphism and fatal cerebral malaria, Lancet, vol.352, issue.9135, pp.1193-1197, 1998.

X. B. Ji and T. C. Hollocher, Reduction of nitrite to nitric oxide by enteric bacteria, Biochem Biophys Res Commun, vol.157, issue.1, pp.106-114, 1988.

B. R. Crane, J. Sudhamsu, and B. A. Patel, Bacterial nitric oxide synthases, Annu Rev Biochem, vol.79, pp.445-70, 2010.

T. Sobko, C. I. Reinders, E. Jansson, E. Norin, T. Midtvedt et al., Gastrointestinal bacteria generate nitric oxide from nitrate and nitrite, Nitric Oxide. déc, vol.13, issue.4, pp.272-280, 2005.

J. Vermeiren, T. Van-de-wiele, W. Verstraete, P. Boeckx, and N. Boon, Nitric oxide production by the human intestinal microbiota by dissimilatory nitrate reduction to ammonium, J Biomed Biotechnol, p.284718, 2009.

D. Modiano, V. Petrarca, B. S. Sirima, I. Nebié, D. Diallo et al., Different response to Plasmodium falciparum malaria in West African sympatric ethnic groups, Proc Natl Acad Sci, vol.93, issue.23, pp.13206-13217, 1996.

R. D. Astronomo and D. R. Burton, Carbohydrate vaccines: developing sweet solutions to sticky situations?, Nat Rev Drug Discov. avr, vol.9, issue.4, pp.308-332, 2010.

C. Davitt and E. C. Lavelle, Delivery strategies to enhance oral vaccination against enteric infections, Adv Drug Deliv Rev. 30 août, vol.91, pp.52-69, 2015.

B. A. Cobb, Q. Wang, A. O. Tzianabos, and D. L. Kasper, Polysaccharide processing and presentation by the MHCII pathway, Cell. 28 mai, vol.117, issue.5, pp.677-87, 2004.

R. A. Seder, L. Chang, M. E. Enama, K. L. Zephir, U. N. Sarwar et al., Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine, Science. 20 sept, vol.341, issue.6152, pp.1359-65, 2013.

C. Cruz, A. Valdés, J. J. De-la-fuente, and J. , Control of vector-borne infectious diseases by human immunity against ?-Gal, Expert Rev Vaccines, vol.15, issue.8, pp.953-958, 2016.

J. Hackett, Use of Salmonella for heterologous gene expression and vaccine delivery systems, Curr Opin Biotechnol. oct, vol.4, issue.5, pp.611-616, 1993.

J. Holmgren and C. Czerkinsky, Cholera as a model for research on mucosal immunity and development of oral vaccines, Current Opinion in Immunology. 1 janv, vol.4, issue.4, pp.387-91, 1992.

Z. Zhang, P. Jiang, N. Li, M. Shi, and W. Huang, Oral vaccination of mice against rodent malaria with recombinant Lactococcus lactis expressing MSP-1(19), World J Gastroenterol, vol.11, issue.44, pp.6975-80, 2005.

A. Mangold, D. Hercher, G. Hlavin, J. Liepert, M. Zimmermann et al., Antialpha-Gal antibody titres remain unaffected by the consumption of fermented milk containing Lactobacillus casei in healthy adults, Int J Food Sci Nutr. mai, vol.63, issue.3, pp.278-82, 2012.

M. Mikelsaar, E. Sepp, J. ?t?epetova, E. Songisepp, and R. Mändar, Biodiversity of Intestinal Lactic Acid Bacteria in the Healthy Population, Adv Exp Med Biol, vol.932, pp.1-64, 2016.

J. Van-hylckama-vlieg, J. Rademaker, H. Bachmann, D. Molenaar, W. J. Kelly et al., Natural diversity and adaptive responses of Lactococcus lactis, Curr Opin Biotechnol. avr, vol.17, issue.2, pp.183-90, 2006.

F. Bottacini, M. Ventura, D. Van-sinderen, O. Motherway, and M. , Diversity, ecology and intestinal function of bifidobacteria, Microb Cell Fact. 29 août, vol.13, 2014.

C. Chen, B. Liu, Y. Xu, N. Utkina, D. Zhou et al., Biochemical characterization of the novel ?-1, 3-galactosyltransferase WclR from Escherichia coli O3, Carbohydr Res. 22 juill, vol.430, pp.36-43, 2016.

W. Han, L. Cai, B. Wu, L. Li, Z. Xiao et al., The wciN gene encodes an ?-1,3-galactosyltransferase involved in the biosynthesis of the capsule repeating unit of Streptococcus pneumoniae serotype 6B, Biochemistry. 24 juill, vol.51, issue.29, pp.5804-5814, 2012.

H. Kumar, S. Salminen, H. Verhagen, I. Rowland, J. Heimbach et al., Novel probiotics and prebiotics: road to the market, Curr Opin Biotechnol. avr, vol.32, pp.99-103, 2015.

N. Jain and W. A. Walker, Diet and host-microbial crosstalk in postnatal intestinal immune homeostasis, Nat Rev Gastroenterol Hepatol. janv, vol.12, issue.1, pp.14-25, 2015.

R. Oozeer, K. Van-limpt, T. Ludwig, B. Amor, K. Martin et al., Intestinal microbiology in early life: specific prebiotics can have similar functionalities as humanmilk oligosaccharides, Am J Clin Nutr. août, vol.98, issue.2, pp.561-71, 2013.

C. Chassard, T. De-wouters, and C. Lacroix, Probiotics tailored to the infant: a window of opportunity, Curr Opin Biotechnol. avr, vol.26, pp.141-148, 2014.

E. C. Gritz and V. Bhandari, The human neonatal gut microbiome: a brief review, Front Pediatr, vol.3, p.17, 2015.