F. Saji, Y. Samejima, S. Kamiura, and M. Koyama, Dynamics of immunoglobulins at the fetomaternal interface, Rev Reprod, vol.4, issue.2, pp.81-90, 1999.

A. Malek, R. Sager, P. Kuhn, K. H. Nicolaides, and H. Schneider, Evolution of maternofetal transport of immunoglobulins during human pregnancy, Am J Reprod Immunol, vol.36, issue.5, pp.248-55, 1996.

P. Palmeira, Y. Ito, L. Arslanian, C. Carneiro-sampaio, and M. M. , Passive immunity acquisition of maternal anti-enterohemorrhagic Escherichia coli (EHEC) O157:H7 IgG antibodies by the newborn

, Eur J Pediatr, vol.166, issue.5, pp.413-422, 2006.

, Bull Acad Natl Med, vol.196, issue.8, pp.1601-1613, 2012.

M. Doroudchi, S. Dehaghani, A. Emad, K. Ghaderi, and A. , Placental transfer of rubellaspecific IgG in fullterm and preterm newborns, 57-van den Berg JP, vol.81, pp.157-62, 2003.

R. M. Elburg, V. L. Krebs, T. B. Brasil, G. N. Pontes, M. Carneiro-sampaio et al., Transplacental transport of IgG antibodies specific for pertussis, diphtheria, tetanus, haemophilus influenzae type b, and Neisseria meningitidis serogroup C is lower in preterm compared with term infants, FEMS Immunol Med Microbiol, vol.29, issue.9, pp.236-279, 2010.

-. Zaman, M. B. , B. S. Ph, E. Roy, M. B. et al., Influenza immunization in pregnancy--antibody responses in mothers and infants, N Engl J Med, vol.359, issue.17, pp.1644-1650, 2008.

I. --benowitz, D. B. Esposito, and K. D. Gracey, Influenza vaccine given to pregnant women reduces hospitalization due to influenza in their infants, Clin. Infect. Dis, vol.51, pp.1355-61, 2010.

A. A. Eick, T. M. Uyeki, A. Klimov, H. Hall, R. Reid et al., Maternal influenza vaccination and effect on influenza virus infection in young infants, Arch Pediatr Adolesc Med, vol.165, issue.2, pp.104-115, 2011.

, Ministère des affaires sociales et de la santé -Vaccination contre la grippe -saison 2012-2013 : Questions / Réponses -Professionnels de santé

O. Ballard and A. L. Morrow, Human Milk Composition: Nutrients and Bioactive Factors, Pediatr Clin North Am, vol.60, issue.1, pp.49-74, 2013.

C. Castellote, R. Casillas, C. Ramírez-santana, F. J. Pérez-cano, M. Castell et al., Premature delivery influences the immunological composition of colostrum and transitional and mature human milk, J Nutr, vol.141, issue.6, pp.1181-1188, 2011.

J. K. Kulski and P. E. Hartmann, Changes in human milk composition during the initiation of lactation

, Aust J Exp Biol Med Sci, vol.59, issue.1, pp.101-115, 1981.

L. Hanson, S. A. Silfverdal, L. Strömbäck, V. Erling, S. Zaman et al., The immunological role of breast feeding, Pediatr Allergy Immunol, vol.12, issue.2, pp.36-40, 2001.

P. Brandtzaeg, The mucosal immune system and its integration with the mammary glands, J Pediatr, vol.156, issue.2, pp.8-15, 2010.

K. A. Kadaoui, B. Corthésy, and . Immunol, Secretory IgA mediates bacterial translocation to dendritic cells in mouse Peyer's patches with restriction to mucosal compartment, vol.179, pp.7751-7758, 2007.

E. M. Lilius and P. Marnila, The role of colostral antibodies in prevention of microbial infections, Curr Opin Infect Dis, vol.14, issue.3, pp.295-300, 2001.

R. I. Glass, A. M. Svennerholm, B. J. Stoll, M. R. Khan, K. M. Hossain et al., Protection against cholera in breast-fed children by antibodies in breast milk, N Engl J Med, vol.308, issue.23, pp.1389-92, 1983.

G. M. -ruiz-palacios, J. J. Calva, L. K. Pickering, Y. Lopez-vidal, P. Volkow et al., Protection of breast-fed infants against Campylobacter diarrhea by antibodies in human milk, J Pediatr, vol.116, issue.5, pp.707-720, 1990.

L. Walter, P. K. Hurley, and . Theil, Perspectives on Immunoglobulins in Colostrum and Milk Nutrients, vol.3, pp.442-474, 2011.

D. Turcka, ;. Vidailhetb, A. Bocquetc, J. Bressond, A. Briende et al., Simeonik Allaitement maternel : les bénéfices pour la santé de l'enfant et de sa mère, Archives de Pédiatrie, vol.20, pp.29-48, 2013.

, 76-Fondation Canadienne de l'Allaitement. (18/10/2018) Comment le lait maternel protège les nouveaux-nés

M. G. Rooks and W. S. Garrett, Gut microbiota, metabolites and host immunity, Nat Rev Immunol, vol.16, issue.6, pp.341-52, 2016.

T. Gensollen, S. S. Iyer, D. I. Kasper, and R. S. Blumberg, How colonization by microbiota in early life shapes the immune system, Science, vol.352, issue.6285, pp.539-583, 2016.

K. Honda and D. R. Littman, The microbiota in adaptive immune homeostasis and disease, Nature, vol.535, issue.7610, pp.75-84, 2016.

S. 80-christian-milani, F. Duranti, E. Bottacini, F. Casey, J. Turroni et al., The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota, Microbiol Mol Biol Rev, vol.81, issue.4, pp.36-53, 2017.

-. Levy, Innate immunity of the newborn: basic mechanisms and clinical correlates, Nature Reviews Immunology, vol.7, pp.379-390, 2007.

N. T. Mueller, E. Bakacs, J. Combellick, Z. Grigoryan, and M. G. Dominguez-bello, The infant microbiome development: mom matters, Trends Mol Med, vol.21, issue.2, pp.109-117, 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, vol.6, issue.237, pp.237-65, 2014.

T. -tatsuo-ichinohe, T. Miyama, Y. Kawase, K. Honjo, H. Kitaura et al., Generation Immune Repertoire Sequencing as a Clue to Elucidate the Landscape of Immune Modulation by Host-Gut Microbiome Interactions, Front Immunol, vol.9, p.668, 2018.

J. J. -faith, F. E. Rey, D. O'donnell, M. Karlsson, N. P. Mcnulty et al., Creating and characterizing communities of human gut microbes in gnotobiotic mice, ISME J, vol.4, issue.9, pp.1094-1102, 2010.

B. Deplancke and H. R. Gaskins, Microbial modulation of innate defense: goblet cells and the intestinal mucus layer, Am J Clin Nutr, vol.73, issue.6, pp.1131-1141, 2001.

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, vol.19, issue.2, pp.59-69, 2007.

O. Pabst, New concepts in the generation and functions of IgA, Nat Rev Immunol, vol.12, issue.12, pp.821-853, 2012.

S. K. Mazmanian, C. H. Liu, A. O. Tzianabos, and D. L. Kasper, An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system, Cell, vol.122, issue.1, pp.107-125, 2005.

I. -ivanov, F. Rde, L. Manel, N. Yoshinaga, K. Rifkin et al., Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine, Cell Host Microbe, vol.4, issue.4, pp.337-386, 2008.

U. G. -strauch, F. Obermeier, N. Grunwald, S. Gürster, N. Dunger et al., Influence of intestinal bacteria on induction of regulatory T cells: lessons from a transfer model of colitis, Gut, vol.54, issue.11, pp.1546-52, 2005.

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

M. G. Rooks and W. S. Garrett, Gut microbiota, metabolites and host immunity, Nat Rev Immunol, vol.16, issue.6, pp.341-352, 2016.

P. M. -smith, M. R. Howitt, N. Panikov, M. Michaud, C. A. Gallini et al., The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis, Science, vol.341, issue.6145, pp.569-73, 2013.

A. Trompette, E. S. Gollwitzer, K. Yadava, A. K. Sichelstiel, N. Sprenger et al., Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis, Nat Med, vol.20, issue.2, pp.159-66, 2014.

A. N. -thorburn, C. I. Mckenzie, S. Shen, D. Stanley, L. Macia et al., Evidence that asthma is a developmental origin disease influenced by maternal diet and bacterial metabolites, Nat Commun, vol.6, p.7320, 2015.

Y. Furusawa, Y. Obata, S. Fukuda, T. A. Endo, G. Nakato et al., Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells, Nature, vol.504, issue.7480, pp.446-50, 2013.

E. Avershina, K. Lundgård, M. Sekelja, C. Dotterud, O. Storrø et al., Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns, Environ Microbiol, vol.18, issue.7, pp.11971-11976, 2010.

D. Chierico, F. Vernocchi, P. Petrucca, A. Paci, P. Fuentes et al., Phylogenetic and Metabolic Tracking of Gut Microbiota during Perinatal Development, PLoS One, vol.10, issue.9, p.137347, 2015.

-. Burcelin, Human gut microbiome viewed across age and geography, Nature, vol.486, issue.7402, pp.222-229, 2009.

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

Y. Belkaid and J. A. Segre, Dialogue between skin microbiota and immunity, Science, vol.346, issue.6212, pp.954-963, 2014.

L. V. Hooper, D. R. Littman, and A. J. Macpherson, Interactions between the microbiota and the immune system, Science, vol.336, issue.6086, pp.1268-73, 2012.

V. Tremaroli and F. Bäckhed, Functional interactions between the gut microbiota and host metabolism, Nature, vol.489, issue.7415, pp.242-251, 2012.

K. J. Maloy and F. Powrie, Intestinal homeostasis and its breakdown in inflammatory bowel disease, Nature, vol.474, issue.7351, pp.298-306, 2011.

M. Pasparakis, I. Haase, and F. O. Nestle, Mechanisms regulating skin immunity and inflammation, Nat Rev Immunol, vol.14, issue.5, pp.289-301, 2014.

E. A. Grice, H. H. Kong, G. Renaud, A. Young, . Program et al., A diversity profile of the human skin microbiota, Genome Res, vol.18, issue.7, pp.1043-50, 2008.

T. C. Scharschmidt, K. S. Vasquez, H. Truong, S. V. Gearty, M. L. Pauli et al., A wave of regulatory T cells into neonatal skin mediates tolerance to commensal microbes, Immunity, vol.43, issue.5, pp.1011-1021, 2015.

Y. E. -chen, M. A. Fischbach, and Y. Belkaid, Skin microbiota-host interactions, Nature, vol.553, issue.7689, pp.427-436, 2018.

T. C. Scharschmidt, Establishing tolerance to commensal skin bacteria: timing is everything, Dermatol Clin, vol.35, issue.1, pp.1-9, 2017.

T. Scharschmidt, K. S. Vasquez, H. A. Truong, S. V. Gearty, M. L. Pauli et al., , vol.43, pp.1011-1032, 2015.

M. Tollin, G. Bergsson, Y. Kai-larsen, J. Lengqvist, J. Sjövall et al., Vernix caseosa as a multi-component defence system based on polypeptides, lipids and their interactions, Cell Mol Life Sci, vol.62, pp.2390-2399, 2005.

M. -zasloff, Vernix, the newborn, and innate defense, Pediatr. Res, vol.53, pp.203-204, 2003.

G. Marchini, A. Nelson, J. Edner, S. Lonne-rahm, A. Stavréus-evers et al., Erythema toxicum neonatorum is an innate immune response to commensal microbes penetrated into the skin of the newborn infant, vol.58, pp.613-619, 2005.

S. Yang, N. Fujikado, D. Kolodin, C. Benoist, and D. Mathis, Immune tolerance. Regulatory T cells generated early in life play a distinct role in maintaining self-tolerance, Science, vol.348, issue.6234, pp.589-94, 2015.

V. O. Rotimi and B. I. Duerden, The development of the bacterial flora in normal neonates, J Med Microbiol, vol.14, issue.1, pp.51-62, 1981.

S. K. Lathrop, S. M. Bloom, S. M. Rao, K. Nutsch, C. W. Lio et al., Peripheral education of the immune system by colonic commensal microbiota, Nature, vol.478, issue.7368, pp.250-254, 2011.

A. -cebula, M. Seweryn, G. A. Rempala, S. S. Pabla, R. A. Mcindoe et al.,

, Thymus-derived regulatory T cells contribute to tolerance to commensal microbiota, Nature, vol.497, issue.7448, pp.258-62, 2009.

C. J. Hill, D. B. Lynch, K. Murphy, M. Ulaszewska, I. B. Jeffery et al., Evolution of gut microbiota composition from birth to 24 weeks in the INFANTMET Cohort, vol.5, p.4, 2017.

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

I. G. Pantoja-feliciano, J. C. Clemente, E. K. Costello, M. E. Perez, M. J. Blaser et al., Biphasic assembly of the murine intestinal microbiota during early development, ISME J, vol.7, issue.6, pp.1112-1117, 2013.

A. J. Macpherson and N. L. Harris, Interactions between commensal intestinal bacteria and the immune system, Nat Rev Immunol, vol.4, issue.6, pp.478-85, 2004.

C. H. Hansen, D. S. Nielsen, M. Kverka, Z. Zakostelska, K. Klimesova et al., Patterns of early gut colonization shape future immune responses of the host, PLoS One, vol.7, issue.3, p.34043, 2012.

M. Grover, ;. Purna-c-kashyap, L. Horowitz-re, . Sm, and . Popper-h, The response of the lymphatic tissue to the microbial flora. Studies on germfree mice, Neurogastroenterol Motil, vol.26, issue.6, pp.471-83, 1963.

I. Mosconi, M. B. Geuking, M. M. Zaiss, J. C. Massacand, C. Aschwanden et al., Intestinal bacteria induce TSLP to promote mutualistic T-cell responses, Mucosal Immunol, vol.6, issue.6, pp.1157-67, 2013.

-. Amenyogbe, T. R. Kollmann, and R. Ben-othman, Early-Life Host-Microbiome Interphase: The Key Frontier for Immune Development, Front Pediatr, vol.5, p.111, 2017.

S. Tamburini, N. Shen, H. C. Wu, and J. C. Clemente, The microbiome in early life: implications for health outcomes, Nat Med, vol.22, issue.7, pp.713-735, 2016.

M. C. Arrieta, L. T. Stiemsma, P. A. Dimitriu, L. Thorson, and S. Russell, Early infancy microbial and metabolic alterations affect risk of childhood asthma, Sci Transl Med, vol.7, issue.307, pp.307-152, 2015.

T. Olszak, D. An, S. Zeissig, M. P. Vera, J. Richter et al., Microbial exposure during early life has persistent effects on natural killer T cell function, vol.336, pp.489-93, 2012.

D. -torsten-olszak, S. An, M. P. Zeissig, J. Vera, A. Richter et al., Microbial Exposure During Early Life Has Persistent Effects on Natural Killer T Cell Function, Science, vol.336, issue.6080, pp.489-493, 2012.

S. Z. Josefowicz and A. Rudensky, Control of regulatory T cell lineage commitment and maintenance, Immunity, vol.30, issue.5, pp.616-641, 2009.

S. K. Lathrop, N. A. Santacruz, D. Pham, J. Luo, and C. S. Hsieh, Antigen-specific peripheral shaping of the natural regulatory T cell population, J Exp Med, vol.205, issue.13, pp.3105-3122, 2008.

A. M. Thornton, P. E. Korty, D. Q. Tran, E. A. Wohlfert, P. E. Murray et al., Expression of Helios, an Ikaros transcription factor family member, differentiates thymicderived from peripherally induced Foxp3+ T regulatory cells, J Immunol, vol.184, issue.7, pp.3433-3474, 2010.

K. Atarashi, T. Tanoue, T. Shima, A. Imaoka, T. Kuwahara et al., Induction of colonic regulatory T cells by indigenous Clostridium species, Science, vol.331, issue.6015, pp.337-378, 2011.

S. K. Lathrop, S. M. Bloom, M. Sindhuja, K. Rao, C. Nutsch et al., Peripheral education of the immune system by colonic commensal microbiota, Nature, vol.478, issue.7368, pp.250-254, 2011.

M. A. -curotto-de-lafaille, N. Kutchukhidze, S. Shen, Y. Ding, H. Yee et al., Adaptive Foxp3+ regulatory T cell-dependent and -independent control of allergic inflammation, Immunity, vol.29, issue.1, pp.114-140, 2008.

C. M. -sun, J. A. Hall, R. B. Blank, N. Bouladoux, M. Oukka et al., Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid, J Exp Med, vol.204, issue.8, pp.1775-85, 2007.

J. L. -round and S. K. Mazmanian, Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota, Proc Natl Acad Sci, vol.107, issue.27, pp.12204-12213, 2010.

S. A. De-pavert and R. E. Mebius, New insights into the development of lymphoid tissues, Nat Rev Immunol, vol.10, issue.9, pp.664-74, 2010.

C. L. Maynard, C. O. Elson, R. D. Hatton, and C. T. Weaver, Reciprocal Interactions of the Intestinal Microbiota and Immune System, Nature, vol.489, issue.7415, pp.231-241, 2012.

G. Eberl and M. Lochner, The development of intestinal lymphoid tissues at the interface of self and microbiota, Mucosal Immunol, vol.2, issue.6, pp.478-85, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-00509629

B. J. Sutton and H. J. Gould, The human IgE network, Nature, vol.366, issue.6454, pp.421-429, 1993.

C. O. Qing-zhao and . Elson, Adaptive immune education by gut microbiota antigens, Immunology, vol.154

J. Cahenzli, Y. Köller, M. Wyss, M. B. Geuking, and K. D. ,

. Mccoy, Intestinal Microbial Diversity during Early-Life Colonization Shapes Long-Term IgE Levels, Cell Host Microbe, vol.14, issue.5, pp.559-570, 2013.

J. Cahenzli, Y. Köller, M. Wyss, M. B. Geuking, and K. D. ,

. Mccoy, Intestinal Microbial Diversity during Early-Life Colonization Shapes Long-Term IgE Levels, Cell Host Microbe, vol.14, issue.5, pp.559-570, 2013.

T. Herbst, A. Sichelstiel, C. Schär, K. Yadava, K. Bürki et al., Dysregulation of allergic airway inflammation in the absence of microbial colonization, Am J Respir Crit Care Med, vol.184, issue.2, pp.198-205, 2011.

K. D. Mccoy, N. L. Harris, P. Diener, S. Hatak, B. Odermatt et al., Natural IgE production in the absence of MHC Class II cognate help, Immunity, vol.24, issue.3, pp.329-368, 2006.

P. G. Holt, Programming for responsiveness to environmental antigens that trigger allergic respiratory disease in adulthood is initiated during the perinatal period. Environ Health Perspect, vol.106, pp.795-800, 1998.

A. J. Macpherson, D. Gatto, E. Sainsbury, G. R. Harriman, H. Hengartner et al., A primitive T cell-independent mechanism of intestinal mucosal IgA responses to commensal bacteria, Science, vol.288, issue.5474, pp.2222-2228, 2000.

A. J. Macpherson and T. Uhr, Induction of protective IgA by intestinal dendritic cells carrying commensal bacteria, Science, vol.303, issue.5664, pp.1662-1667, 2004.

K. Moor, M. Diard, M. E. Sellin, B. Felmy, S. Y. Wotzka et al., High-avidity IgA protects the intestine by enchaining growing bacteria, Nature, vol.544, issue.7651, pp.498-502, 2017.

S. Johnson, W. D. Sypura, D. N. Gerding, S. L. Ewing, and E. N. Janoff, Selective neutralization of a bacterial enterotoxin by serum immunoglobulin A in response to mucosal disease, Infect Immun, vol.63, issue.8, pp.3166-73, 1995.

D. 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, pp.328-367, 2007.

C. Lindner, B. Wahl, L. Fohse, S. Suerbaum, A. J. Macpherson et al., Age, microbiota, and T cells shape diverse individual IgA repertoires in the intestine, J Exp Med, vol.209, pp.365-77, 2012.

S. Hapfelmeier, M. A. Lawson, E. Slack, J. K. Kirundi, M. Stoel et al., Reversible microbial colonization of germ-free mice reveals the dynamics of IgA immune responses, Science, vol.328, issue.5986, pp.1705-1714, 2010.

C. Lindner, I. Thomsen, B. Wahl, M. Ugur, M. K. Sethi et al., Diversification of memory B cells drives the continuous adaptation of secretory antibodies to gut microbiota, Nat Immunol, vol.16, pp.880-888, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01604354

E. -calderón-gómez, H. Bassolas-molina, R. Mora-buch, D. I. Planell, N. Esteller et al., Commensal-Specific CD4(+) Cells From Patients With Crohn's Disease Have a, p.17

. Inflammatory-profile, U. Gastroenterology-;--pirzer, A. Schonhaar, B. Fleischer, E. Hermann et al., Reactivity of infiltrating T lymphocytes with microbial antigens in Crohn's disease, Lancet, vol.151, issue.3, pp.1238-1247, 1991.

A. N. Hegazy, N. R. West, M. Stubbington, E. Wendt, K. Suijker et al., Circulating and Tissue-Resident CD4+ T Cells With Reactivity to Intestinal Microbiota Are Abundant in Healthy Individuals and Function Is Altered During Inflammation, Gastroenterology, vol.153, issue.5, pp.1320-1337, 2017.

A. -pianta, S. Arvikar, K. Strle, E. E. Drouin, Q. Wang et al., Evidence of the immune relevance of Prevotella copri, a gut microbe, in patients with rheumatoid arthritis, Arthritis Rheumatol, vol.69, pp.964-75, 2017.

A. -pianta, S. Arvikar, K. Strle, E. E. Drouin, Q. Wang et al., Evidence of the immune relevance of Prevotella copri, a gut microbe, in patients with rheumatoid arthritis, Arthritis Rheumatol, vol.69, pp.964-75, 2017.

M. K. Tulic, P. O. Fiset, J. J. Manoukian, S. Frenkiel, F. Lavigne et al., Role of toll-like receptor 4 in protection by bacterial lipopolysaccharide in the nasal mucosa of atopic children but not adults, Lancet, vol.363, issue.9422, pp.1689-97, 2004.

H. Alenius, J. Pakarinen, O. Saris, M. A. Andersson, M. Leino et al., Contrasting immunological effects of two disparate dusts -preliminary observations, Int Arch Allergy Immunol, vol.149, issue.1, pp.81-90, 2009.

S. H. Butchart, M. Walpole, C. B. Van-strien, A. Scharlemann, J. P. Almond et al., Global biodiversity: indicators of recent declines, Science, vol.328, issue.5982, pp.1164-1172, 2010.

I. -leena-von-hertzen, T. Hanski, and . Haahtela, Biodiversity loss and inflammatory diseases are two global megatrends that might be related, EMBO Rep, vol.12, issue.11, pp.1089-1093, 2011.

A. E. -thomas and . Platts-mills, The Allergy Epidemics: 1870-2010, J Allergy Clin Immunol, vol.136, issue.1, pp.3-13, 2015.

T. A. Platts-mills, The allergy epidemics: 1870-2010, J Allergy Clin Immunol, vol.136, issue.1, pp.3-13, 2015.

T. Leah, L. A. Stiemsma, S. E. Reynolds, B. Turvey, and . Brett, The hygiene hypothesis: current perspectives and future therapies, Finlay Immunotargets Ther, vol.4, pp.143-157, 2015.

H. Okada, C. Kuhn, H. Feillet, and J. F. Bach, The 'hygiene hypothesis' for autoimmune and allergic diseases: an update, Clin Exp Immunol, vol.160, issue.1, pp.1-9, 2010.

, The International Study of Asthma and Allergies in Childhood (ISAAC) Steering Committee. Worldwide variation in prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and atopic eczema, Lancet, vol.351, issue.9111, pp.1225-1257, 1998.

D. P. Strachan, Hay fever, hygiene, and household size, BMJ, vol.299, issue.6710, pp.1259-60, 1989.

D. P. Strachan, E. M. Taylor, and R. G. Carpenter, Family structure, neonatal infection, and hay fever in adolescence, Arch Dis Child, vol.74, issue.5, pp.422-428, 1996.

D. R. -ownby, C. C. Johnson, and E. L. Peterson, Exposure to dogs and cats in the first year of life and risk of allergic sensitization at 6 to 7 years of age, JAMA, vol.288, issue.8, pp.963-72, 2002.

C. S. -benn, M. Melbye, J. Wohlfahrt, B. Björkstén, and P. Aaby, Cohort study of sibling effect, infectious diseases, and risk of atopic dermatitis during first 18 months of life, BMJ, vol.328, issue.7450, p.1223, 2004.

P. M. -matricardi, F. Rosmini, S. Riondino, M. Fortini, L. Ferrigno et al., Exposure to foodborne and orofecal microbes versus airborne viruses in relation to atopy and allergic asthma: epidemiological study, BMJ, vol.320, issue.7232, pp.412-419, 2000.

A. W. Graham, C. A. Rook, C. L. Lowry, and . Microbial, Old Friends', immunoregulation and stress resilience, Raison Evol Med Public Health, vol.2013, issue.1, pp.46-64, 2013.

T. R. -mosmann, H. Cherwinski, M. W. Bond, M. A. Giedlin, and R. L. Coffman, Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins, J Immunol, vol.136, issue.7, pp.2348-57, 1986.

B. Bosnjak, B. Stelzmueller, K. J. Erb, and M. M. Epstein, Treatment of allergic asthma: modulation of Th2 cells and their responses, Respir Res, vol.12, p.114, 2011.

T. B. Oriss, S. A. Mccarthy, B. F. Morel, M. A. Campana, and P. A. Morel, Crossregulation between T helper cell (Th)1 and Th2: inhibition of Th2 proliferation by IFN-gamma involves interference with IL-1, J Immunol, vol.158, issue.8, pp.3666-72, 1997.

L. Huang, A. M. Krieg, N. Eller, and D. E. Scott, Induction and regulation of Th1-inducing cytokines by bacterial DNA, lipopolysaccharide, and heat-inactivated bacteria, Infect Immun, vol.67, issue.12, pp.6257-63, 1999.

A. Graham and . Rook, Review series on helminths, immune modulation and the hygiene hypothesis: The broader implications of the hygiene hypothesis, Immunology, 2009.

R. M. Maizels, H. J. Mcsorley, and D. J. Smyth, 187-Rook GA, Brunet LR. Microbes, immunoregulation, and the gut, Clin Exp Immunol, vol.177, issue.1, pp.317-337, 2005.

N. Krug, J. Madden, A. E. Redington, P. Lackie, R. Djukanovic et al., T-cell cytokine profile evaluated at the single cell level in BAL and blood in allergic asthma, Am J Respir Cell Mol Biol, vol.14, issue.4, pp.319-345, 1996.

S. Klunker, A. Trautmann, M. Akdis, J. Verhagen, P. Schmid-grendelmeier et al., A second step of chemotaxis after transendothelial migration: keratinocytes undergoing apoptosis release IFN-gamma-inducible protein 10, monokine induced by IFN-gamma, and IFN-gamma-inducible alpha-chemoattractant for T cell chemotaxis toward epidermis in atopic dermatitis, J Immunol, vol.171, issue.2, pp.1078-84, 2003.

D. A. Lammas, J. L. Casanova, and D. S. Kumararatne, Clinical consequences of defects in the IL-12-dependent interferon-gamma (IFN-gamma) pathway, Clin Exp Immunol, vol.121, issue.3, pp.417-442, 2000.

G. Hansen, G. Berry, R. H. Dekruyff, D. T. Umetsu, G. Hansen et al., Allergen-specific Th1 cells fail to counterbalance Th2 cell-induced airway hyperreactivity but cause severe airway inflammation, J Clin Invest, vol.103, issue.2, pp.175-83, 1999.

M. Yazdanbakhsh, P. G. Kremsner, and R. Van-ree, Allergy, parasites, and the hygiene hypothesis, Science, vol.296, issue.5567, pp.490-494, 2002.

J. F. Bach, The effect of infections on susceptibility to autoimmune and allergic diseases, N Engl J Med, vol.347, issue.12, pp.911-931, 2002.

L. C. Stene and P. Nafstad, Relation between occurrence of type 1 diabetes and asthma, Lancet, vol.357, issue.9256, pp.607-615, 2001.

G. Rook, C. L-raison, and C. Lowry, Microbial 'old friends', immunoregulation and socioeconomic status, Clin Exp Immunol, vol.177, issue.1, pp.1-12, 2014.

M. Versini, P. Jeandel, T. Bashi, G. Bizzaro, M. Blank et al., Unraveling the Hygiene Hypothesis of helminthes and autoimmunity: origins, pathophysiology, and clinical applications, BMC Med, vol.13, p.81, 2015.

T. Dunder, T. Tapiainen, T. Pokka, and M. Uhari, Infections in child day care centers and later development of asthma, allergic rhinitis, and atopic dermatitis: prospective follow-up survey 12 years after controlled randomized hygiene intervention, Arch Pediatr Adolesc Med, vol.161, issue.10, pp.972-979, 2007.

J. Yoo, H. Tcheurekdjian, S. V. Lynch, M. Cabana, and H. A. Boushey, Microbial manipulation of immune function for asthma prevention: inferences from clinical trials, Proc Am Thorac Soc, vol.4, issue.3, pp.277-82, 2007.

N. A. Koloski, L. Bret, and G. Radford-smith, Hygiene hypothesis in inflammatory bowel disease: a critical review of the literature, World J Gastroenterol, vol.14, issue.2, pp.165-73, 2008.

C. R. Cardwell, D. J. Carson, J. Yarnell, M. D. Shields, and C. C. Patterson, Atopy, home environment and the risk of childhood-onset type 1 diabetes: a population-based case-control study, Pediatr Diabetes, vol.9, issue.3, pp.191-197, 2008.

D. K. Amre, P. Lambrette, L. Law, A. Krupoves, V. Chotard et al., Investigating the hygiene hypothesis as a risk factor in pediatric onset Crohn's disease: a case-control study, Am J Gastroenterol, vol.101, issue.5, pp.1005-1016, 2006.

C. N. Bernstein, P. Rawsthorne, M. Cheang, J. F. Blanchard, G. A. ;--rook et al., 204-Rook GA. 99th Dahlem conference on infection, inflammation and chronic inflammatory disorders: darwinian medicine and the 'hygiene' or 'old friends' hypothesis, .x. 205-von Hertzen L, Hanski I, Haahtela T. Natural immunity. Biodiversity loss and inflammatory diseases are two global megatrends that might be related. EMBO Rep, vol.101, pp.1089-93, 2003.

T. R. Mosmann and R. L. Coffman, TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties, Annu Rev Immunol, vol.7, pp.145-73, 1989.

F. Annunziato and S. Romagnani, Heterogeneity of human effector CD4+ T cells, Arthritis Res Ther, vol.11, issue.6, p.257, 2009.

W. C. Gause, J. F. Urban, and M. J. Stadecker, The immune response to parasitic helminths: insights from murine models, Trends Immunol, vol.24, issue.5, pp.269-77, 2003.

E. Bettelli, M. Oukka, and V. K. Kuchroo, T(H)-17 cells in the circle of immunity and autoimmunity, Nat Immunol, vol.8, issue.4, pp.345-50, 2007.

R. P. Singh, S. Hasan, S. Sharma, S. Nagra, D. T. Yamaguchi et al., Th17 cells in inflammation and autoimmunity, Autoimmun Rev, vol.13, issue.12, pp.1174-81, 2014.

S. Sakaguchi, Regulatory T cells: key controllers of immunologic self-tolerance, Cell, vol.101, issue.5, pp.455-463, 2000.

C. M. -hawrylowicz, O. Garra, and A. , Potential role of interleukin-10-secreting regulatory T cells in allergy and asthma, Nat Rev Immunol, vol.5, issue.4, pp.271-83, 2005.

J. M. Kim, J. P. Rasmussen, and A. Y. Rudensky, Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice, Nat Immunol, vol.8, issue.2, pp.191-198, 2007.

M. Akdis, J. Verhagen, A. Taylor, F. Karamloo, C. Karagiannidis et al., Immune responses in healthy and allergic individuals are characterized by a fine balance between allergen-specific T regulatory 1 and T helper 2 cells, S J Exp Med, vol.199, issue.11, pp.1567-75, 2004.

V. Viglietta, C. Baecher-allan, H. L. Weiner, and D. A. Hafler, Loss of functional suppression by CD4+CD25+ regulatory T cells in patients with multiple sclerosis, J Exp Med, vol.199, issue.7, pp.971-980, 2004.

T. A. Kraus, L. Toy, L. Chan, J. Childs, and L. Mayer, Failure to induce oral tolerance to a soluble protein in patients with inflammatory bowel disease, Gastroenterology, vol.126, issue.7, pp.1771-1779, 2004.

F. Powrie, S. Read, C. Mottet, H. Uhlig, and K. Maloy, Control of immune pathology by regulatory T cells, Novartis Found Symp, vol.252, pp.106-120, 2003.

D. T. Umetsu and R. H. Dekruyff, 99th Dahlem conference on infection, inflammation and chronic inflammatory disorders: microbes, apoptosis and TIM-1 in the development of asthma, Clin Exp Immunol, vol.160, issue.1, pp.125-134, 2010.

. Stoll, This wormy world, J. Parasitol, vol.33, pp.1-18, 1947.

N. W. Schröder, The role of innate immunity in the pathogenesis of asthma, Curr Opin Allergy Clin Immunol, vol.9, issue.1, pp.38-43, 2009.

, Out-of-Africa migration and Neolithic coexpansion of Mycobacterium tuberculosis with modern humans, Nat. Genet, 2013.

, An African origin for the intimate association between humans and Helicobacter pylori, Nature, vol.445, pp.915-918, 2007.

. 223-wolfe, Origins of major human infectious diseases Nature, vol.447, pp.279-283, 2007.

G. A. -rook, C. A. Lowry, and C. L. Raison, Hygiene and other early childhood influences on the subsequent function of the immune system, Brain Res, vol.1617, pp.47-62, 2015.

G. A. -rook, Hygiene hypothesis and autoimmune diseases, Clin Rev Allergy Immunol, vol.42, issue.1, pp.5-15, 2012.

G. A. -rook, C. L. Raison, and C. A. Lowry, Microbial 'old friends', immunoregulation and socioeconomic status, Clin Exp Immunol, vol.177, issue.1, pp.1-12, 2014.

L. Bert, N. Chain, B. M. Rook, G. Noursadeghi, and M. , DC priming by M. vaccae inhibits Th2 responses in contrast to specific TLR2 priming and is associated with selective activation of the CREB pathway, PLoS One, vol.6, issue.4, p.18346, 2011.

B. -linz, F. Balloux, Y. Moodley, A. Manica, H. Liu et al., An African origin for the intimate association between humans and Helicobacter pylori, Nature, vol.445, issue.7130, pp.915-918, 2007.

-. Mpairwe, E. L. Webb, L. Muhangi, J. Ndibazza, D. Akishule et al., Anthelminthic treatment during pregnancy is associated with increased risk of infantile eczema: randomised-controlled trial results, Pediatr Allergy Immunol, vol.22, issue.3, pp.305-312, 2011.

C. C. -obihara, N. Beyers, R. P. Gie, P. C. Potter, B. J. Marais et al., Inverse association between Mycobacterium tuberculosis infection and atopic rhinitis in children, Allergy, vol.60, issue.9, pp.1121-1126, 2005.

J. L. 231-round, S. M. Lee, J. Li, G. Tran, B. Jabri et al., The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota, Science, vol.332, issue.6032, pp.974-981, 2011.

J. R. Grainger, K. A. Smith, J. P. Hewitson, H. J. Mcsorley, Y. Harcus et al., Helminth secretions induce de novo T cell Foxp3 expression and regulatory function through the TGF-? pathway, J Exp Med, vol.207, issue.11, pp.2331-2372, 2010.

I. Hanski, L. Von-hertzen, N. Fyhrquist, K. Koskinen, K. Torppa et al., Environmental biodiversity, human microbiota, and allergy are interrelated, Proc Natl Acad Sci, vol.109, issue.21, pp.8334-8343, 2012.

S. Singhal, D. Dian, A. Keshavarzian, L. Fogg, J. Z. Fields et al., 235-Rook GA. Review series on helminths, immune modulation and the hygiene hypothesis: the broader implications of the hygiene hypothesis, Dig Dis Sci, vol.56, issue.1, pp.3-11, 2009.

Y. Osada and T. Kanazawa, Parasitic helminths: new weapons against immunological disorders, J Biomed Biotechnol, p.743758, 2010.

K. Karimi, M. D. Inman, J. Bienenstock, and P. Forsythe, Lactobacillus reuteri-induced regulatory T cells protect against an allergic airway response in mice, Am J Respir Crit Care Med, vol.179, issue.3, pp.186-93, 2009.

H. H. Smits, A. Engering, D. Van-der-kleij, E. C. De-jong, K. Schipper et al., Selective probiotic bacteria induce IL-10-producing regulatory T cells in vitro by modulating dendritic cell function through dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin, J Allergy Clin Immunol, vol.115, issue.6, pp.1260-1267, 2005.

J. Correale, M. Farez, and . Ann, Association between parasite infection and immune responses in multiple sclerosis, Neurol, vol.61, issue.2, pp.97-108, 2007.

J. Correale and M. F. Farez, The impact of parasite infections on the course of multiple sclerosis, J Neuroimmunol, vol.233, issue.1-2, pp.6-11, 2011.

J. O. Fleming, A. Isaak, J. E. Lee, C. C. Luzzio, M. D. Carrithers et al., Probiotic helminth administration in relapsing-remitting multiple sclerosis: a phase 1 study, Mult Scler, vol.17, issue.6, pp.743-54, 2011.

H. J. Mcsorley and R. M. Maizels, Helminth infections and host immune regulation, Clin Microbiol Rev, vol.25, issue.4, pp.585-608, 2012.

P. J. Hotez, P. J. Brindley, J. M. Bethony, C. H. King, E. J. Pearce et al., Helminth infections: the great neglected tropical diseases, J Clin Invest, vol.118, issue.4, pp.1311-1332, 2008.

K. Ziegelbauer, B. Speich, D. Mäusezahl, R. Bos, J. Keiser et al., Effect of sanitation on soil-transmitted helminth infection: systematic review and meta-analysis, PLoS Med, vol.9, issue.1, p.1001162, 2012.

S. Babu, C. P. Blauvelt, V. Kumaraswami, T. B. Nutman, and . Immunol, Regulatory networks induced by live parasites impair both Th1 and Th2 pathways in patent lymphatic filariasis: implications for parasite persistence, vol.176, pp.3248-56, 2006.

D. 246-van-der-kleij, E. Latz, J. F. Brouwers, Y. C. Kruize, M. Schmitz et al., A novel host-parasite lipid cross-talk. Schistosomal lyso-phosphatidylserine activates toll-like receptor 2 and affects immune polarization, 247-Weinstock JV, vol.277, pp.211-217, 2002.

D. B. Shor and Y. Shoenfeld, Autoimmunity: Will worms cure rheumatoid arthritis?, Nat Rev Rheumatol, vol.9, issue.3, pp.138-178, 2013.

H. Torres-aguilar, M. Blank, L. J. Jara, and Y. Shoenfeld, Tolerogenic dendritic cells in autoimmune diseases: crucial players in induction and prevention of autoimmunity, Autoimmun Rev, vol.10, issue.1, pp.8-17, 2010.

H. H. Smits, C. H. Hokke, and M. Yazdanbakhsh, Helminths and dendritic cells: sensing and regulating via pattern recognition receptors, Th2 and Treg responses. Everts B, Eur J Immunol, vol.40, issue.6, pp.1525-1562, 2010.

S. S. -diebold, Determination of T-cell fate by dendritic cells, Immunol Cell Biol, vol.86, issue.5, pp.389-97, 2008.

B. Pulendran, H. Tang, and S. Manicassamy, Programming dendritic cells to induce T(H)2 and tolerogenic responses, Nat Immunol, vol.11, issue.8, pp.647-55, 2010.

L. Hang, T. Setiawan, A. M. Blum, J. Urban, K. Stoyanoff et al., Heligmosomoides polygyrus infection can inhibit colitis through direct interaction with innate immunity, J Immunol, vol.185, issue.6, pp.3184-3193, 2010.

A. M. -blum, L. Hang, T. Setiawan, J. P. Urban, K. M. Stoyanoff et al., Heligmosomoides polygyrus bakeri induces tolerogenic dendritic cells that block colitis and prevent antigen-specific gut T cell responses, J Immunol, vol.189, issue.5, pp.2512-2532, 2012.

D. E. Elliott, A. Metwali, J. Leung, T. Setiawan, A. M. Blum et al., Colonization with Heligmosomoides polygyrus suppresses mucosal IL-17 production, J Immunol, vol.181, issue.4, pp.2414-2423, 2008.

Z. Wu, I. Nagano, K. Asano, and Y. Takahashi, Infection of non-encapsulated species of Trichinella ameliorates experimental autoimmune encephalomyelitis involving suppression of Th17 and Th1 response, Parasitol Res, vol.107, issue.5, pp.1173-88, 2010.

A. Gruden-movsesijan, N. Ilic, M. Mostarica-stojkovic, S. Stosic-grujicic, M. Milic et al., Mechanisms of modulation of experimental autoimmune encephalomyelitis by chronic Trichinella spiralis infection in Dark Agouti rats, Parasite Immunol, vol.32, issue.6, pp.450-459, 2010.

Y. -osada, S. Shimizu, T. Kumagai, S. Yamada, and T. Kanazawa, Schistosoma mansoni infection reduces severity of collagen-induced arthritis via down-regulation of proinflammatory mediators, Int J Parasitol, vol.39, issue.4, pp.457-64, 2009.

M. A. Pineda, M. A. Mcgrath, P. C. Smith, L. Al-riyami, J. Rzepecka et al., The parasitic helminth product ES-62 suppresses pathogenesis in collageninduced arthritis by targeting the interleukin-17-producing cellular network at multiple sites, Arthritis Rheum, vol.64, issue.10, pp.3168-78, 2012.

K. A. Saunders, T. Raine, A. Cooke, and C. E. Lawrence, Inhibition of autoimmune type 1 diabetes by gastrointestinal helminth infection. Infect Immun, vol.75, pp.397-407, 2007.

M. P. Hübner, J. T. Stocker, and E. Mitre, Inhibition of type 1 diabetes in filaria-infected nonobese diabetic mice is associated with a T helper type 2 shift and induction of FoxP3+ regulatory T cells, Immunology, vol.127, issue.4, pp.512-534, 2009.

P. Zaccone, O. Burton, N. Miller, F. M. Jones, D. W. Dunne et al., Schistosoma mansoni egg antigens induce Treg that participate in diabetes prevention in NOD mice, Eur J Immunol, vol.39, issue.4, pp.1098-107, 2009.

P. Zaccone, O. T. Burton, S. E. Gibbs, N. Miller, F. M. Jones et al., The S. mansoni glycoprotein ?-1 induces Foxp3 expression in NOD mouse CD4? T cells, Eur J Immunol, vol.41, issue.9, pp.2709-2727, 2011.
URL : https://hal.archives-ouvertes.fr/in2p3-00021182

I. B. Mcinnes, B. P. Leung, M. Harnett, J. A. Gracie, F. Y. Liew et al., A novel therapeutic approach targeting articular inflammation using the filarial nematode-derived phosphorylcholine-containing glycoprotein ES-62, J Immunol, vol.171, issue.4, pp.2127-2160, 2003.

I. Kalampokis, A. Yoshizaki, and T. F. Tedder, IL-10-producing regulatory B cells (B10 cells) in autoimmune disease, Arthritis Res Ther, vol.15, 2013.

M. Yang, K. Rui, S. Wang, and L. Lu, Regulatory B cells in autoimmune diseases, Cell Mol Immunol, vol.10, issue.2, pp.122-154, 2013.

A. R. Khan, S. Amu, S. P. Saunders, and P. G. Fallon, The generation of regulatory B cells by helminth parasites, Methods Mol Biol, vol.1190, pp.143-62, 2014.

L. E. 269-van-der-vlugt, J. F. Zinsou, A. Ozir-fazalalikhan, P. G. Kremsner, M. Yazdanbakhsh et al., Protection against collagen-induced arthritis in mice afforded by the parasitic worm product, ES-62, is associated with restoration of the levels of interleukin-10-producing B cells and reduced plasma cell infiltration of the joints, J Infect Dis, vol.210, issue.8, pp.457-66, 2014.

J. Correale, M. Farez, and G. Razzitte, Helminth infections associated with multiple sclerosis induce regulatory B cells, Ann Neurol, vol.64, issue.2, pp.187-99, 2008.

A. -sica and A. Mantovani, Macrophage plasticity and polarization: in vivo veritas, J Clin Invest, vol.122, issue.3, pp.787-95, 2012.

D. R. Herbert, C. Hölscher, M. Mohrs, B. Arendse, A. Schwegmann et al., Alternative macrophage activation is essential for survival during schistosomiasis and downmodulates T helper 1 responses and immunopathology, Immunity, vol.20, issue.5, pp.623-658, 2004.

A. H. Liu, Revisiting the hygiene hypothesis for allergy and asthma, J Allergy Clin Immunol, vol.136, issue.4, pp.860-865, 2015.

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.

S. V. -lynch, R. A. Wood, H. Boushey, L. B. Bacharier, G. R. Bloomberg et al., Effects of early-life exposure to allergens and bacteria on recurrent wheeze and atopy in urban children, J Allergy Clin Immunol, vol.134, issue.3, pp.593-601, 2014.

H. Bisgaard, M. N. Hermansen, F. Buchvald, L. Loland, L. B. Halkjaer et al., Childhood asthma after bacterial colonization of the airway in neonates, N Engl J Med, vol.357, issue.15, pp.1487-95, 2007.

T. C. -scharschmidt, Establishing Tolerance to Commensal Skin Bacteria: Timing Is Everything, Dermatol Clin, vol.35, issue.1, pp.1-9, 2017.

H. H. Smits and C. A. Akdis, In utero priming by worms protects against respiratory allergies, J Allergy Clin Immunol, vol.134, issue.6, pp.1280-1281, 2014.

J. Philip, M. E. Cooper, I. Chico, . Guadalupe, A. Carlos et al., Impact of early life exposures to geohelminth infections on the development of vaccine immunity, allergic sensitization, and allergic inflammatory diseases in children living in tropical Ecuador: the ECUAVIDA birth cohort study, BMC Infect Dis, vol.11, p.184, 2011.

P. J. Cooper, M. L. Barreto, and L. C. Rodrigues, Human allergy and geohelminth infections: a review of the literature and a proposed conceptual model to guide the investigation of possible causal associations, Br Med Bull, pp.203-221, 2006.

L. C. Rodrigues, P. J. Newcombe, S. S. Cunha, N. M. Alcantara-neves, B. Genser et al., Early infection with Trichuris trichiura and allergen skin test reactivity in later childhood, Clin Exp Allergy, vol.38, issue.11, pp.1769-77, 2008.

C. A. -figueiredo, N. M. Alcantara-neves, L. D. Amorim, N. B. Silva, L. C. Carvalho et al., Evidence for a modulatory effect of IL-10 on both Th1 and Th2 cytokine production: the role of the environment, Clin Immunol, vol.139, issue.1, pp.57-64, 2011.

J. D. Turner, J. A. Jackson, H. Faulkner, J. Behnke, K. J. Else et al., Intensity of intestinal infection with multiple worm species is related to regulatory cytokine output and immune hyporesponsiveness, J Infect Dis, vol.197, issue.8, pp.1204-1216, 2008.

C. A. -figueiredo, N. M. Alcântara-neves, R. Veiga, L. D. Amorim, V. Dattoli et al., Spontaneous cytokine production in children according to biological characteristics and environmental exposures. Environ Health Perspect, Infect Immun, vol.117, issue.5, pp.3160-3167, 2009.

P. J. Cooper, Interactions between helminth parasites and allergy, Curr Opin Allergy Clin Immunol, vol.9, issue.1, pp.29-37, 2009.

B. Kampmann, S. K. Mazmanian, A. Marchant, and O. Levy, Protecting the Newborn and Young Infant from Infectious Diseases: Lessons from Immune Ontogeny, vol.46, pp.350-363, 2017.

N. Wood and C. A. Siegrist, Neonatal immunization: where do we stand?, Curr Opin Infect Dis, vol.24, issue.3, pp.190-195, 2011.

A. Saso and B. Kampmann, Vaccine responses in newborns Semin Immunopathol, vol.39, pp.627-642, 2017.

H. Dunand, P. C. Noel, T. Pauli, C. J. Wilson-;--colditz, G. A. Brewer et al., Efficacy of BCG vaccine in the prevention of tuberculosis. Meta-analysis of the published literature, Goldhaber-Fiebert JD, vol.2, p.13842, 1994.

M. M. 295-van-den-ent, D. W. Brown, E. J. Hoekstra, A. Christie, and S. L. Cochi, Measles mortality reduction contributes substantially to reduction of all cause mortality among children less than five years of age, J Infect Dis, issue.1, pp.18-23, 0204.

M. A. Koenig, N. C. Roy, T. Mcelrath, M. Shahidullah, and B. Wojtyniak, Duration of protective immunity conferred by maternal tetanus toxoid immunization: further evidence from Matlab, Bangladesh, Am J Public Health, vol.88, issue.6, pp.903-910, 1998.

D. R. -feikin, B. Flannery, and M. J. Hamel, A novel in vivo follicular dendritic cell-dependent iccosome-mediated mechanism for delivery of antigen to antigen-processing cells, ) Recommandations vaccinales et rappels chez l'enfants, vol.140, pp.341-53, 1988.

T. -junt, E. A. Moseman, M. Iannacone, S. Massberg, P. A. Lang et al., Subcapsular sinus macrophages in lymph nodes clear lymph-borne viruses and present them to antiviral B cells, Nature, vol.450, issue.7166, pp.110-114, 2007.

T. G. -phan, J. A. Green, E. E. Gray, Y. Xu, and J. G. Cyster, Immune complex relay by subcapsular sinus macrophages and noncognate B cells drives antibody affinity maturation, Nat Immunol, vol.10, issue.7, pp.786-93, 2009.

A. -lanzavecchia, Receptor-mediated antigen uptake and its effect on antigen presentation to class II-restricted T lymphocytes, Annu Rev Immunol, vol.8, pp.773-93, 1990.

T. Okada, M. J. Miller, I. Parker, M. F. Krummel, M. Neighbors et al., Antigenengaged B cells undergo chemotaxis toward the T zone and form motile conjugates with helper T cells, PLoS Biol, vol.3, issue.6, p.150, 2005.

N. M. Haynes, C. D. Allen, L. R. Ansel, K. M. Killeen, N. Cyster et al., Role of CXCR5 and CCR7 in follicular Th cell positioning and appearance of a programmed cell death gene-1high germinal center-associated subpopulation, J Immunol, vol.179, issue.8, pp.5099-108, 2007.

J. 306-banchereau and F. Rousset, Growing human B lymphocytes in the CD40 system, Nature, vol.353, issue.6345, pp.678-687, 1991.

A. I. Jaiswal and M. Croft, CD40 ligand induction on T cell subsets by peptide-presenting B cells: implications for development of the primary T and B cell response, J Immunol, vol.159, issue.5, pp.2282-91, 1997.

Y. Liu, J. Zhang, P. J. Lane, E. Y. Chan, and I. C. Maclennan, Sites of specific B cell activation in primary and secondary responses to T cell-dependent and T cell-independent antigens, Eur J Immunol, vol.21, issue.12, pp.2951-62, 1991.

-. Dw, Allergy and immunology, Annu Rev Med, vol.8, pp.239-56, 1957.

M. G. Weigert, I. M. Cesari, S. J. Yonkovich, and M. Cohn, Variability in the lambda light chain sequences of mouse antibody, Nature, vol.228, issue.5276, pp.1045-1052, 1970.

M. Muramatsu, K. Kinoshita, S. Fagarasan, S. Yamada, Y. Shinkai et al., Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme, Cell, vol.102, issue.5, pp.553-63, 2000.

P. Revy, T. Muto, Y. Levy, F. Geissmann, A. Plebani et al., Activation-induced cytidine deaminase (AID) deficiency causes the autosomal recessive form of the Hyper-IgM syndrome (HIGM2), Cell, vol.102, issue.5, pp.565-75, 2000.

P. W. Kincade, A. R. Lawton, D. E. Bockman, and M. D. Cooper, Suppression of immunoglobulin G synthesis as a result of antibody-mediated suppression of immunoglobulin M synthesis in chickens, Proc Natl Acad Sci, vol.67, issue.4, pp.1918-1943, 1970.

C. M. Snapper and W. E. Paul, Interferon-gamma and B cell stimulatory factor-1 reciprocally regulate Ig isotype production, Science, vol.236, issue.4804, pp.944-951, 1987.

P. Nieuwenhuis and D. Opstelten, Functional anatomy of germinal centers, Am J Anat, vol.170, issue.3, pp.421-456, 1984.

G. D. -victora, T. A. Schwickert, D. R. Fooksman, A. O. Kamphorst, M. Meyer-hermann et al., Germinal center dynamics revealed by multiphoton microscopy with a photoactivatable fluorescent reporter, Cell, vol.143, issue.4, pp.592-605, 2010.

Y. J. Liu, D. E. Joshua, G. T. Williams, C. A. Smith, J. Gordon et al., Germinal center cells express bcl-2 protein after activation by signals which prevent their entry into apoptosis, Eur J Immunol, vol.342, issue.6252, pp.1243-52, 1989.

C. Arpin, J. Déchanet, C. Van-kooten, P. Merville, G. Grouard et al., Generation of memory B cells and plasma cells in vitro, Science, vol.268, issue.5211, pp.720-722, 1995.

N. L. -bernasconi, E. Traggiai, and A. Lanzavecchia, Maintenance of serological memory by polyclonal activation of human memory B cells. Science, vol.298, pp.2199-202, 2002.

I. Dogan, B. Bertocci, V. Vilmont, F. Delbos, J. Mégret et al., Multiple layers of B cell memory with different effector functions, Nat Immunol, vol.10, issue.12, pp.1292-1301, 2009.

K. A. Pape, J. J. Taylor, R. W. Maul, P. J. Gearhart, and M. K. Jenkins, Different B cell populations mediate early and late memory during an endogenous immune response, Science, vol.331, issue.6021, pp.1203-1210, 2011.

V. Ilja, S. Khavrutskii, S. M. Chaudhury, D. W. Stronsky, J. G. Lee et al., Quantitative Analysis of Repertoire-Scale Immunoglobulin Properties in Vaccine-Induced B-Cell Responses, Front Immunol, vol.8, p.910, 2017.

L. J. Mcheyzer-williams and M. Mg, Antigen-specific memory B cell development, Annu Rev Immunol, vol.23, pp.487-513, 2005.

I. C. Maclennan, K. M. Toellner, A. F. Cunningham, K. Serre, D. M. Sze et al., Extrafollicular antibody responses, Immunol Rev, vol.194, pp.8-18, 2003.

C. -goodnow, J. Sprent, F. De-st-groth, B. Vinuesa, and C. G. , Cellular and genetic mechanisms of self tolerance and autoimmunity, Nature, vol.435, issue.7042, pp.590-597, 2005.

T. G. Phan, E. E. Gray, and J. G. Cyster, The microanatomy of B cell activation, Curr Opin Immunol, vol.21, issue.3, pp.258-65, 2009.

S. Crotty, Follicular helper CD4 T cells (TFH), Annu Rev Immunol, vol.29, pp.621-63, 2011.

R. Gomez, M. Talke, Y. Goebel, N. Hermann, F. Reich et al., Activation of virus-specific memory B cells in the absence of T cell help, J Immunol, vol.185, issue.12, pp.593-602, 2004.

N. Engels, L. M. König, C. Heemann, J. Lutz, T. Tsubata et al., Recruitment of the cytoplasmic adaptor Grb2 to surface IgG and IgE provides antigen receptor-intrinsic costimulation to class-switched B cells, Nat Immunol, vol.10, issue.9, pp.1018-1043, 2009.

-. Castellino, G. Galli, G. D. Giudice, R. Rappuoli, C. L. Trotter et al., Optimising the use of conjugate vaccines to prevent disease caused by Haemophilus influenzae type b, Neisseria meningitidis and Streptococcus pneumoniae. Vaccine, European Journal of ImmunologyVolume, vol.39, issue.35, pp.4434-4479, 2008.

R. -borrow, E. Miller, E. A. ;--clutterbuck, S. Oh, M. Hamaluba et al., Serotypespecific and age-dependent generation of pneumococcal polysaccharide-specific memory B-cell and antibody responses to immunization with a pneumococcal conjugate vaccine, Clin Vaccine Immunol, vol.5, issue.6, pp.182-93, 2006.

D. F. Kelly, A. J. Pollard, and E. R. Moxon, Immunological memory: the role of B cells in longterm protection against invasive bacterial pathogens, JAMA, vol.294, issue.23, pp.3019-3042, 2005.

C. L. Trotter, N. J. Andrews, E. B. Kaczmarski, E. Miller, and M. E. Ramsay, Effectiveness of meningococcal serogroup C conjugate vaccine 4 years after introduction, Lancet, vol.364, issue.9431, pp.365-372, 2004.

H. Nakajima, H. Kariya, R. Ohata, and H. Ogura, Investigation of immunity level against diphtheria and reinforcement of immunity by booster vaccination for infection control staff in Okayama prefecture, Jpn J Infect Dis, vol.61, issue.2, pp.104-110, 2008.

D. P. Francis, S. C. Hadler, S. E. Thompson, J. E. Maynard, D. G. Ostrow et al., The prevention of hepatitis B with vaccine. Report of the centers for disease control multicenter efficacy trial among homosexual men, Ann Intern Med, vol.97, issue.3, pp.362-368, 1982.

E. Leuridan and P. Van-damme, Hepatitis B and the need for a booster dose, Clin Infect Dis, vol.53, issue.1, pp.68-75, 2011.

W. -jilg, M. Schmidt, and F. Deinhardt, Vaccination against hepatitis B: comparison of three different vaccination schedules, J Infect Dis, vol.160, issue.5, pp.766-775, 1989.

J. P. Ting, J. A. Duncan, and Y. Lei, The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors, Nat Immunol, vol.11, issue.5, pp.286-90, 2010.

C. Wilkins and M. Gale, Recognition of viruses by cytoplasmic sensors, Curr Opin Immunol, vol.22, issue.1, pp.41-48, 2010.

B. Pulendran and R. Ahmed, Translating innate immunity into immunological memory: implications for vaccine development, Cell, vol.124, issue.4, pp.849-63, 2006.

R. M. Steinman, Dendritic cells in vivo: a key target for a new vaccine science. Immunity, vol.29, pp.319-343, 2008.

K. A. Heldwein, M. D. Liang, T. K. Andresen, K. E. Thomas, A. M. Marty et al., TLR2 and TLR4 serve distinct roles in the host immune response against Mycobacterium bovis BCG, J Leukoc Biol, vol.74, issue.2, pp.277-86, 2003.

A. P. Soares, K. Chung, C. K. Choice, T. Hughes, E. J. Jacobs et al., Longitudinal changes in CD4(+) T-cell memory responses induced by BCG vaccination of newborns, J Infect Dis, vol.207, issue.7, 2013.

, Humoral and cellular immunity to varicella-zoster virus: an overview, J Infect Dis, issue.2, pp.58-60, 0197.

B. Pulendran and R. Ahmed, Immunological mechanisms of vaccination, Nat Immunol, vol.12, issue.6, pp.509-517, 2011.

G. A. -kolumam, S. Thomas, L. J. Thompson, J. Sprent, and K. Murali-krishna, Type I interferons act directly on CD8 T cells to allow clonal expansion and memory formation in response to viral infection, J Exp Med, vol.202, issue.5, pp.637-50, 2005.

C. Pasare and R. Medzhitov, Toll pathway-dependent blockade of CD4+CD25+ T cellmediated suppression by dendritic cells, Science, vol.299, issue.5609, pp.1033-1039, 2003.

B. Pulendran, J. L. Smith, G. Caspary, K. Brasel, D. Pettit et al., Distinct dendritic cell subsets differentially regulate the class of immune response in vivo, 370-den Haan JM, Lehar SM, Bevan MJ, vol.96, pp.1685-96, 1999.

F. Sallusto, A. Lanzavecchia, K. Araki, and R. Ahmed, From vaccines to memory and back. Immunity, vol.33, pp.451-63, 2010.

S. C. Jameson and D. Masopust, Diversity in T cell memory: an embarrassment of riches, Immunity, vol.31, issue.6, pp.859-71, 2009.

S. M. -kaech, E. J. Wherry, S. A. Plotkin, O. Wa, P. A. Offit et al., Heterogeneity and cell-fate decisions in effector and memory CD8+ T cell differentiation during viral infection, Immunity, vol.27, issue.3, pp.393-405, 2007.

S. Crotty, P. Felgner, H. Davies, J. Glidewell, L. Villarreal et al., Cutting edge: long-term B cell memory in humans after smallpox vaccination, J Immunol, vol.171, issue.10, pp.4969-73, 2003.

E. Hammarlund, M. W. Lewis, S. G. Hansen, L. I. Strelow, J. A. Nelson et al., Duration of antiviral immunity after smallpox vaccination, Nat Med, vol.9, issue.9, pp.1131-1138, 2003.

T. D. -querec, R. S. Akondy, E. K. Lee, W. Cao, H. I. Nakaya et al., Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans, Nat Immunol, vol.10, issue.1, pp.116-125, 2009.

M. L. -precopio, M. R. Betts, J. Parrino, D. A. Price, E. Gostick et al., Immunization with vaccinia virus induces polyfunctional and phenotypically distinctive CD8(+) T cell responses, J Exp Med, vol.204, issue.6, pp.1405-1421, 2007.

J. Zhu, J. Martinez, X. Huang, and Y. Yang, Innate immunity against vaccinia virus is mediated by TLR2 and requires TLR-independent production of IFN-beta. Blood, vol.109, pp.619-644, 2007.

T. Querec, S. Bennouna, S. Alkan, Y. Laouar, K. Gorden et al., Yellow fever vaccine YF-17D activates multiple dendritic cell subsets via TLR2, 7, 8, and 9 to stimulate polyvalent immunity, J Exp Med, vol.203, issue.2, pp.413-437, 2006.

A. J. Pollard, K. P. Perrett, and P. C. Beverley, Maintaining protection against invasive bacteria with protein-polysaccharide conjugate vaccines, Nat Rev Immunol, vol.9, issue.3, pp.213-233, 2009.

G. Blanchard-rohner, M. D. Snape, D. F. Kelly, T. John, A. Morant et al., The magnitude of the antibody and memory B cell responses during priming with a protein-polysaccharide conjugate vaccine in human infants is associated with the persistence of antibody and the intensity of booster response, vol.180, pp.2165-73, 2008.

E. Mohr and C. A. Siegrist, Vaccination in early life: standing up to the challenges, Curr Opin Immunol, vol.41, pp.1-8, 2016.

D. J. Dowling and O. Levy, Ontogeny of early life immunity, Trends Immunol, vol.35, issue.7, pp.299-310, 2014.

S. Fadel and M. Sarzotti, Cellular immune responses in neonates, Int Rev Immunol, vol.19, issue.2-3, pp.173-93, 2000.

A. Marchant and M. Goldman, T cell-mediated immune responses in human newborns: ready to learn?, Clin Exp Immunol, vol.141, issue.1, pp.10-18, 2005.

A. Demirjian and O. Levy, Safety and efficacy of neonatal vaccination, Eur J Immunol

, , vol.39, pp.36-46

S. Basha, N. Surendran, and M. Pichichero, Immune responses in neonates, Expert Rev Clin Immunol, vol.10, issue.9, pp.1171-84, 2014.

I. Debock and V. Flamand, Unbalanced Neonatal CD4(+) T-Cell Immunity. Front Immunol, vol.5, p.393, 2014.

M. -prabhudas, B. Adkins, H. Gans, C. King, O. Levy et al., Challenges in infant immunity: implications for responses to infection and vaccines, Nat Immunol, vol.12, issue.3, pp.189-94, 2011.

C. A. -siegrist, The challenges of vaccine responses in early life: selected examples, J Comp Pathol, vol.137, issue.1, pp.4-9, 2007.

O. Levy, Innate immunity of the newborn: basic mechanisms and clinical correlates, Nat Rev Immunol, vol.7, issue.5, pp.379-90, 2007.

M. E. -belderbos, O. Levy, L. Meyaard, and L. Bont, Plasma-mediated immune suppression: a neonatal perspective, Pediatr Allergy Immunol, vol.24, issue.2, pp.102-115, 2013.

-. Levy, M. Coughlin, B. N. Cronstein, R. M. Roy, A. Desai et al., The Adenosine System Selectively Inhibits TLR-Mediated TNF-? Production in the Human Newborn, J Immunol, vol.177, issue.3, pp.1956-1966, 2006.

D. Krumbiegel, F. Zepp, and C. U. Meyer, Combined Toll-like receptor agonists synergistically increase production of inflammatory cytokines in human neonatal dendritic cells, Hum Immunol, vol.68, issue.10, pp.813-835, 2007.

P. J. Fink, The biology of recent thymic emigrants, Annu Rev Immunol, vol.31, pp.31-50, 2012.

C. J. Haines, T. D. Giffon, L. S. Lu, X. Lu, M. Tessier-lavigne et al., Human CD4+ T cell recent thymic emigrants are identified by protein tyrosine kinase 7 and have reduced immune function, J Exp Med, vol.206, issue.2, pp.275-85, 2009.

H. Zaghouani, C. M. Hoeman, and B. Adkins, Neonatal immunity: faulty T-helpers and the shortcomings of dendritic cells, Trends Immunol, vol.30, issue.12, pp.585-91, 2009.

M. C. Morris and N. Surendran, Neonatal Vaccination: Challenges and Intervention Strategies, Neonatology, vol.109, issue.3, pp.161-170, 2016.

M. C. Boer, S. Joosten, and T. H. Ottenhoff, Regulatory T-Cells at the Interface between Human Host and Pathogens in Infectious Diseases and Vaccination. Front Immunol, vol.6, p.217, 2015.

T. D. Burt, Fetal regulatory T cells and peripheral immune tolerance in utero: implications for development and disease, Am J Reprod Immunol, vol.69, issue.4, pp.346-58, 2013.

C. A. -siegrist and R. Aspinall, B-cell responses to vaccination at the extremes of age, Nat Rev Immunol, vol.9, issue.3, pp.185-94, 2009.

I. Debock, K. Jaworski, H. Chadlaoui, S. Delbauve, N. Passon et al., Neonatal follicular Th cell responses are impaired and modulated by IL, J Immunol, vol.191, issue.3, pp.1231-1240, 2013.

M. Pihlgren, C. Tougne, P. Bozzotti, A. Fulurija, M. A. Duchosal et al., Unresponsiveness to lymphoid-mediated signals at the neonatal follicular dendritic cell precursor level contributes to delayed germinal center induction and limitations of neonatal antibody responses to T-dependent antigens, J Immunol, vol.170, issue.6, pp.2824-2856, 2003.

M. O. Ota, J. Vekemans, S. E. Schlegel-haueter, K. Fielding, H. Whittle et al., Hepatitis B immunisation induces higher antibody and memory Th2 responses in new-borns than in adults, Vaccine, vol.22, issue.3-4, pp.511-520, 2004.

J. Vekemans, M. O. Ota, E. C. Wang, M. Kidd, L. K. Borysiewicz et al., T cell responses to vaccines in infants: defective IFNgamma production after oral polio vaccination, Clin Exp Immunol, vol.127, issue.3, pp.495-503, 2002.

M. O. Ota, J. Vekemans, S. E. Schlegel-haueter, K. Fielding, M. Sanneh et al., Influence of Mycobacterium bovis bacillus Calmette-Guérin on antibody and cytokine responses to human neonatal vaccination, J Immunol, vol.168, issue.2, pp.919-944, 2002.

S. Cobey, P. Wilson, and F. A. Matsen, The evolution within us, Philos Trans R Soc Lond B Biol Sci, vol.370, 1676.

K. B. Hoehn, A. Fowler, G. Lunter, and O. G. Pybus, The Diversity and Molecular Evolution of B-Cell Receptors during Infection, Mol Biol Evol, vol.33, issue.5, pp.1147-57, 2016.

V. Greiff, P. Bhat, S. C. Cook, U. Menzel, W. Kang et al., A bioinformatic framework for immune repertoire diversity profiling enables detection of immunological status, Genome Med, vol.7, issue.1, p.49, 2015.

R. N. Germain, Vaccines and the future of human immunology, Immunity, vol.33, issue.4, pp.441-50, 2010.

R. Rappuoli, Bridging the knowledge gaps in vaccine design, Nat Biotechnol, vol.25, issue.12, pp.1361-1367, 2007.

B. Pulendran and R. Ahmed, Immunological mechanisms of vaccination, Nat Immunol, vol.12, issue.6, pp.509-526, 2011.

M. Kuraoka, A. G. Schmidt, T. Nojima, F. Feng, A. Watanabe et al., Complex Antigens Drive Permissive Clonal Selection in Germinal Centers, Immunity, vol.44, issue.3, pp.542-552, 2016.

J. J. Lavinder, Y. Wine, C. Giesecke, G. C. Ippolito, A. P. Horton et al., Identification and characterization of the constituent human serum antibodies elicited by vaccination, Proc Natl Acad Sci, vol.111, issue.6, pp.2259-64, 2014.

E. Mark, D. Mcgovern, and . Canning, Vaccination and All-Cause Child Mortality From 1985 to 2011: Global Evidence From the Demographic and Health Surveys, Am J Epidemiol, vol.182, issue.9, pp.791-798, 2015.

P. Aaby, J. Bukh, I. M. Lisse, and . Smits, Measles vaccination and reduction in child mortality: a community study from Guinea-Bissau, AJ J Infect, vol.8, issue.1, pp.13-21, 1984.

H. S. Goodridge, S. Ahmed, N. Curtis, T. R. Kollmann, and O. Levy, Harnessing the beneficial heterologous effects of vaccination, Nat Rev Immunol, vol.16, issue.6, pp.392-400, 2016.

M. J. De-castro, J. Pardo-seco, F. Martinón-torres, and . Nonspecific, Heterologous) Protection of Neonatal BCG Vaccination Against Hospitalization Due to Respiratory Infection and Sepsis, Clin Infect Dis, vol.60, issue.11, pp.1611-1620, 2015.

S. Sørup, C. S. Benn, A. Poulsen, T. G. Krause, P. Aaby et al., Live vaccine against measles, mumps, and rubella and the risk of hospital admissions for nontargeted infections, vol.311, pp.826-861, 2014.

S. Sørup, M. Villumsen, H. Ravn, C. S. Benn, T. I. Sørensen et al., Smallpox vaccination and all-cause infectious disease hospitalization: a Danish registerbased cohort study, Int J Epidemiol, vol.40, issue.4, pp.955-63, 2011.

S. -sørup, L. G. Stensballe, T. G. Krause, P. Aaby, C. S. Benn et al., Oral Polio Vaccination and Hospital Admissions With Non-Polio Infections in Denmark: Nationwide Retrospective Cohort Study. Open Forum Infect Dis, vol.3, p.204, 2015.

P. Aaby, T. R. Kollmann, and C. S. Benn, Nonspecific effects of neonatal and infant vaccination: public-health, immunological and conceptual challenges, Nat Immunol, vol.15, issue.10, pp.895-904, 2014.

K. L. Flanagan, R. Van-crevel, N. Curtis, F. Shann, and L. O. Heterologous, nonspecific") and sex-differential effects of vaccines: epidemiology, clinical trials, and emerging immunologic mechanisms, Clin Infect Dis, vol.57, issue.2, pp.283-292, 2013.

H. S. -goodridge, S. S. Ahmed, N. Curtis, T. R. Kollmann, O. Levy et al., Harnessing the beneficial heterologous effects of vaccination, -Meeting of the Strategic Advisory Group of Experts on immunization, vol.89, pp.392-400, 2014.

N. Lund, A. Andersen, A. S. Hansen, F. S. Jepsen, A. Barbosa et al., The Effect of Oral Polio Vaccine at Birth on Infant Mortality: A Randomized Trial, Clin Infect Dis, vol.61, issue.10, pp.1504-1515, 2015.

S. Khurana, N. Verma, J. W. Yewdell, A. K. Hilbert, F. Castellino et al., MF59 adjuvant enhances diversity and affinity of antibody-mediated immune response to pandemic influenza vaccines, Sci Transl Med, vol.3, issue.85, pp.85-133, 2011.

M. A. Koenig, M. A. Khan, B. Wojtyniak, J. D. Clemens, J. Chakraborty et al., Impact of measles vaccination on childhood mortality in rural Bangladesh, Bull World Health Organ, vol.68, issue.4, pp.441-448, 1990.

P. Aaby, H. Jensen, A. Rodrigues, M. L. Garly, C. S. Benn et al., Divergent female-male mortality ratios associated with different routine vaccinations among female-male twin pairs, Int J Epidemiol, vol.33, issue.2, pp.367-73, 2004.

M. G. -netea, R. Van-crevel, and . Semin-immunol, BCG-induced protection: effects on innate immune memory, vol.26, pp.512-519, 2014.

J. Kleinnijenhuis, R. Van-crevel, and M. G. Netea, Trained immunity: consequences for the heterologous effects of BCG vaccination, Trans R Soc Trop Med Hyg, vol.109, issue.1, pp.29-35, 2015.

J. Kleinnijenhuis, J. Quintin, F. Preijers, C. S. Benn, L. A. Joosten et al., Longlasting effects of BCG vaccination on both heterologous Th1/Th17 responses and innate trained immunity, J Innate Immun, vol.6, issue.2, pp.152-160, 2014.

D. H. -libraty, L. Zhang, M. Woda, L. P. Acosta, A. Obcena et al., Neonatal BCG vaccination is associated with enhanced T-helper 1 immune responses to heterologous infant vaccines, Capeding RZTrials Vaccinol, vol.3, pp.1-5, 2014.

J. Kleinnijenhuis, J. Quintin, F. Preijers, L. A. Joosten, D. C. Ifrim et al., Bacille Calmette-Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes, Proc Natl Acad Sci, vol.109, issue.43, pp.17537-17579, 2012.

Q. Li and H. H. Shen, Neonatal bacillus Calmette-Guérin vaccination inhibits de novo allergic inflammatory response in mice via alteration of CD4+CD25+ T-regulatory cells, Acta Pharmacol Sin, vol.30, issue.1, pp.125-158, 2009.

N. Curtis, Potential role for BCG in treatment of autoimmune diseases, BMJ, vol.355, p.6091, 2016.

M. C. Rousseau, M. E. Parent, and Y. St-pierre, Potential health effects from non-specific stimulation of the immune function in early age: the example of BCG vaccination, Pediatr Allergy Immunol, vol.19, issue.5, pp.438-486, 2008.

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

M. Fulde and M. W. Hornef, Maturation of the enteric mucosal innate immune system during the postnatal period, Immunol Rev, vol.260, issue.1, pp.21-34, 2014.

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

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

M. -gomez-de-agüero, S. C. Ganal-vonarburg, T. Fuhrer, S. Rupp, Y. Uchimura et al., The maternal microbiota drives early postnatal innate immune development, Science, vol.351, issue.6279, pp.1296-302, 2016.

S. Rautava, M. C. Collado, S. Salminen, and . Isolauri, Probiotics modulate host-microbe interaction in the placenta and fetal gut: a randomized, double-blind, placebo-controlled trial, ENeonatology, vol.102, issue.3, pp.178-84, 2012.

S. L. Prescott, K. Wickens, L. Westcott, W. Jung, H. Currie et al., Supplementation with Lactobacillus rhamnosus or Bifidobacterium lactis probiotics in pregnancy increases cord blood interferon-gamma and breast milk transforming growth factor-beta and immunoglobin A detection, 446-Jakobsson HE, vol.38, pp.559-66, 2008.

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.

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, vol.17, issue.5, pp.690-703, 2015.

M. Jay, Huilin Li. Antibiotics, birth mode, and diet shape microbiome maturation during early life, Sci Transl Med, vol.1, issue.2, pp.343-82, 2016.

J. -romano-keeler and J. H. Weitkamp, Maternal influences on fetal microbial colonization and immune development, Pediatr Res, vol.77, issue.1-2, pp.189-95, 2015.

C. H. Hansen, S. B. Metzdorff, and A. K. Hansen, Customizing laboratory mice by modifying gut microbiota and host immunity in an early "window of opportunity, Gut Microbes

K. M. Bendtsen, L. Fisker, A. K. Hansen, C. H. Hansen, and D. S. Nielsen, The influence of the young microbiome on inflammatory diseases--Lessons from animal studies, Birth Defects Res C Embryo Today, vol.105, issue.4, pp.278-95, 2015.

L. M. Cox, S. Yamanishi, J. Sohn, A. V. Alekseyenko, J. M. Leung et al., Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences, Cell, vol.158, issue.4, pp.705-721, 2014.

H. Chu and S. K. Mazmanian, Innate immune recognition of the microbiota promotes hostmicrobial symbiosis, Nat Immunol, vol.14, issue.7, pp.668-75, 2013.

A. Khosravi, A. Yáñez, J. G. Price, A. Chow, M. Merad et al., Gut microbiota promote hematopoiesis to control bacterial infection, Cell Host Microbe, vol.15, issue.3, pp.374-81, 2014.

M. G. Dominguez-bello and M. J. Blaser, Asthma: Undoing millions of years of coevolution in early life?, Sci Transl Med, vol.7, issue.307, pp.307-346, 2015.

T. Lavender, G. J. Hofmeyr, J. P. Neilson, C. Kingdon, and G. M. Gyte, High caesarean rates in Madras (India): a population-based cross sectional study, CD004660. 458-Sreevidya S, Sathiyasekaran BW, vol.110, pp.106-117, 2003.

W. J. Ledger and M. J. Blaser, Are we using too many antibiotics during pregnancy? BJOG, vol.120, pp.1450-1452, 2013.

-. Health-;-gibbons, L. Belizán, J. M. Lauer, J. A. Betrán, A. P. Merialdi et al., The Global Numbers and Costs of Additionally Needed and Unnecessary Caesarean Sections Performed per Year: Overuse as a Barrier to Universal Coverage, Geneva: World Health Organisation, 2010.

P. Bager, M. Melbye, K. Rostgaard, C. S. Benn, T. Westergaard et al., Mode of delivery and risk of allergic rhinitis and asthma, Biol Rev Camb Philos Soc, vol.111, issue.1, pp.229-272, 2003.

M. -wills-karp, J. Santeliz, and C. L. Karp, The germless theory of allergic disease: revisiting the hygiene hypothesis, Nat Rev Immunol, vol.1, issue.1, pp.69-75, 2001.

J. Neu, J. Rushing-;-465-palmer, C. Bik, E. M. Digiulio, D. B. Relman et al., Cesarean versus Vaginal Delivery: Long term infant outcomes and the Hygiene Hypothesis, Clin Perinatol, vol.38, issue.2, p.177, 2007.

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

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, vol.28, issue.1, pp.19-25, 1999.

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.

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, vol.118, issue.2, pp.511-532, 2006.

E. Rutayisire, K. Huang, Y. Liu, and F. Tao, The mode of delivery affects the diversity and colonization pattern of the gut microbiota during the first year of infants' life: a systematic review, BMC Gastroenterol, vol.16, issue.1, p.86, 2016.

D. M. Chu, J. Ma, A. L. Prince, K. M. Antony, M. D. Seferovic et al., Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery, Nat Med, vol.23, issue.3, pp.314-326, 2017.

N. A. -bokulich, J. Chung, T. Battaglia, N. Henderson, M. Jay et al., Antibiotics, birth mode, and diet shape microbiome maturation during early life, Sci Transl Med, vol.8, issue.343, pp.343-82, 2016.

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, vol.53, issue.9, pp.1388-1397, 2004.

K. Negele, J. Heinrich, M. Borte, V. Berg, A. Schaaf et al., Mode of delivery and development of atopic disease during the first 2 years of life, Pediatr Allergy Immunol, vol.15, issue.1, pp.48-54, 2004.

J. S. Debley, J. M. Smith, G. J. Redding, and C. W. Critchlow, Childhood asthma hospitalization risk after cesarean delivery in former term and premature infants, Ann Allergy Asthma Immunol, vol.94, issue.2, pp.228-261, 2005.

B. Laubereau, B. Filipiak-pittroff, V. Berg, A. Grübl, A. Reinhardt et al., Caesarean section and gastrointestinal symptoms, atopic dermatitis, and sensitisation during the first year of life, Arch Dis Child, vol.89, issue.11, pp.993-1000, 2004.

M. Eggesbø, G. Botten, H. Stigum, P. Nafstad, and P. Magnus, Is delivery by cesarean section a risk factor for food allergy?, J Allergy Clin Immunol, vol.112, issue.2, pp.420-426, 2003.

S. Thavagnanam, J. Fleming, A. Bromley, M. D. Shields, and C. R. Cardwell, A meta-analysis of the association between Caesarean section and childhood asthma, Clin Exp Allergy, vol.38, issue.4, pp.629-662, 2008.

P. Bager, J. Wohlfahrt, and T. Westergaard, Caesarean delivery and risk of atopy and allergic disease: meta-analyses, Clin Exp Allergy, vol.38, issue.4, pp.634-676, 2008.

C. R. Cardwell, L. C. Stene, G. Joner, O. Cinek, and J. Svensson, Caesarean section is associated with an increased risk of childhood-onset type 1 diabetes mellitus: a metaanalysis of observational studies, Diabetologia, vol.51, issue.5, pp.726-761, 2008.

E. Bonifacio, K. Warncke, C. Winkler, M. Wallner, and A. G. Ziegler, Cesarean section and interferon-induced helicase gene polymorphisms combine to increase childhood type 1 diabetes risk, Diabetes, vol.60, issue.12, pp.3300-3306, 2011.

G. V. Guibas, G. Moschonis, P. Xepapadaki, E. Roumpedaki, O. Androutsos et al., Conception via in vitro fertilization and delivery by Caesarean section are associated with paediatric asthma incidence, Clin Exp Allergy, vol.43, issue.9, pp.1058-66, 2013.

B. Hesselmar, F. Sjöberg, R. Saalman, N. Aberg, I. Adlerberth et al., Pacifier cleaning practices and risk of allergy development, Pediatrics, vol.131, issue.6, pp.1829-1866, 2013.

J. Penders, K. Gerhold, E. E. Stobberingh, C. Thijs, K. Zimmermann et al., Establishment of the intestinal microbiota and its role for atopic dermatitis in early childhood, J Allergy Clin Immunol, vol.132, issue.3, pp.601-607, 2013.

K. G. Dewey, L. A. Nommsen-rivers, M. J. Heinig, and R. J. Cohen, Risk factors for suboptimal infant breastfeeding behavior, delayed onset of lactation, and excess neonatal weight loss, Pediatrics, vol.112, issue.3, pp.607-626, 2003.

K. C. Evans, R. G. Evans, R. Royal, A. J. Esterman, and S. L. James, Effect of caesarean section on breast milk transfer to the normal term newborn over the first week of life, Arch Dis Child Fetal Neonatal Ed, vol.88, issue.5, pp.380-382, 2003.

S. L. Prescott, C. Macaubas, T. Smallacombe, B. J. Holt, P. D. Sly et al., Development of allergen-specific T-cell memory in atopic and normal children, Lancet, vol.353, issue.9148, pp.196-200, 1999.

J. -neu and J. Rushing, Cesarean versus vaginal delivery: long-term infant outcomes and the hygiene hypothesis, Clin Perinatol, vol.38, issue.2, pp.321-352, 2011.

D. Munblit, D. G. Peroni, A. Boix-amorós, P. S. Hsu, B. Van't-land et al., Human Milk and Allergic Diseases: An Unsolved Puzzle, vol.9, p.894, 2017.

D. , A. A. Scaloni, A. Zolla, and L. , Human milk proteins: an interactomics and updated functional overview, J Proteome Res, vol.9, issue.7, pp.3339-73, 2010.

T. Hennet and L. Borsig, Breastfed at Tiffany's, Trends Biochem Sci, vol.41, issue.6, pp.508-518, 2016.

S. Rautava and W. A. Walker, Academy of Breastfeeding Medicine founder's lecture 2008: breastfeeding--an extrauterine link between mother and child, Breastfeed Med, vol.4, issue.1, pp.3-10, 2009.

V. -verhasselt, V. Milcent, J. Cazareth, A. Kanda, S. Fleury et al., Breast milk-mediated transfer of an antigen induces tolerance and protection from allergic asthma, Nat Med, vol.14, issue.2, pp.170-175, 2008.

T. -pozo-rubio, A. Capilla, J. R. Mujico, G. De-palma, M. A. Sanz et al., Influence of breastfeeding versus formula feeding on lymphocyte subsets in infants at risk of coeliac disease: the PROFICEL study, Eur J Nutr, vol.52, issue.2, pp.637-683, 2013.

E. Kainonen, S. Rautava, and E. Isolauri, Immunological programming by breast milk creates an anti-inflammatory cytokine milieu in breast-fed infants compared to formula-fed infants, J Nutr, vol.109, issue.11, pp.1962-70, 2013.

C. Gale, K. M. Logan, S. Santhakumaran, J. R. Parkinson, M. J. Hyde et al., Effect of breastfeeding compared with formula feeding on infant body composition: a systematic review and meta-analysis, Am J Clin Nutr, vol.95, issue.3, pp.656-69, 2012.

S. M. -innis, Human milk: maternal dietary lipids and infant development, Proc Nutr Soc, vol.66, issue.3, pp.397-404, 2007.

L. Bode, Human milk oligosaccharides: every baby needs a sugar mama, Glycobiology, vol.22, issue.9, pp.1147-62, 2012.

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, vol.30, issue.1, pp.61-68, 2000.

H. Yoshioka, K. Iseki, and K. Fujita, Development and differences of intestinal flora in the neonatal period in breast-fed and bottle-fed infants, Pediatrics, vol.72, issue.3, pp.317-338, 1983.

M. B. Azad, T. Konya, R. R. Persaud, D. S. Guttman, R. S. Chari et al., Association of Exposure to Formula in the Hospital and Subsequent Infant Feeding Practices With Gut Microbiota and Risk of Overweight in the First Year of Life, JAMA Pediatr, vol.123, issue.6, p.181161, 2016.

J. D. Forbes and M. B. Azad, Canadian Healthy Infant Longitudinal Development (CHILD) Study Investigators. Association of Exposure to Formula in the Hospital and Subsequent Infant Feeding Practices With Gut Microbiota and Risk of Overweight in the First Year of Life, JAMA Pediatr, vol.172, issue.7, p.181161, 2018.

J. L. Kaplan, H. N. Shi, and W. A. Walker, The role of microbes in developmental immunologic programming, Pediatr Res, vol.69, issue.6, pp.465-72, 2011.

S. Moossavi, K. Miliku, S. Sepehri, E. Khafipour, and M. B. Azad, The Prebiotic and Probiotic Properties of Human Milk: Implications for Infant Immune Development and Pediatric Asthma. Front Pediatr, Nature, vol.6, issue.7164, pp.811-819, 2007.

T. R. Abrahamsson, H. E. Jakobsson, A. F. Andersson, B. Björkstén, L. Engstrand et al., Low gut microbiota diversity in early infancy precedes asthma at school age, Clin Exp Allergy, vol.44, issue.6, pp.842-50, 2014.

K. Miliku and M. B. Azad, Breastfeeding and the Developmental Origins of Asthma: Current Evidence, Possible Mechanisms, and Future Research Priorities Nutrients, vol.10, p.995, 2018.

C. M. -dogaru, D. Nyffenegger, A. M. Pescatore, B. D. Spycher, and C. E. Kuehni, Breastfeeding and childhood asthma: systematic review and meta-analysis, Am J Epidemiol, vol.179, issue.10, pp.1153-67, 2014.

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, Clin Exp Allergy, vol.39, issue.12, pp.1842-51, 2009.

B. E. -snijders, C. Thijs, P. C. Dagnelie, F. F. Stelma, M. Mommers et al., Breast-feeding duration and infant atopic manifestations, by maternal allergic status, in the first 2 years of life (KOALA study), J Pediatr, vol.151, issue.4, pp.1-2, 2007.

I. Kull, E. Melen, J. Alm, J. Hallberg, M. Svartengren et al., Breast-feeding in relation to asthma, lung function, and sensitization in young schoolchildren, J Allergy Clin Immunol, vol.125, issue.5, pp.1013-1022, 2010.

J. Montoya, N. B. Matta, P. Suchon, M. C. Guzian, N. C. Lambert et al., Patients with ankylosing spondylitis have been breast fed less often than healthy controls: a casecontrol retrospective study, Ann Rheum Dis, vol.75, issue.5, pp.879-82, 2016.

M. S. Ehlayel and A. Bener, Duration of breast-feeding and the risk of childhood allergic diseases in a developing country, Allergy Asthma Proc, vol.29, issue.4, pp.386-91, 2008.

A. -sepa, A. Frodi, and J. Ludvigsson, Mothers' experiences of serious life events increase the risk of diabetes-related autoimmunity in their children, Diabetes Care, vol.28, issue.10, pp.2394-2403, 2005.

H. Vlajinac, S. Sipeti?, J. Marinkovi?, M. Bjeki?, N. Kocev et al., The Belgrade childhood diabetes study -comparison of children with type 1 diabetes with their siblings, Paediatr Perinat Epidemiol, vol.20, issue.3, pp.238-281, 2006.

H. Peng and W. Hagopian, Environmental factors in the development of Type 1 diabetes, Rev Endocr Metab Disord, vol.7, issue.3, pp.149-62, 2006.

E. Chen, G. E. Miller, H. A. Walker, J. M. Arevalo, C. Y. Sung et al., Perinatal risk factors for inflammatory bowel disease: a case-control study, Am J Epidemiol, vol.64, issue.1, pp.1111-1120, 1990.

M. Gurven, H. Kaplan, J. Winking, C. Finch, E. M. Crimmins et al., Analysis of variability of high sensitivity C-reactive protein in lowland Ecuador reveals no evidence of chronic low-grade inflammation, J Gerontol A Biol Sci Med Sci, vol.63, issue.2, pp.196-205, 2008.

A. W. Graham, C. L. Rook, C. A. Raison, and . Lowry, Childhood microbial experience, immunoregulation, inflammation and adult susceptibility to psychosocial stressors and depression in rich and poor countries, Evol Med Public Health, vol.2013, issue.1, pp.14-17, 2013.

T. W. Mcdade, J. Rutherford, L. Adair, and C. W. Kuzawa, Early origins of inflammation: microbial exposures in infancy predict lower levels of C-reactive protein in adulthood, Proc Biol Sci, vol.277, pp.1129-1166, 1684.

H. A. Hong, R. Khaneja, N. M. Tam, A. Cazzato, S. Tan et al., 526-Nicholson WL. Roles of Bacillus endospores in the environment, Cell Mol Life Sci, vol.160, issue.2, pp.410-416, 2002.

G. Casula and S. M. Cutting, Bacillus probiotics: spore germination in the gastrointestinal tract, Appl Environ Microbiol, vol.68, issue.5, pp.2344-52, 2002.

N. K. Tam, N. Q. Uyen, H. A. Hong, H. Duc-le, T. T. Hoa et al., Role of commensal bacteria in development of gut-associated lymphoid tissues and preimmune antibody repertoire, J Bacteriol, vol.188, issue.7, pp.1118-1142, 2004.

H. A. Hong, R. Khaneja, N. M. Tam, A. Cazzato, S. Tan et al., Bacillus subtilis isolated from the human gastrointestinal tract, Res Microbiol, vol.160, issue.2, pp.134-177, 2009.

H. A. Hong, E. To, S. Fakhry, L. Baccigalupi, R. E. Cutting et al., Defining the natural habitat of Bacillus spore-formers, Res Microbiol, vol.160, issue.6, pp.375-384, 2009.

J. Riedler, C. Braun-fahrländer, W. Eder, M. Schreuer, M. Waser et al.,

. Alex-study-team, Exposure to farming in early life and development of asthma and allergy: a cross-sectional survey, Lancet, vol.358, issue.9288, pp.1129-1162, 2001.

N. Aichbhaumik, E. M. Zoratti, R. Strickler, G. Wegienka, D. R. Ownby et al., Prenatal exposure to household pets influences fetal immunoglobulin E production, Clin Exp Allergy, vol.38, issue.11, pp.1787-94, 2008.

S. J. Song, C. Lauber, E. K. Costello, C. A. Lozupone, G. Humphrey et al., Cohabiting family members share microbiota with one another and with their dogs. Elife, vol.2, p.458, 2013.

K. E. Fujimura, C. C. Johnson, D. R. Ownby, M. J. Cox, E. L. Brodie et al., Man's best friend? The effect of pet ownership on house dust microbial communities, J Allergy Clin Immunol, vol.126, issue.2, pp.1-3, 2010.

R. R. Dunn, N. Fierer, J. B. Henley, J. W. Leff, and H. L. Menninger, Home life: factors structuring the bacterial diversity found within and between homes, PLoS One, vol.8, issue.5, p.64133, 2013.

J. Pakarinen, A. Hyvärinen, M. Salkinoja-salonen, S. Laitinen, A. Nevalainen et al., Predominance of Gram-positive bacteria in house dust in the low-allergy risk Russian Karelia, Environ Microbiol, vol.10, issue.12, pp.3317-3342, 2008.

D. Pearl, . Houghteling, W. Mda, C. Walker-;-braun-fahrländer, M. Gassner et al., Prevalence of hay fever and allergic sensitization in farmer's children and their peers living in the same rural community. SCARPOL team. Swiss Study on Childhood Allergy and Respiratory Symptoms with Respect to Air Pollution, J Clin Gastroenterol, vol.539, issue.1, pp.28-34, 1999.

G. Loss, S. Apprich, M. Waser, W. Kneifel, J. Genuneit et al., GABRIELA study group. The protective effect of farm milk consumption on childhood asthma and atopy: the GABRIELA study, J Allergy Clin Immunol, vol.128, issue.4, pp.766-773, 2011.

, Exposure to environmental microorganisms and childhood asthma, N Engl J Med, vol.364, issue.8, pp.701-710, 2011.

K. L. Alexander and S. R. Targan, Elson CO 3rd. Microbiota activation and regulation of innate and adaptive immunity, Immunol Rev, vol.260, issue.1, pp.206-226, 2014.

M. C. Arrieta, L. T. Stiemsma, N. Amenyogbe, E. M. Brown, B. Finlay et al., Use of antibiotics during pregnancy increases the risk of asthma in early childhood, J Pediatr, vol.5, issue.4, pp.832-838, 2013.

S. L. Russell, M. J. Gold, M. Hartmann, B. P. Willing, L. Thorson et al., Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic asthma, EMBO Rep, vol.13, issue.5, pp.440-447, 2012.

J. Metsälä, A. Lundqvist, L. J. Virta, M. Kaila, M. Gissler et al., Mother's and offspring's use of antibiotics and infant allergy to cow's milk, Epidemiology, vol.24, issue.2, pp.303-312, 2013.

A. A. Villarreal, F. J. Aberger, R. Benrud, and J. D. Gundrum, Use of broad-spectrum antibiotics and the development of irritable bowel syndrome, WMJ, vol.111, issue.1, pp.17-20, 2012.

F. -fouhy, C. M. Guinane, S. Hussey, R. Wall, C. A. Ryan et al., Highthroughput 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.

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, vol.108, issue.1, pp.4554-61, 2011.

M. B. Azad, S. L. Bridgman, A. B. Becker, and A. L. Kozyrskyj, Infant antibiotic exposure and the development of childhood overweight and central adiposity, Int J Obes, vol.38, issue.10, pp.1290-1298, 2014.

A. Kilkkinen, S. M. Virtanen, T. Klaukka, M. G. Kenward, M. Salkinoja-salonen et al., Use of antimicrobials and risk of type 1 diabetes in a population-based motherchild cohort, Diabetologia, vol.49, issue.1, pp.66-70, 2006.

A. Hviid, H. Svanström, and M. Frisch, Antibiotic use and inflammatory bowel diseases in childhood, Gut, vol.60, issue.1, pp.49-54, 2011.

W. Murk, K. R. Risnes, M. Bracken, F. A. Van-nimwegen, J. Penders et al., Prenatal or early-life exposure to antibiotics and risk of childhood asthma: a systematic review, J Allergy Clin Immunol, vol.127, issue.6, pp.1-3, 2011.

G. -gonzalez-perez, A. L. Hicks, T. M. Tekieli, C. M. Radens, B. L. Williams et al., Maternal Antibiotic Treatment Impacts Development of the Neonatal Intestinal Microbiome and Antiviral Immunity, J Immunol, vol.196, issue.9, pp.3768-79, 2016.

I. Aloisio, A. Quagliariello, S. De-fanti, D. Luiselli, D. Filippo et al., Evaluation of the effects of intrapartum antibiotic prophylaxis on newborn intestinal microbiota using a sequencing approach targeted to multi hypervariable 16S rDNA regions, Appl Microbiol Biotechnol, vol.100, issue.12, pp.5537-5583, 2016.

I. Aloisio, G. Mazzola, L. T. Corvaglia, G. Tonti, G. Faldella et al., Influence of intrapartum antibiotic prophylaxis against group B Streptococcus on the early newborn gut composition and evaluation of the anti-Streptococcus activity of Bifidobacterium strains, Appl Microbiol Biotechnol, vol.98, issue.13, pp.6051-60, 2014.

L. Corvaglia, G. Tonti, S. Martini, A. Aceti, G. Mazzola et al., Influence of Intrapartum Antibiotic Prophylaxis for Group B Streptococcus on Gut Microbiota in the First Month of Life, J Pediatr Gastroenterol Nutr, vol.62, issue.2, pp.304-312, 2016.

G. Mazzola, K. Murphy, R. P. Ross, D. Gioia, D. Biavati et al., Early Gut Microbiota Perturbations Following Intrapartum Antibiotic Prophylaxis to Prevent Group B Streptococcal Disease, PLoS One, vol.11, issue.6, p.157527, 2016.

M. -fahima, Asthme allergique induit par un allergène d'acarien, House Dust Mite (HDM) : rôles de la caspase-1 et de la protéine kinase C thêta (PKC-?). Allergologie. Université d'Orléans, 2014.

L. Shannon, . Russell, J. Matthew, M. Gold, . Hartmann et al., Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic asthma EMBO Rep, 2012.

S. L. Russell, M. J. Gold, B. P. Willing, L. Thorson, K. M. Mcnagny et al., Perinatal antibiotic treatment affects murine microbiota, immune responses and allergic asthma, Gut Microbes, vol.4, issue.2, pp.158-64, 2013.

D. A. Hill and D. Artis, Intestinal bacteria and the regulation of immune cell homeostasis, Annu Rev Immunol, vol.28, pp.623-67, 2010.

N. Oyama, N. Sudo, H. Sogawa, and C. Kubo, Antibiotic use during infancy promotes a shift in the T(H)1/T(H)2 balance toward T(H)2-dominant immunity in mice, J Allergy Clin Immunol, vol.107, issue.1, pp.153-162, 2001.

M. E. -bashir, S. Louie, H. N. Shi, and C. Nagler-anderson, Toll-like receptor 4 signaling by intestinal microbes influences susceptibility to food allergy, J Immunol, vol.172, issue.11, pp.6978-87, 2004.

B. Alm, L. Erdes, P. Möllborg, R. Pettersson, S. G. Norvenius et al., Neonatal antibiotic treatment is a risk factor for early wheezing, Pediatrics, vol.121, issue.4, pp.697-702, 2008.

B. Alm, E. Goksör, R. Pettersson, P. Möllborg, L. Erdes et al., Antibiotics in the first week of life is a risk factor for allergic rhinitis at school age, Pediatr Allergy Immunol, vol.25, issue.5, pp.468-72, 2014.

F. Marra, C. A. Marra, K. Richardson, L. D. Lynd, M. Fitzgerald et al., 569-World Health Organization. (18/04/2018) WHO guidelines on hand hygiene in health care: first global patient safety challenge. Geneva: World Health Organization, BMC Pulm Med, vol.11, issue.5, pp.462-74, 2009.

R. E. -black, A. C. Dykes, K. E. Anderson, J. G. Wells, S. P. Sinclair et al., The global water supply and sanitation assessment. Geneva: World Health Organisation, Am J Epidemiol, vol.113, issue.4, pp.445-51, 1981.

A. V. Bartlett, B. A. Jarvis, V. Ross, T. M. Katz, M. A. Dalia et al., Diarrheal illness among infants and toddlers in day care centers: effects of active surveillance and staff training without subsequent monitoring, Am J Epidemiol, vol.127, issue.4, pp.808-825, 1988.

L. Roberts, W. Smith, L. Jorm, M. Patel, R. M. Douglas et al., Effect of infection control measures on the frequency of upper respiratory infection in child care: a randomized, controlled trial, Pediatrics, vol.105, issue.4, pp.738-780, 2000.

A. E. Aiello and E. L. Larson, What is the evidence for a causal link between hygiene and infections? Lancet Infect Dis, vol.2, pp.103-113, 2002.

V. Curtis and S. Cairncross, Effect of washing hands with soap on diarrhoea risk in the community: a systematic review, Lancet Infect Dis, vol.3, issue.5, pp.275-81, 2003.

A. E. Aiello, R. M. Coulborn, V. Perez, and E. L. Larson, Effect of hand hygiene on infectious disease risk in the community setting: a meta-analysis, Am J Public Health, vol.98, issue.8, pp.1372-81, 2008.

T. Rabie and V. Curtis, Handwashing and risk of respiratory infections: a quantitative systematic review, Trop Med Int Health, vol.11, issue.3, pp.258-67, 2006.

A. M. -snelling, T. Saville, D. Stevens, and C. B. Beggs, Comparative evaluation of the hygienic efficacy of an ultra-rapid hand dryer vs conventional warm air hand dryers, J Appl Microbiol, vol.110, issue.1, pp.19-26, 2011.

E. L. -best and K. Redway, Comparison of different hand-drying methods: the potential for airborne microbe dispersal and contamination, J Hosp Infect, vol.89, issue.3, pp.215-222, 2015.

P. T. Kimmitt and K. F. Redway, Evaluation of the potential for virus dispersal during hand drying: a comparison of three methods, J Appl Microbiol, vol.120, issue.2, pp.478-86, 2016.

J. T. Staley and A. Konopka, Measurement of in situ activities of nonphotosynthetic microorganisms in aquatic and terrestrial habitats, Annu Rev Microbiol, vol.39, pp.369-94, 1985.

H. H. Kong, B. Andersson, T. Clavel, J. E. Common, S. A. Jackson et al., Performing Skin Microbiome Research: A Method to the Madness, J Invest Dermatol, vol.137, issue.3, pp.561-568, 2017.

H. H. Kong and J. A. Segre, The Molecular Revolution in Cutaneous Biology: Investigating the Skin Microbiome, J Invest Dermatol, vol.137, issue.5, pp.119-122, 2017.

C. Zapka, J. Leff, J. Henley, J. Tittl, D. Nardo et al., Comparison of Standard Culture-Based Method to Culture-Independent Method for Evaluation of Hygiene Effects on the Hand Microbiome, vol.8, pp.93-110, 2017.

D. A. -somerville-millar and W. C. Noble, Resident and transient bacteria of the skin, J Cutan Pathol, vol.1, issue.6, pp.260-264, 1974.

I. Cho and M. J. Blaser, The human microbiome: at the interface of health and disease, Nat Rev Genet, vol.13, issue.4, pp.260-70, 2012.

P. B. -price, The bacteriology of normal skin; a new quantitative test applied to a study of the bacterial flora and the disinfectant action of mechanical cleansing, J Infect Dis, vol.1938, pp.301-319, 1938.

D. Pittet, S. Dharan, S. Touveneau, V. Sauvan, and T. V. Perneger, Bacterial contamination of the hands of hospital staff during routine patient care, Arch Intern Med, vol.159, issue.8, pp.821-827, 1999.

A. L. Cogen, V. Nizet, R. L. Gallo, and . Br, Skin microbiota: a source of disease or defence?, J Dermatol, vol.158, issue.3, pp.442-55, 2008.

E. A. Grice and J. A. Segre, The skin microbiome, Nat Rev Microbiol, vol.9, issue.4, pp.244-53, 2011.

I. Uçkay, D. Pittet, P. Vaudaux, H. Sax, D. Lew et al., Foreign body infections due to Staphylococcus epidermidis, Ann Med, vol.41, issue.2, pp.109-128, 2009.

S. -naik, N. Bouladoux, C. Wilhelm, M. J. Molloy, R. Salcedo et al., Compartmentalized control of skin immunity by resident commensals, Science, vol.337, issue.6098, pp.1115-1124, 2012.

J. Weber, S. Illi, D. Nowak, R. Schierl, O. Holst et al., Asthma and the hygiene hypothesis. Does cleanliness matter?, Am J Respir Crit Care Med, vol.191, issue.5, pp.522-531, 2015.

. Alex-study-team, Microbial exposure of rural school children, as assessed by levels of N-acetyl-muramic acid in mattress dust, and its association with respiratory health, J Allergy Clin Immunol, vol.113, issue.5, pp.860-867, 2004.

K. Sprunt, W. Redman, and G. Leidy, Antibacterial effectiveness of routine hand washing, Pediatrics, vol.52, issue.2, pp.264-71, 1973.

C. L. -pessoa-silva, S. Dharan, S. Hugonnet, S. Touveneau, K. Posfay-barbe et al., Dynamics of bacterial hand contamination during routine neonatal care, Infect Control Hosp Epidemiol, vol.25, issue.3, pp.192-199, 2004.

C. A. -mackintosh and P. N. Hoffman, An extended model for transfer of micro-organisms via the hands: differences between organisms and the effect of alcohol disinfection, J Hyg, vol.92, issue.3, pp.345-55, 1984.

-. Vandegrift, A. C. Bateman, K. N. Siemens, and M. Nguyen, Cleanliness in context: reconciling hygiene with a modern microbial perspective Microbiome, vol.5, p.76, 2017.

S. Higaki, M. Morohashi, T. Yamagishi, and Y. Hasegawa, Comparative study of staphylococci from the skin of atopic dermatitis patients and from healthy subjects, Int J Dermatol, vol.38, issue.4, pp.265-274, 1999.

J. A. Sanford and R. L. Gallo, Functions of the skin microbiota in health and disease, Semin Immunol, vol.25, issue.5, pp.370-377, 2013.

M. R. Williams and R. L. Gallo, The role of the skin microbiome in atopic dermatitis, Curr Allergy Asthma Rep, vol.15, issue.11, p.65, 2015.

H. H. Kong, J. Oh, C. Deming, S. Conlan, E. A. Grice et al., Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis, Genome Res, vol.22, issue.5, pp.850-859, 2012.

S. F. -bloomfield, G. A. Rook, E. A. Scott, F. Shanahan, R. Stanwell-smith et al., Time to abandon the hygiene hypothesis: new perspectives on allergic disease, the human microbiome, infectious disease prevention and the role of targeted hygiene, vol.136, pp.213-237, 2016.

-. Scudellari, Environmental Sciences News Feature: Cleaning up the hygiene hypothesis, Immunology and Inflammation, vol.114, issue.7, pp.1433-1436, 2017.

S. F. -bloomfield, M. Exner, G. M. Fara, K. J. Nath, E. A. Scott et al., An internetdelivered handwashing intervention to modify influenza-like illness and respiratory infection transmission (PRIMIT): a primary care randomised trial, Lancet, vol.386, pp.1631-1640, 2009.

B. Winther, K. Mccue, K. Ashe, J. Rubino, and J. O. Hendley, Rhinovirus contamination of surfaces in homes of adults with natural colds: transfer of virus to fingertips during normal daily activities, J Med Virol, vol.83, issue.5, pp.906-915, 2011.

S. F. -bloomfield, M. Exner, G. M. Fara, K. J. Nath, and E. A. Scott, Van der Voorden C. The global burden of hygiene-related diseases in relation to the home and community. International Scientific Forum on Home Hygiene, pp.16-19, 2009.

K. E. Jones, N. G. Patel, M. A. Levy, A. Storeygard, D. Balk et al., 613-World Health Organization (WHO). (16/08/2019) global influenza preparedness plan: The role of WHO and recommendations for national measures before and during pandemics, Nature, vol.451, issue.7181, pp.990-993, 2005.

R. G. -behling, J. Eifert, M. C. Erickson, J. B. Gurtler, J. L. Kornacki et al., Selected pathogens of concern to industrial food processors: Infectious, toxigenic, toxico-infectious, selected emerging pathogenic bacteria, Principles of Microbiological Troubleshooting in the Industrial Food Processing Environment, pp.5-61, 2010.

A. J. -hall, Noroviruses: the perfect human pathogens?, J Infect Dis, vol.205, issue.11, pp.1622-1626, 2012.

, 22/04/2019) Recommendations for future collaboration between the U.S. and EU. Transatlantic Taskforce on Antimicrobial Resistance, 2011.

;. Ar_report, . Hygiene, . Health, . Bloomfield, M. G. Dominguez-bello et al., Spread of antibiotic resistant strains in the home and community. International Scientific Forum on Home Hygiene2013, Nat Med, vol.22, issue.3, pp.250-253, 2016.

G. Vitaliti, P. Pavone, F. Guglielmo, G. Spataro, and R. Falsaperla, The immunomodulatory effect of probiotics beyond atopy: an update, J Asthma, vol.51, issue.3, pp.320-352, 2014.

L. Vitetta, G. Vitetta, and S. Hall, Immunological Tolerance and Function: Associations Between Intestinal Bacteria, Probiotics, Prebiotics, and Phages. Front Immunol, vol.9, p.2240, 2018.

A. Makrgeorgou, J. Leonardi-bee, F. J. Bath-hextall, D. F. Murrell, M. L. Tang et al., Probiotics for treating eczema, Cochrane Database Syst Rev, vol.11, p.6135, 2018.

E. Bezirtzoglou and E. Stavropoulou, Immunology and probiotic impact of the newborn and young children intestinal microflora, Anaerobe, vol.17, issue.6, pp.369-74, 2011.

P. Bodera and A. Chcialowski, Immunomodulatory effect of probiotic bacteria. Recent Pat Inflamm Allergy Drug Discov, vol.3, pp.58-64, 2009.

B. -pot, B. Foligné, C. Daniel, C. Grangette, K. Hufnagl et al., Distinctive anti-allergy properties of two probiotic bacterial strains in a mouse model of allergic poly-sensitization. Vaccine, Nestle Nutr Inst Workshop Ser, vol.77, issue.10, pp.1981-90, 2011.

, Lactobacillus johnsonii N6.2 mitigates the development of type 1 diabetes in BB-DP rats, PLoS One, vol.5, issue.5, p.10507, 2010.

S. -rausch, J. Held, A. Fischer, M. M. Heimesaat, A. A. Kühl et al., Small intestinal nematode infection of mice is associated with increased enterobacterial loads alongside the intestinal tract, PLoS One, vol.8, issue.9, p.74026, 2013.

M. Schultz, C. Veltkamp, L. A. Dieleman, W. B. Grenther, P. B. Wyrick et al., Lactobacillus plantarum 299V in the treatment and prevention of spontaneous colitis in interleukin-10-deficient mice, J Gastroenterol Hepatol, vol.8, issue.2, pp.1834-1843, 2002.

F. Calcinaro, S. Dionisi, M. Marinaro, P. Candeloro, V. Bonato et al., Oral probiotic administration induces interleukin-10 production and prevents spontaneous autoimmune diabetes in the non-obese diabetic mouse, Diabetologia, vol.48, issue.8, pp.1565-75, 2005.

F. F. Duan, J. H. Liu, and J. C. March, Engineered commensal bacteria reprogram intestinal cells into glucose-responsive insulin-secreting cells for the treatment of diabetes, Diabetes, vol.64, issue.5, pp.1794-803, 2015.

F. Calcinaro, S. Dionisi, M. Marinaro, P. Candeloro, V. Bonato et al., Oral probiotic administration induces interleukin-10 production and prevents spontaneous autoimmune diabetes in the non-obese diabetic mouse, Diabetologia, vol.48, issue.8, pp.1565-75, 2005.

M. L. Tang, A. L. Ponsonby, F. Orsini, D. Tey, M. Robinson et al., Administration of a probiotic with peanut oral immunotherapy: A randomized trial, J Allergy Clin Immunol, vol.135, issue.3, pp.737-781, 2015.

T. R. Abrahamsson, T. Jakobsson, B. Björkstén, G. Oldaeus, and M. C. Jenmalm, No effect of probiotics on respiratory allergies: a seven-year follow-up of a randomized controlled trial in infancy, Pediatr Allergy Immunol, vol.24, issue.6, pp.556-61, 2013.

R. J. Boyle, F. J. Bath-hextall, J. Leonardi-bee, D. F. Murrell, M. L. Tang et al., Effects of Lactobacillus salivarius 433118 on intestinal inflammation, immunity status and in vitro colon function in two mouse models of inflammatory bowel disease, Clin Exp Allergy, vol.39, issue.8, pp.2495-506, 2008.

J. H. Cummings, G. T. Macfarlane, and H. N. Englyst, Prebiotic digestion and fermentation, Am J Clin Nutr, vol.73, issue.2, pp.415-420, 2001.

J. M. Wong, R. De-souza, C. W. Kendall, A. Emam, D. J. Jenkins et al., Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway, J Clin Gastroenterol, vol.40, issue.3, pp.235-278, 2006.

S. Arslanoglu, G. E. Moro, J. Schmitt, L. Tandoi, S. Rizzardi et al., Early dietary intervention with a mixture of prebiotic oligosaccharides reduces the incidence of allergic manifestations and infections during the first two years of life, J Nutr, vol.138, issue.6, pp.1091-1096, 2008.

A. P. Vos, B. C. Van-esch, B. Stahl, M. 'rabet, L. Folkerts et al., Dietary supplementation with specific oligosaccharide mixtures decreases parameters of allergic asthma in mice, J. Int Immunopharmacol, vol.7, issue.12, pp.1582-1589, 2007.

M. Kalliomäki, S. Salminen, H. Arvilommi, P. Kero, P. Koskinen et al., Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial, Lancet, vol.357, issue.9262, pp.1076-1085, 2001.

M. Kalliomäki, S. Salminen, T. Poussa, H. Arvilommi, and E. Isolauri, Probiotics and prevention of atopic disease: 4-year follow-up of a randomised placebo-controlled trial, Lancet, vol.361, issue.9372, pp.1869-71, 2003.

T. R. Abrahamsson, T. Jakobsson, M. F. Böttcher, M. Fredrikson, M. C. Jenmalm et al., Probiotics in prevention of IgE-associated eczema: a double-blind, randomized, placebo-controlled trial, J Allergy Clin Immunol, vol.119, issue.5, pp.1174-80, 2007.

S. Kivit, E. Saeland, A. D. Kraneveld, H. J. Van-de-kant, B. Schouten et al., Galectin-9 induced by dietary synbiotics is involved in suppression of allergic symptoms in mice and humans, 646-van der Aa LB, Heymans HS, van Aalderen WM, vol.67, pp.343-52, 2012.

K. Amor, Effect of a new synbiotic mixture on atopic dermatitis in infants: a randomized-controlled trial, 647-van der Aa LB, van Aalderen WM, Heymans HS, vol.40, pp.795-804, 2010.

. Synbad-study-group, Synbiotics prevent asthma-like symptoms in infants with atopic dermatitis, Allergy, vol.66, issue.2, pp.170-177, 2011.

E. Furrie, S. Macfarlane, A. Kennedy, J. H. Cummings, S. V. Walsh et al., Synbiotic therapy (Bifidobacterium longum/Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: a randomised controlled pilot trial, Gut, vol.54, issue.2, pp.242-251, 2005.

N. Apiwattanakul, P. G. Thomas, A. R. Iverson, and J. A. Mccullers, Chronic helminth infections impair pneumococcal vaccine responses. Vaccine, vol.32, pp.5405-5415, 2014.

S. Bobat, M. Darby, D. Mrdjen, C. Cook, E. Logan et al., Natural and vaccinemediated immunity to Salmonella Typhimurium is impaired by the helminth Nippostrongylus brasiliensis, PLoS Negl Trop Dis, vol.8, issue.12, p.3341, 2014.

J. O. Fleming and J. V. Weinstock, Clinical trials of helminth therapy in autoimmune diseases: rationale and findings, Parasite Immunol, vol.37, issue.6, pp.277-92, 2015.

J. Fleming, G. Hernandez, L. Hartman, J. Maksimovic, S. Nace et al., Safety and efficacy of helminth treatment in relapsing-remitting multiple sclerosis: Results of the HINT 2 clinical trial, Mult Scler, vol.25, issue.1, pp.81-91, 2019.

W. J. Sandborn, D. E. Elliott, J. Weinstock, R. W. Summers, A. Landry-wheeler et al., Randomised clinical trial: the safety and tolerability of Trichuris suis ova in patients with Crohn's disease, Aliment Pharmacol Ther, vol.38, issue.3, pp.255-63, 2013.

J. Croese, J. Masson, J. Cooke, S. Melrose, W. Pritchard et al., A proof of concept study establishing Necator americanus in Crohn's patients and reservoir donors, Gut, vol.55, issue.1, pp.136-143, 2006.

R. W. -summers, D. E. Elliott, K. Qadir, J. F. Urban, R. Thompson et al.,

, Trichuris suis seems to be safe and possibly effective in the treatment of inflammatory bowel disease, Am J Gastroenterol, vol.98, issue.9, pp.2034-2075, 2003.

R. W. Summers, D. E. Elliott, J. F. Urban, R. A. Thompson, and J. V. Weinstock, Trichuris suis therapy for active ulcerative colitis: a randomized controlled trial, Gastroenterology, vol.128, issue.4, pp.825-857, 2005.

J. P. Hewitson, J. R. Grainger, and R. M. Maizels, Helminth immunoregulation: the role of parasite secreted proteins in modulating host immunity, Mol Biochem Parasitol, vol.167, issue.1, pp.1-11, 2009.

H. J. -mcsorley, O. 'gorman, M. T. Blair, N. Sutherland, T. E. Filbey et al., Suppression of type 2 immunity and allergic airway inflammation by secreted products of the helminth Heligmosomoides polygyrus, Eur J Immunol, vol.42, issue.10, pp.2667-82, 2012.

H. J. Mcsorley, N. F. Blair, K. A. Smith, A. N. Mckenzie, and R. M. Maizels, Blockade of IL-33 release and suppression of type 2 innate lymphoid cell responses by helminth secreted products in airway allergy, Mucosal Immunol, vol.7, issue.5, pp.1068-78, 2014.

N. E. Ruyssers, D. Winter, B. Y. De-man, J. G. Loukas, A. Pearson et al., Therapeutic potential of helminth soluble proteins in TNBS-induced colitis in mice, Inflamm Bowel Dis, vol.15, issue.4, pp.491-500, 2009.

C. Schnoeller, S. Rausch, S. Pillai, A. Avagyan, B. M. Wittig et al., A helminth immunomodulator reduces allergic and inflammatory responses by induction of IL-10-producing macrophages, J Immunol, vol.180, issue.6, pp.4265-72, 2008.

I. Ferreira, D. Smyth, S. Gaze, A. Aziz, P. Giacomin et al., Hookworm excretory/secretory products induce interleukin-4 (IL-4)+ IL-10+ CD4+ T cell responses and suppress pathology in a mouse model of colitis, Infect Immun, vol.81, issue.6, pp.2104-2115, 2013.

P. Zaccone, Z. Fehérvári, F. M. Jones, S. Sidobre, M. Kronenberg et al., Schistosoma mansoni antigens modulate the activity of the innate immune response and prevent onset of type 1 diabetes, Eur J Immunol, vol.33, issue.5, pp.1439-1488, 2003.

L. A. Reynolds, K. A. Smith, K. J. Filbey, Y. Harcus, J. P. Hewitson et al.,

S. T. -walk, A. M. Blum, S. A. Ewing, J. V. Weinstock, and V. B. Young, Commensal-pathogen interactions in the intestinal tract: lactobacilli promote infection with, and are promoted by, helminth parasites, Inflamm Bowel Dis, vol.5, issue.4, pp.1841-1850, 2010.

A. Lyons, D. O'mahony, O. 'brien, F. Macsharry, J. Sheil et al., Bacterial strain-specific induction of Foxp3+ T regulatory cells is protective in murine allergy models, Clin Exp Allergy, vol.40, issue.5, pp.811-820, 2010.

D. -zeevi, T. Korem, N. Zmora, D. Israeli, D. Rothschild et al., Personalized Nutrition by Prediction of Glycemic Responses, Cell, vol.163, issue.5, pp.1079-1094, 2015.

. Serment-de-galien,

, Je jure, en présence des maîtres de la Faculté

, D'honorer ceux qui m'ont instruit(e) dans les préceptes de mon art et de leur témoigner ma reconnaissance en restant fidèle à leur enseignement

, intérêt de la santé publique, ma profession avec conscience et de respecter non seulement la législation en vigueur, mais aussi les règles de l'honneur

, De ne jamais oublier ma responsabilité et mes devoirs envers le malade et sa dignité humaine

, En aucun cas, je ne consentirai à utiliser mes connaissances et mon état pour corrompre les moeurs et favoriser des actes criminels

, Que les hommes m'accordent leur estime si je suis fidèle à mes promesses

, Que je sois couvert(e) d'opprobre et méprisé(e) de mes confrères si j'y manque