C. Des, V. Gag-gfp-par-les-cellules-de-l-'intestin, and .. , 64 a. Stratégie de définition des populations de cellules immunitaires 64 b. Capture in vivo des VLP-GFP par les cellules de l'intestin, p.66

.. Capture-in-vivo-des-vlp-gfp-par-les-cellules-des-poumons, 69 a 69 b. Capture in vivo des VLP-GFP par les cellules des poumons, p.71

C. A. Akdis, T. Blesken, M. Akdis, B. Wüthrich, and K. Blaser, Role of interleukin 10 in specific immunotherapy., Journal of Clinical Investigation, vol.102, issue.1, pp.98-106, 1998.
DOI : 10.1172/JCI2250

M. Andrawiss, Y. Takeuchi, L. Hewlett, C. , and M. , Murine Leukemia Virus Particle Assembly Quantitated by Fluorescence Microscopy: Role of Gag-Gag Interactions and Membrane Association, Journal of Virology, vol.77, issue.21, pp.11651-11660, 2003.
DOI : 10.1128/JVI.77.21.11651-11660.2003

K. Atarashi, T. Tanoue, T. Shima, A. Imaoka, T. Kuwahara et al., Induction of Colonic Regulatory T Cells by Indigenous Clostridium Species, Science, vol.188, issue.10, pp.337-341, 2011.
DOI : 10.1084/jem.188.10.1929

P. Bager, J. Wohlfahrt, and T. Westergaard, Caesarean delivery and risk of atopy and allergic disesase: meta-analyses, Clinical & Experimental Allergy, vol.76, issue.4, pp.634-642, 2008.
DOI : 10.1034/j.1398-9995.2001.056006491.x

B. Bellier, C. Dalba, B. Clerc, D. Desjardins, R. Drury et al., DNA vaccines encoding retrovirus-based virus-like particles induce efficient immune responses without adjuvant, Vaccine, vol.24, issue.14, pp.2643-2655, 2006.
DOI : 10.1016/j.vaccine.2005.11.034

B. Bellier, C. Huret, M. Miyalou, D. Desjardins, M. Frenkiel et al., DNA vaccines expressing retrovirus-like particles are efficient immunogens to induce neutralizing antibodies, Vaccine, vol.27, issue.42, pp.5772-5780, 2009.
DOI : 10.1016/j.vaccine.2009.07.059

URL : https://hal.archives-ouvertes.fr/hal-00420201

M. J. Benson, K. Pino-lagos, M. Rosemblatt, N. , and R. J. , All-trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation, The Journal of Experimental Medicine, vol.154, issue.8, pp.1765-1774, 2007.
DOI : 10.1093/intimm/10.12.1969

S. Bhattacharyya, P. Sen, M. Wallet, B. Long, A. S. Baldwin et al., Immunoregulation of dendritic cells by IL-10 is mediated through suppression of the PI3K/Akt pathway and of I??B kinase activity, Blood, vol.104, issue.4, pp.1100-1109, 2004.
DOI : 10.1182/blood-2003-12-4302

A. B. Blázquez and M. C. Berin, Gastrointestinal Dendritic Cells Promote Th2 Skewing via OX40L, The Journal of Immunology, vol.180, issue.7, pp.4441-4450, 1950.
DOI : 10.4049/jimmunol.180.7.4441

H. Bour-jordan, J. L. Grogan, Q. Tang, J. A. Auger, R. M. Locksley et al., CTLA-4 regulates the requirement for cytokine-induced signals in TH2 lineage commitment, Nature Immunology, vol.4, issue.2, pp.182-188, 2003.
DOI : 10.1038/ni884

P. J. Ceponis, F. Botelho, C. D. Richards, and D. M. Mckay, Interleukins 4 and 13 Increase Intestinal Epithelial Permeability by a Phosphatidylinositol 3-Kinase Pathway, Journal of Biological Chemistry, vol.162, issue.37, pp.29132-29137, 2000.
DOI : 10.4049/jimmunol.164.1.404

URL : http://www.jbc.org/content/275/37/29132.full.pdf

B. Chackerian, Virus-like particles: flexible platforms for vaccine development, Expert Review of Vaccines, vol.23, issue.3, pp.381-390, 2007.
DOI : 10.1016/j.vaccine.2005.05.007

X. Chen, C. Song, Z. Liu, B. Feng, P. Zheng et al., Intestinal epithelial cells express galectin-9 in patients with food allergy that plays a critical role in sustaining allergic status in mouse intestine, Allergy, vol.284, issue.8, pp.1038-1046, 2011.
DOI : 10.1074/jbc.M109.035196

D. Chiang and M. C. Berin, An Examination of Clinical and Immunologic Outcomes in Food Allergen Immunotherapy by Route of Administration, Current Allergy and Asthma Reports, vol.133, issue.3, p.35, 2015.
DOI : 10.1016/j.jaci.2014.01.007

D. K. Chu, A. Llop-guevara, T. D. Walker, K. Flader, S. Goncharova et al., IL-33, but not thymic stromal lymphopoietin or IL-25, is central to mite and peanut allergic sensitization, Journal of Allergy and Clinical Immunology, vol.131, issue.1, pp.187-200, 2013.
DOI : 10.1016/j.jaci.2012.08.002

D. K. Chu, R. Jimenez-saiz, C. P. Verschoor, T. D. Walker, S. Goncharova et al., Indigenous enteric eosinophils control DCs to initiate a primary Th2 immune response in vivo, The Journal of Experimental Medicine, vol.3, issue.8, pp.1657-1672, 2014.
DOI : 10.1084/jem.20020656

G. Churlaud, V. Jimenez, J. Ruberte, M. Amadoudji-zin, G. Fourcade et al., Sustained stimulation and expansion of Tregs by IL2 control autoimmunity without impairing immune responses to infection, vaccination and cancer, Clinical Immunology, vol.151, issue.2, 2014.
DOI : 10.1016/j.clim.2014.02.003

J. L. Coombes, K. R. Siddiqui, C. V. Arancibia-cárcamo, J. Hall, C. Sun et al., regulatory T cells via a TGF-????? and retinoic acid???dependent mechanism, The Journal of Experimental Medicine, vol.204, issue.8, pp.1757-1764, 2007.
DOI : 10.1093/nar/29.9.e45

C. Davidoff, R. J. Payne, S. H. Willis, B. J. Doranz, and J. B. Rucker, Maturation of the Gag core decreases the stability of retroviral lipid membranes, Virology, vol.433, issue.2, pp.401-409, 2012.
DOI : 10.1016/j.virol.2012.08.023

T. L. Denning, N. A. Campbell, F. Song, R. P. Garofalo, G. R. Klimpel et al., Expression of IL-10 receptors on epithelial cells from the murine small and large intestine, International Immunology, vol.12, issue.2, pp.133-139, 2000.
DOI : 10.1093/intimm/12.2.133

T. L. Denning, Y. Wang, S. R. Patel, I. R. Williams, and B. Pulendran, Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17???producing T cell responses, Nature Immunology, vol.146, issue.10, pp.1086-1094, 2007.
DOI : 10.1038/ni1212

J. Descotes and G. Choquet-kastylevsky, Gell and Coombs's classification: is it still valid?, Toxicology, vol.158, issue.1-2, pp.43-49, 2001.
DOI : 10.1016/S0300-483X(00)00400-5

L. Ding and E. M. Shevach, IL-10 inhibits mitogen-induced T cell proliferation by selectively inhibiting macrophage costimulatory function, J. Immunol. Baltim. Md, pp.148-3133, 1950.

C. P. Frossard, L. Tropia, C. Hauser, and P. A. Eigenmann, Lymphocytes in Peyer patches regulate clinical tolerance in a murine model of food allergy???, Journal of Allergy and Clinical Immunology, vol.113, issue.5, pp.958-964, 2004.
DOI : 10.1016/j.jaci.2003.12.017

T. Fukaya, H. Takagi, Y. Sato, K. Sato, K. Eizumi et al., Crucial roles of B7-H1 and B7-DC expressed on mesenteric lymph node dendritic cells in the generation of antigen-specific CD4+Foxp3+ regulatory T cells in the establishment of oral tolerance, Blood, vol.116, issue.13, pp.2266-2276, 2010.
DOI : 10.1182/blood-2009-10-250472

URL : https://hal.archives-ouvertes.fr/hal-00605852

S. Gaffen and K. Liu, Overview of interleukin-2 function, production and clinical applications, Cytokine, vol.28, issue.3, 2004.
DOI : 10.1016/j.cyto.2004.06.010

C. Gamazo, G. Gastaminza, M. Ferrer, M. L. Sanz, and J. M. Irache, Nanoparticle based-immunotherapy against allergy, Immunotherapy, vol.6, issue.7, pp.885-897, 2014.
DOI : 10.2217/imt.14.63

G. Gri, S. Piconese, B. Frossi, V. Manfroi, S. Merluzzi et al., CD4+CD25+ Regulatory T Cells Suppress Mast Cell Degranulation and Allergic Responses through OX40-OX40L Interaction, Immunity, vol.29, issue.5, pp.771-781, 2008.
DOI : 10.1016/j.immuni.2008.08.018

URL : http://doi.org/10.1016/j.immuni.2008.08.018

R. Gupta, A. Sheikh, D. P. Strachan, A. , and H. R. , Time trends in allergic disorders in the UK, Thorax, vol.62, issue.1, pp.91-96, 2007.
DOI : 10.1136/thx.2004.038844

U. Hadis, B. Wahl, O. Schulz, M. Hardtke-wolenski, A. Schippers et al., Intestinal Tolerance Requires Gut Homing and Expansion of FoxP3+ Regulatory T Cells in the Lamina Propria, Immunity, vol.34, issue.2, pp.237-246, 2011.
DOI : 10.1016/j.immuni.2011.01.016

U. Haupts, S. Maiti, P. Schwille, W. , and W. W. , Dynamics of fluorescence fluctuations in green fluorescent protein observed by fluorescence correlation spectroscopy, Proc. Natl. Acad. Sci. U, 1998.
DOI : 10.1021/bi980857x

F. Heller, P. Florian, C. Bojarski, J. Richter, M. Christ et al., Interleukin-13 Is the Key Effector Th2 Cytokine in Ulcerative Colitis That Affects Epithelial Tight Junctions, Apoptosis, and Cell Restitution, Gastroenterology, vol.129, issue.2, pp.550-564, 2005.
DOI : 10.1016/j.gastro.2005.05.002

M. R. Hepworth, L. A. Monticelli, T. C. Fung, C. G. Ziegler, S. Grunberg et al., Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria, Nature, vol.7, issue.7452, pp.113-117, 2013.
DOI : 10.1038/nmeth.f.303

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3699860

D. A. Hill, M. C. Siracusa, M. C. Abt, B. S. Kim, D. Kobuley et al., Commensal bacteria???derived signals regulate basophil hematopoiesis and allergic inflammation, Nature Medicine, vol.180, issue.4, pp.538-546, 2012.
DOI : 10.4049/jimmunol.180.11.7327

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3321082

S. Huber, N. Gagliani, E. Esplugues, W. O-'connor, F. J. Huber et al., Th17 Cells Express Interleukin-10 Receptor and Are Controlled by Foxp3??? and Foxp3+ Regulatory CD4+ T Cells in an Interleukin-10-Dependent Manner, Immunity, vol.34, issue.4, pp.554-565, 2011.
DOI : 10.1016/j.immuni.2011.01.020

C. Huret, D. Desjardins, M. Miyalou, B. Levacher, M. Amadoudji-zin et al., Recombinant retrovirus-derived virus-like particle-based vaccines induce hepatitis C virus-specific cellular and neutralizing immune responses in mice, Vaccine, vol.31, issue.11, pp.1540-1547, 2013.
DOI : 10.1016/j.vaccine.2012.05.025

C. Jacob, P. Yang, D. Darmoul, S. Amadesi, T. Saito et al., Mast Cell Tryptase Controls Paracellular Permeability of the Intestine, Journal of Biological Chemistry, vol.276, issue.36, pp.31936-31948, 2005.
DOI : 10.1139/y03-080

S. J. Jenkins, G. Perona-wright, A. G. Worsley, N. Ishii, and A. S. Macdonald, Dendritic Cell Expression of OX40 Ligand Acts as a Costimulatory, Not Polarizing, Signal for Optimal Th2 Priming and Memory Induction In Vivo, The Journal of Immunology, vol.179, issue.6, pp.3515-3523, 1950.
DOI : 10.4049/jimmunol.179.6.3515

S. G. Johansson, T. Bieber, R. Dahl, P. S. Friedmann, B. Q. Lanier et al., Revised Nomenclature for Allergy for Global Use, Allergy & Clinical Immunology International - Journal of the World Allergy Organization, vol.17, issue.01, 2003.
DOI : 10.1027/0838-1925.17.1.4

M. Jutel, U. R. Müller, M. Fricker, S. Rihs, W. J. Pichler et al., Influence of bee venom immunotherapy on degranulation and leukotriene generation in human blood basophils, 1996.

G. Lack, Update on risk factors for food allergy, Journal of Allergy and Clinical Immunology, vol.129, issue.5, pp.1187-1197, 2012.
DOI : 10.1016/j.jaci.2012.02.036

M. Larché, C. A. Akdis, and R. Valenta, Immunological mechanisms of allergen-specific immunotherapy, Nature Reviews Immunology, vol.302, issue.10, pp.761-771, 2006.
DOI : 10.1001/jama.1940.02810270025005

D. Leo, V. Yang, P. Berin, M. C. Perdue, and M. H. , Factors Regulating the Effect of IL-4 on Intestinal Epithelial Barrier Function, International Archives of Allergy and Immunology, vol.129, issue.3, pp.219-227, 2002.
DOI : 10.1159/000066778

G. Lescaille, F. Pitoiset, R. Macedo, C. Baillou, C. Huret et al., Efficacy of DNA Vaccines Forming E7 Recombinant Retroviral Virus-Like Particles for the Treatment of Human Papillomavirus-Induced Cancers, Human Gene Therapy, vol.24, issue.5, pp.533-544, 2013.
DOI : 10.1089/hum.2012.037

T. C. Li, Y. Yamakawa, K. Suzuki, M. Tatsumi, M. A. Razak et al., Expression and self-assembly of empty virus-like particles of hepatitis E virus, J. Virol, vol.71, pp.7207-7213, 1997.

E. M. Ling, T. Smith, X. D. Nguyen, C. Pridgeon, M. Dallman et al., Relation of CD4+CD25+ regulatory T-cell suppression of allergen-driven T-cell activation to atopic status and expression of allergic disease, The Lancet, vol.363, issue.9409, pp.608-615, 2004.
DOI : 10.1016/S0140-6736(04)15592-X

Z. Ling, Z. Li, X. Liu, Y. Cheng, Y. Luo et al., Altered Fecal Microbiota Composition Associated with Food Allergy in Infants, Applied and Environmental Microbiology, vol.80, issue.8, pp.2546-2554, 2014.
DOI : 10.1128/AEM.00003-14

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3993190

A. Link, F. Zabel, Y. Schnetzler, A. Titz, F. Brombacher et al., Innate Immunity Mediates Follicular Transport of Particulate but Not Soluble Protein Antigen, The Journal of Immunology, vol.188, issue.8, 2012.
DOI : 10.4049/jimmunol.1103312

URL : http://www.jimmunol.org/content/jimmunol/188/8/3724.full.pdf

A. Llop-guevara, D. K. Chu, T. D. Walker, S. Goncharova, R. Fattouh et al., A GM-CSF/IL-33 Pathway Facilitates Allergic Airway Responses to Sub-Threshold House Dust Mite Exposure, PLoS ONE, vol.177, issue.2, p.88714, 2014.
DOI : 10.1371/journal.pone.0088714.s003

URL : http://doi.org/10.1371/journal.pone.0088714

A. Magnan, H. , and M. , Is deficient tolerance the true paradigm for atopic diseases?, Clinical <html_ent glyph="@amp;" ascii="&"/> Experimental Allergy, vol.35, issue.12, pp.1507-1510, 2005.
DOI : 10.1165/rcmb.2002-0220OC

A. Marçais, S. Viel, M. Grau, T. Henry, J. Marvel et al., Regulation of Mouse NK Cell Development and Function by Cytokines, Frontiers in Immunology, vol.4, p.450, 2013.
DOI : 10.3389/fimmu.2013.00450

G. Matteoli, E. Mazzini, I. D. Iliev, E. Mileti, F. Fallarino et al., Gut CD103+ dendritic cells express indoleamine 2,3-dioxygenase which influences T regulatory/T effector cell balance and oral tolerance induction, Gut, vol.59, issue.5, pp.595-604, 2010.
DOI : 10.1136/gut.2009.185108

E. Mazzini, L. Massimiliano, G. Penna, and M. Rescigno, Oral Tolerance Can Be Established via Gap Junction Transfer of Fed Antigens from CX3CR1+ Macrophages to CD103+ Dendritic Cells, Immunity, vol.40, issue.2, pp.248-261, 2014.
DOI : 10.1016/j.immuni.2013.12.012

F. Meiler, S. Klunker, M. Zimmermann, C. A. Akdis, and M. Akdis, Distinct regulation of IgE, IgG4 and IgA by T regulatory cells and toll-like receptors, Allergy, vol.119, issue.11, pp.1455-1463, 2008.
DOI : 10.4049/jimmunol.172.6.3580

A. M. Mowat, Anatomical basis of tolerance and immunity to intestinal antigens, Nature Reviews Immunology, vol.3, issue.4, pp.331-341, 2003.
DOI : 10.1038/nri1057

B. Niggemann, U. Staden, C. Rolinck-werninghaus, and K. Beyer, Specific oral tolerance induction in food allergy, Allergy, vol.44, issue.7, pp.808-811, 2006.
DOI : 10.1111/j.1365-2222.2005.02150.x

N. Van-panhuys, F. Klauschen, G. , and R. N. , T-Cell-Receptor-Dependent Signal Intensity Dominantly Controls CD4+ T Cell Polarization In??Vivo, Immunity, vol.41, issue.1, pp.63-74, 2014.
DOI : 10.1016/j.immuni.2014.06.003

M. C. Pereira-santos, A. P. Baptista, A. Melo, R. R. Alves, R. S. Soares et al., Expansion of circulating Foxp3+CD25bright CD4+ T cells during specific venom immunotherapy, Clinical & Experimental Allergy, vol.55, issue.2, pp.291-297, 2008.
DOI : 10.4049/jimmunol.171.11.5760

A. M. Piliponsky, G. J. Gleich, I. Bar, and F. Levi-schaffer, Effects of eosinophils on mast cells: a new pathway for the perpetuation of allergic inflammation, Molecular Immunology, vol.38, issue.16-18, pp.1369-1372, 2002.
DOI : 10.1016/S0161-5890(02)00090-1

S. L. Prescott, R. Pawankar, K. J. Allen, D. E. Campbell, J. K. Sinn et al., A global survey of changing patterns of food allergy burden in children, World Allergy Organization Journal, vol.6, issue.1, 2013.
DOI : 10.1016/j.jaci.2007.08.023

C. Primard, N. Rochereau, E. Luciani, C. Genin, T. Delair et al., Traffic of poly(lactic acid) nanoparticulate vaccine vehicle from intestinal mucus to sub-epithelial immune competent cells, Biomaterials, vol.31, issue.23, pp.6060-6068, 2010.
DOI : 10.1016/j.biomaterials.2010.04.021

URL : https://hal.archives-ouvertes.fr/hal-00595098

L. S. Van-rijt, S. Jung, A. Kleinjan, N. Vos, M. Willart et al., dendritic cells during allergen challenge abrogates the characteristic features of asthma, The Journal of Experimental Medicine, vol.14, issue.6, pp.981-991, 2005.
DOI : 10.1164/ajrccm.156.3.9606031

C. Roduit, R. Frei, M. Depner, B. Schaub, G. Loss et al., Increased food diversity in the first year of life is inversely associated with allergic diseases, Journal of Allergy and Clinical Immunology, vol.133, issue.4, pp.1056-1064, 2014.
DOI : 10.1016/j.jaci.2013.12.1044

URL : https://hal.archives-ouvertes.fr/hal-01063983

R. J. Rona, T. Keil, C. Summers, D. Gislason, L. Zuidmeer et al., The prevalence of food allergy: A meta-analysis, Journal of Allergy and Clinical Immunology, vol.120, issue.3, 2007.
DOI : 10.1016/j.jaci.2007.05.026

P. Roy and R. Noad, Virus-like particles as a vaccine delivery system: myths and facts, 2009.
DOI : 10.1007/978-1-4419-1132-2_11

M. Salimi, J. L. Barlow, S. P. Saunders, L. Xue, D. Gutowska-owsiak et al., A role for IL-25 and IL-33???driven type-2 innate lymphoid cells in atopic dermatitis, The Journal of Experimental Medicine, vol.139, issue.13, pp.2939-2950, 2013.
DOI : 10.1038/ni.2208

I. Schöll, E. Untersmayr, N. Bakos, F. Roth-walter, A. Gleiss et al., Antiulcer drugs promote oral sensitization and hypersensitivity to hazelnut allergens in BALB/c mice and humans. Am, J. Clin. Nutr, vol.81, pp.154-160, 2005.

G. Senti, S. Von-moos, F. Tay, N. Graf, P. Johansen et al., Determinants of efficacy and safety in epicutaneous allergen immunotherapy: summary of three clinical trials, Allergy, vol.127, issue.6, pp.707-710, 2015.
DOI : 10.1016/j.jaci.2011.02.016

E. M. Shevach and A. M. Thornton, tTregs, pTregs, and iTregs: similarities and differences, Immunological Reviews, vol.35, issue.1, pp.88-102, 2014.
DOI : 10.2337/dc12-0038

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3982187

S. H. Sicherer, Epidemiology of food allergy, Journal of Allergy and Clinical Immunology, vol.127, issue.3, pp.594-602, 2011.
DOI : 10.1016/j.jaci.2010.11.044

S. H. Sicherer and H. A. Sampson, Food allergy: Epidemiology, pathogenesis, diagnosis, and treatment, Journal of Allergy and Clinical Immunology, vol.133, issue.2, pp.291-307, 2014.
DOI : 10.1016/j.jaci.2013.11.020

M. C. Siracusa, S. A. Saenz, D. A. Hill, B. S. Kim, M. B. Headley et al., TSLP promotes interleukin-3-independent basophil haematopoiesis and type 2 inflammation, Nature, vol.181, issue.7363, pp.229-233, 2011.
DOI : 10.4049/jimmunol.181.7.4709

P. L. Smaldini, M. L. Orsini-delgado, C. A. Fossati, and G. H. Docena, Orally-Induced Intestinal CD4+ CD25+ FoxP3+ Treg Controlled Undesired Responses towards Oral Antigens and Effectively Dampened Food Allergic Reactions, PLOS ONE, vol.153, issue.1, 2015.
DOI : 10.1371/journal.pone.0141116.t001

URL : http://doi.org/10.1371/journal.pone.0141116

C. L. Sokol, G. M. Barton, A. G. Farr, and R. Medzhitov, A mechanism for the initiation of allergen-induced T helper type 2 responses, Nature Immunology, vol.444, issue.3, pp.310-318, 2008.
DOI : 10.4049/jimmunol.165.7.3620

V. V. Temchura, M. Tenbusch, G. Nchinda, G. Nabi, B. Tippler et al., Enhancement of immunostimulatory properties of exosomal vaccines by incorporation of fusion-competent G protein of vesicular stomatitis virus, Vaccine, vol.26, issue.29-30, pp.3662-3672, 2008.
DOI : 10.1016/j.vaccine.2008.04.069

T. C. Theoharides, K. Alysandratos, A. Angelidou, D. Delivanis, N. Sismanopoulos et al., Mast cells and inflammation, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, vol.1822, issue.1, 2012.
DOI : 10.1016/j.bbadis.2010.12.014

J. Travers and M. E. Rothenberg, Eosinophils in mucosal immune responses, Mucosal Immunology, vol.92, issue.3, pp.464-475, 2015.
DOI : 10.1016/j.jaci.2013.11.041

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4476057

R. Troncone, N. Caputo, G. Florio, and E. Finelli, Increased intestinal sugar permeability after challenge in children with cow's milk allergy or intolerance., Allergy, vol.348, issue.3, pp.142-146, 1994.
DOI : 10.1111/j.1365-2222.1986.tb00764.x

H. Turner and J. P. Kinet, Signalling through the high-affinity IgE receptor Fc??RI, Nature, vol.402, issue.6760supp, pp.24-30, 1999.
DOI : 10.1038/35037021

E. Untersmayr, I. Schöll, I. Swoboda, W. J. Beil, E. Förster-waldl et al., Antacid medication inhibits digestion of dietary proteins and causes food allergy A fish allergy model in balb/c mice, Journal of Allergy and Clinical Immunology, vol.112, issue.3, pp.616-623, 2003.
DOI : 10.1016/S0091-6749(03)01719-6

M. Vazquez-ortiz, M. Alvaro, M. Piquer, O. Dominguez, A. Machinena et al., Baseline specific IgE levels are useful to predict safety of oral immunotherapy in egg-allergic children, Clinical & Experimental Allergy, vol.59, issue.1, pp.130-141, 2014.
DOI : 10.1111/j.1398-9995.2004.00466.x

A. Vereda, M. Van-hage, S. Ahlstedt, M. D. Ibañez, J. Cuesta-herranz et al., Peanut allergy: Clinical and immunologic differences among patients from 3 different geographic regions, Journal of Allergy and Clinical Immunology, vol.127, issue.3, pp.603-607, 2011.
DOI : 10.1016/j.jaci.2010.09.010

J. Wang and H. A. Sampson, Food allergy, Journal of Clinical Investigation, vol.121, issue.3, pp.827-835, 2011.
DOI : 10.1172/JCI45434

URL : https://hal.archives-ouvertes.fr/hal-01262509

F. Van-wijk, S. Hoeks, S. Nierkens, S. J. Koppelman, P. Van-kooten et al., CTLA-4 Signaling Regulates the Intensity of Hypersensitivity Responses to Food Antigens, but is Not Decisive in the Induction of Sensitization, The Journal of Immunology, vol.174, issue.1, pp.174-179, 1950.
DOI : 10.4049/jimmunol.174.1.174

K. Wing, Y. Onishi, P. Prieto-martin, T. Yamaguchi, M. Miyara et al., CTLA-4 Control over Foxp3+ Regulatory T Cell Function, Science, vol.105, issue.29, pp.271-275, 2008.
DOI : 10.1073/pnas.0711106105

T. Worbs, U. Bode, S. Yan, M. W. Hoffmann, G. Hintzen et al., Oral tolerance originates in the intestinal immune system and relies on antigen carriage by dendritic cells, The Journal of Experimental Medicine, vol.56, issue.3, pp.519-527, 2006.
DOI : 10.1126/science.1091334

C. Xiao, S. M. Puddicombe, S. Field, J. Haywood, V. Broughton-head et al., Defective epithelial barrier function in asthma, Journal of Allergy and Clinical Immunology, vol.128, issue.3, pp.549-556, 2011.
DOI : 10.1016/j.jaci.2011.05.038

H. Yamashita, K. Takahashi, H. Tanaka, H. Nagai, and N. Inagaki, Overcoming food allergy through acquired tolerance conferred by transfer of Tregs in a murine model, Allergy, vol.123, issue.2, pp.201-209, 2012.
DOI : 10.1016/j.jaci.2008.09.051

J. Zhu, J. Cote-sierra, L. Guo, P. , and W. E. , Stat5 Activation Plays a Critical Role in Th2 Differentiation, Immunity, vol.19, issue.5, pp.739-748, 2003.
DOI : 10.1016/S1074-7613(03)00292-9

URL : http://doi.org/10.1016/s1074-7613(03)00292-9

J. Zhu, B. Min, J. Hu-li, C. J. Watson, A. Grinberg et al., Conditional deletion of Gata3 shows its essential function in TH1-TH2 responses, Conditional deletion of Gata3 shows its essential function in T(H)1-T(H)2 responses, pp.1157-1165, 2004.
DOI : 10.1016/S1074-7613(01)00084-X