K. Ashjaei, Differential T-helper cell polarization after allergen-specific stimulation of autologous dendritic cells in polysensitized allergic patients, International Archives of Allergy and Immunology, vol.166, issue.2, pp.97-106, 2015.

C. Asselin-paturel, Mouse type I IFN-producing cells are immature APCs with plasmacytoid morphology, Nat Immunol, vol.2, issue.12, pp.1144-1150, 2001.

F. Awad, Inflammasome biology, molecular pathology and therapeutic implications, Pharmacol Ther, 2018.

M. F. Bachmann, M. Kopf, and B. J. Marsland, Chemokines: more than just road signs, Nat Rev Immunol, vol.6, issue.2, pp.159-164, 2006.

V. P. Badovinac and J. T. Harty, Programming, demarcating, and manipulating CD8+ T-cell memory, Immunol Rev, vol.211, pp.67-80, 2006.

L. Baird and A. T. Dinkova-kostova, The cytoprotective role of the Keap1-Nrf2 pathway, Arch Toxicol, vol.85, issue.4, pp.241-272, 2011.

M. Bajenoff, S. Granjeaud, and S. Guerder, The strategy of T cell antigen-presenting cell encounter in antigen-draining lymph nodes revealed by imaging of initial T cell activation, J Exp Med, vol.198, issue.5, pp.715-724, 2003.

G. Bakdash, The nature of activatory and tolerogenic dendritic cell-derived signal II, Front Immunol, vol.4, p.53, 2013.

M. Ban, Simultaneous analysis of the local and systemic immune responses in mice to study the occupational asthma mechanisms induced by chromium and platinum, Toxicology, vol.277, issue.1-3, pp.29-37, 2010.

J. Banchereau, Immunobiology of dendritic cells, Annu Rev Immunol, vol.18, pp.767-811, 2000.

J. Banchereau and R. M. Steinman, Dendritic cells and the control of immunity, Nature, vol.392, issue.6673, pp.245-252, 1998.

G. M. Barton and J. C. Kagan, A cell biological view of Toll-like receptor function: regulation through compartmentalization, Nat Rev Immunol, vol.9, issue.8, pp.535-542, 2009.

D. A. Basketter, Local lymph node assay -validation, conduct and use in practice, Food Chem Toxicol, vol.40, issue.5, pp.593-598, 2002.

D. A. Basketter, Threshold for classification as a skin sensitizer in the local lymph node assay: a statistical evaluation, Food Chem Toxicol, vol.37, issue.12, pp.1167-1174, 1999.

D. A. Basketter, Nothing is perfect, not even the local lymph node assay: a commentary and the implications for REACH, Contact Dermatitis, vol.60, issue.2, pp.65-69, 2009.

R. Basu, Th22 cells are an important source of IL-22 for host protection against enteropathogenic bacteria, Immunity, vol.37, issue.6, pp.1061-1075, 2012.

F. Battais, In vitro detection of chemical allergens: an optimized assay using mouse bone marrow-derived dendritic cells, Contact Dermatitis, vol.77, issue.5, pp.311-322, 2017.

I. Bellinghausen, Importance of the inducible costimulator molecule for the induction of allergic immune responses and its decreased expression on T helper cells after venom immunotherapy, Immunology, vol.112, issue.1, pp.80-86, 2004.

I. Bellinghausen, Regulatory activity of human CD4 CD25 T cells depends on allergen concentration, type of allergen and atopy status of the donor, Immunology, vol.116, issue.1, pp.103-111, 2005.

C. Benezra, A systematic search for structure-activity relationships of skin contact sensitizers: methodology, J Invest Dermatol, vol.85, issue.4, pp.351-356, 1985.

F. Berard, J. P. Marty, and J. F. Nicolas, Allergen penetration through the skin, Eur J Dermatol, vol.13, issue.4, pp.324-330, 2003.

J. C. Bessot, G. Pauli, and O. Vandenplas, Phosphorylation of Nrf2 at Ser40 by protein kinase C in response to antioxidants leads to the release of Nrf2 from INrf2, but is not required for Nrf2 stabilization/accumulation in the nucleus and transcriptional activation of antioxidant response element-mediated NAD(P)H:quinone oxidoreductase-1 gene expression, J Biol Chem, vol.278, issue.45, pp.44675-44682, 2003.

F. Boisleve, S. Kerdine-romer, and M. Pallardy, Implication of the MAPK pathways in the maturation of human dendritic cells induced by nickel and TNF-alpha, Toxicology, vol.206, issue.2, pp.233-244, 2005.

F. Boisleve, S. Kerdine-romer, N. Rougier-larzat, and M. Pallardy, Nickel and DNCB induce CCR7 expression on human dendritic cells through different signalling pathways: role of TNFalpha and MAPK, J Invest Dermatol, vol.123, issue.3, pp.494-502, 2004.

M. Bonmort, Killer dendritic cells: IKDC and the others, Curr Opin Immunol, vol.20, issue.5, pp.558-565, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00294468

O. Boyman, S. Letourneau, C. Krieg, and J. Sprent, Homeostatic proliferation and survival of naive and memory T cells, Eur J Immunol, vol.39, issue.8, pp.2088-2094, 2009.

D. Breitfeld, Follicular B helper T cells express CXC chemokine receptor 5, localize to B cell follicles, and support immunoglobulin production, J Exp Med, vol.192, issue.11, pp.1545-1552, 2000.

M. Breloer and B. Fleischer, CD83 regulates lymphocyte maturation, activation and homeostasis, Trends Immunol, vol.29, issue.4, pp.186-194, 2008.

R. Brigelius-flohe and L. Flohe, Basic principles and emerging concepts in the redox control of transcription factors, Antioxid Redox Signal, vol.15, issue.8, pp.2335-2381, 2011.

H. K. Bryan, A. Olayanju, C. E. Goldring, and B. K. Park, The Nrf2 cell defence pathway: Keap1-dependent and -independent mechanisms of regulation, Biochem Pharmacol, vol.85, issue.6, pp.705-717, 2013.

E. V. Buehler, Delayed Contact Hypersensitivity in the Guinea Pig, Arch Dermatol, vol.91, pp.171-177, 1965.

D. Byamba, The Roles of Reactive Oxygen Species Produced by Contact Allergens and Irritants in Monocyte-derived Dendritic Cells, Ann Dermatol, vol.22, issue.3, pp.269-278, 2010.

Y. Cai, Pivotal role of dermal IL-17-producing gammadelta T cells in skin inflammation, Immunity, vol.35, issue.4, pp.596-610, 2011.

X. Cao, CD1 molecules efficiently present antigen in immature dendritic cells and traffic independently of MHC class II during dendritic cell maturation, J Immunol, vol.169, issue.9, pp.4770-4777, 2002.

A. Caruso, Flow cytometric analysis of activation markers on stimulated T cells and their correlation with cell proliferation, Cytometry, vol.27, issue.1, pp.71-76, 1997.

S. Casati, Integrated Approaches to Testing and Assessment, Basic Clin Pharmacol Toxicol, 2018.

S. Casati, Selection of chemicals for the development and evaluation of in vitro methods for skin sensitisation testing, Altern Lab Anim, vol.37, issue.3, pp.305-312, 2009.

S. Casati, A. Worth, P. Sven, and M. Whelan, EURL ECVAM Strategy for Replacement of Animal Testing for Skin Sensitisation Hazard Identification and Classification, 2013.

M. Cebrian, Triggering of T cell proliferation through AIM, an activation inducer molecule expressed on activated human lymphocytes, J Exp Med, vol.168, issue.5, pp.1621-1637, 1988.

M. Cella, Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I interferon, Nat Med, vol.5, issue.8, pp.919-923, 1999.

K. Chan and Y. W. Kan, Nrf2 is essential for protection against acute pulmonary injury in mice, Proc Natl Acad Sci U S A, vol.96, issue.22, pp.12731-12736, 1999.

A. Chary, Respiratory sensitization: toxicological point of view on the available assays, Arch Toxicol, vol.92, issue.2, pp.803-822, 2018.

A. I. Chen, Ox40-ligand has a critical costimulatory role in dendritic cell:T cell interactions, Immunity, vol.11, issue.6, pp.689-698, 1999.

L. Chen and D. B. Flies, Molecular mechanisms of T cell co-stimulation and co-inhibition, Nat Rev Immunol, vol.13, issue.4, pp.227-242, 2013.

X. Chen, J. J. Oppenheim, and O. M. Howard, BALB/c mice have more CD4+CD25+ T regulatory cells and show greater susceptibility to suppression of their CD4+CD25-responder T cells than C57BL/6 mice, Journal of Leukocyte Biology, vol.78, issue.1, pp.114-121, 2005.

C. Cheong, Microbial stimulation fully differentiates monocytes to DC-SIGN/CD209(+) dendritic cells for immune T cell areas, Cell, vol.143, issue.3, pp.416-429, 2010.

I. Ferreira, Nature and kinetics of redox imbalance triggered by respiratory and skin chemical sensitizers on the human monocytic cell line THP-1, Redox Biol, vol.16, pp.75-86, 2018.

V. Finger, I. Sari-minodier, and D. Charpin, Epidémiologie. L'asthme Professionnel. M. Orange, pp.13-30, 2012.

J. M. Fletcher, Cytomegalovirus-specific CD4+ T cells in healthy carriers are continuously driven to replicative exhaustion, J Immunol, vol.175, issue.12, pp.8218-8225, 2005.

R. Forster, A. Braun, and T. Worbs, Lymph node homing of T cells and dendritic cells via afferent lymphatics, Trends Immunol, vol.33, issue.6, pp.271-280, 2012.

L. Franchi, N. Warner, K. Viani, and G. Nunez, Function of Nod-like receptors in microbial recognition and host defense, Immunol Rev, vol.227, pp.106-128, 2009.

J. Frombach, Lymphocyte surface markers and cytokines are suitable for detection and potency assessment of skin-sensitizing chemicals in an in vitro model of allergic contact dermatitis: the LCSA-ly, Arch Toxicol, vol.92, issue.4, pp.1495-1505, 2018.

R. E. Fruge, Real-time visualization of macromolecule uptake by epidermal Langerhans cells in living animals, J Invest Dermatol, vol.132, pp.609-614, 2012.

M. Fujita, Development of a prediction method for skin sensitization using novel cysteine and lysine derivatives, J Pharmacol Toxicol Methods, vol.70, issue.1, pp.94-105, 2014.

K. Gamerdinger, A new type of metal recognition by human T cells: contact residues for peptide-independent bridging of T cell receptor and major histocompatibility complex by nickel, J Exp Med, vol.197, issue.10, pp.1345-1353, 2003.

J. J. Garcia-vallejo and Y. Van-kooyk, The physiological role of DC-SIGN: a tale of mice and men, Trends Immunol, vol.34, issue.10, pp.482-486, 2013.

F. Geissmann, Development of monocytes, macrophages, and dendritic cells, Science, vol.327, issue.5966, pp.656-661, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00502972

G. F. Gerberick, Selective modulation of T cell memory markers CD62L and CD44 on murine draining lymph node cells following allergen and irritant treatment, Toxicol Appl Pharmacol, vol.146, issue.1, pp.1-10, 1997.

F. Gerberick, Chemical reactivity measurement and the predicitve identification of skin sensitisers. The report and recommendations of ECVAM Workshop 64, Altern Lab Anim, vol.36, issue.2, pp.215-242, 2008.

G. F. Gerberick, The use of peptide reactivity assays for skin sensitisation hazard identification and risk assessment, Altern Lab Anim, vol.44, issue.5, pp.437-442, 2016.

G. F. Gerberick, A chemical dataset for evaluation of alternative approaches to skinsensitization testing, Contact Dermatitis, vol.50, issue.5, pp.274-288, 2004.

G. F. Gerberick, Investigation of peptide reactivity of pro-hapten skin sensitizers using a peroxidase-peroxide oxidation system, Toxicol Sci, vol.112, issue.1, pp.164-174, 2009.

G. F. Gerberick, Development of a peptide reactivity assay for screening contact allergens, Toxicol Sci, vol.81, issue.2, pp.332-343, 2004.

G. F. Gerberick, Quantification of chemical peptide reactivity for screening contact allergens: a classification tree model approach, Toxicol Sci, vol.97, issue.2, pp.417-427, 2007.

B. Gerritsen and A. Pandit, The memory of a killer T cell: models of CD8(+) T cell differentiation, Immunol Cell Biol, vol.94, issue.3, pp.236-241, 2016.

A. Gibson, In Vitro Priming of Naive T-cells with p-Phenylenediamine and Bandrowski's Base, Chem Res Toxicol, vol.28, issue.10, pp.2069-2077, 2015.

C. E. Goldring, Activation of hepatic Nrf2 in vivo by acetaminophen in CD-1 mice, Hepatology, vol.39, issue.5, pp.1267-1276, 2004.

A. Goossens and J. P. Lepoittevin, Contact allergy to cosmetics and their perfume components: new clinical, chemical and diagnostic aspects, Revue Francaise D'Allergologie, vol.43, issue.5, pp.294-300, 2003.

A. V. Gorbachev and R. L. Fairchild, Induction and regulation of T-cell priming for contact hypersensitivity, Crit Rev Immunol, vol.21, issue.5, pp.451-472, 2001.

M. Goutet, Identification of contact and respiratory sensitizers using flow cytometry, Toxicol Appl Pharmacol, vol.205, issue.3, pp.259-270, 2005.

M. Goutet, Identification of contact and respiratory sensitizers according to IL-4 receptor alpha expression and IL-2 production, Toxicol Appl Pharmacol, vol.260, issue.2, pp.95-104, 2012.

E. Gros and N. Novak, Cutaneous dendritic cells in allergic inflammation, Clinical and Experimental Allergy, vol.42, issue.8, pp.1161-1175, 2012.

G. Grouard, The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand, J Exp Med, vol.185, issue.6, pp.1101-1111, 1997.

H. Hadeiba, CCR9 expression defines tolerogenic plasmacytoid dendritic cells able to suppress acute graft-versus-host disease, Nat Immunol, vol.9, issue.11, pp.1253-1260, 2008.

H. Hammad and B. N. Lambrecht, Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma, Nat Rev Immunol, vol.8, issue.3, pp.193-204, 2008.

H. Hammad and B. N. Lambrecht, Dendritic cells and airway epithelial cells at the interface between innate and adaptive immune responses, Allergy, vol.66, issue.5, pp.579-587, 2011.

W. W. Hancock, R. Buelow, M. H. Sayegh, and L. A. Turka, Antibody-induced transplant arteriosclerosis is prevented by graft expression of anti-oxidant and anti-apoptotic genes, Nat Med, vol.4, issue.12, pp.1392-1396, 1998.

S. E. Hardison and G. D. Brown, C-type lectin receptors orchestrate antifungal immunity, Nat Immunol, vol.13, issue.9, pp.817-822, 2012.

L. E. Harrington, Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages, Nat Immunol, vol.6, issue.11, pp.1123-1132, 2005.

L. A. Harshyne, S. C. Watkins, A. Gambotto, and S. M. Barratt-boyes, Dendritic cells acquire antigens from live cells for cross-presentation to CTL, J Immunol, vol.166, issue.6, pp.3717-3723, 2001.

J. T. Harty and V. P. Badovinac, Shaping and reshaping CD8+ T-cell memory, Nat Rev Immunol, vol.8, issue.2, pp.107-119, 2008.

O. Hausmann, B. Schnyder, and W. J. Pichler, Etiology and pathogenesis of adverse drug reactions, Chem Immunol Allergy, vol.97, pp.32-46, 2012.

B. F. Haynes, CD44 a molecule involved in leukocyte adherence and T-cell activation, Immunol Today, vol.10, issue.12, pp.423-431, 1989.

S. Henri, The dendritic cell populations of mouse lymph nodes, J Immunol, vol.167, issue.2, pp.741-748, 2001.

S. E. Henrickson, T cell sensing of antigen dose governs interactive behavior with dendritic cells and sets a threshold for T cell activation, Nat Immunol, vol.9, issue.3, pp.282-291, 2008.

S. T. Holgate, The sentinel role of the airway epithelium in asthma pathogenesis, Immunol Rev, vol.242, issue.1, pp.205-219, 2011.

S. T. Holgate, Innate and adaptive immune responses in asthma, Nat Med, vol.18, issue.5, pp.673-683, 2012.

U. E. Hopken, The ratio between dendritic cells and T cells determines the outcome of their encounter: proliferation versus deletion, Eur J Immunol, vol.35, issue.10, pp.2851-2863, 2005.

N. A. Hosken, The effect of antigen dose on CD4+ T helper cell phenotype development in a T cell receptor-alpha beta-transgenic model, J Exp Med, vol.182, issue.5, pp.1579-1584, 1995.

H. Huang, Mite allergen decreases DC-SIGN expression and modulates human dendritic cell differentiation and function in allergic asthma, Mucosal Immunology, vol.4, issue.5, pp.519-527, 2011.

H. C. Huang, T. Nguyen, and C. B. Pickett, Phosphorylation of Nrf2 at Ser-40 by protein kinase C regulates antioxidant response element-mediated transcription, J Biol Chem, vol.277, issue.45, pp.42769-42774, 2002.

M. Huber and M. Lohoff, Change of paradigm: CD8+ T cells as important helper for CD4+ T cells during asthma and autoimmune encephalomyelitis, Allergo J Int, vol.24, issue.1, pp.8-15, 2015.

J. B. Huppa and M. M. Davis, T-cell-antigen recognition and the immunological synapse, Nat Rev Immunol, vol.3, issue.12, pp.973-983, 2003.

K. M. Huster, Unidirectional development of CD8+ central memory T cells into protective Listeria-specific effector memory T cells, Eur J Immunol, vol.36, issue.6, pp.1453-1464, 2006.

P. T. Illing, Immune self-reactivity triggered by drug-modified HLA-peptide repertoire, Nature, vol.486, issue.7404, pp.554-558, 2012.

K. Inaba, Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor, J Exp Med, vol.176, issue.6, pp.1693-1702, 1992.

K. Inaba, The tissue distribution of the B7-2 costimulator in mice: abundant expression on dendritic cells in situ and during maturation in vitro, J Exp Med, vol.180, issue.5, pp.1849-1860, 1994.

A. Iwasaki and R. Medzhitov, Toll-like receptor control of the adaptive immune responses, Nat Immunol, vol.5, issue.10, pp.987-995, 2004.

C. Janeway, Immunobiologie. Bruxelles, 2009.

L. Jia and C. Wu, The biology and functions of Th22 cells, Adv Exp Med Biol, vol.841, pp.209-230, 2014.

L. Jia and C. Wu, Differentiation, regulation and function of Th9 cells, Adv Exp Med Biol, vol.841, pp.181-207, 2014.

J. D. Johansen and T. Menne, The fragrance mix and its constituents: a 14-year material, Contact Dermatitis, vol.32, issue.1, pp.18-23, 1995.

H. Johansson, A. S. Albrekt, C. A. Borrebaeck, and M. Lindstedt, The GARD assay for assessment of chemical skin sensitizers, Toxicol In Vitro, vol.27, issue.3, pp.1163-1169, 2013.

S. G. Johansson, A revised nomenclature for allergy. An EAACI position statement from the EAACI nomenclature task force, Allergy, vol.56, issue.9, pp.813-824, 2001.

S. M. Kaech, E. J. Wherry, and R. Ahmed, Effector and memory T-cell differentiation: implications for vaccine development, Nat Rev Immunol, vol.2, issue.4, pp.251-262, 2002.

A. T. Kamath, The development, maturation, and turnover rate of mouse spleen dendritic cell populations, J Immunol, vol.165, issue.12, pp.6762-6770, 2000.

D. H. Kaplan, B. Z. Igyarto, and A. A. Gaspari, Early immune events in the induction of allergic contact dermatitis, Nat Rev Immunol, vol.12, issue.2, pp.114-124, 2012.

A. T. Karlberg, Allergic contact dermatitis--formation, structural requirements, and reactivity of skin sensitizers, Chem Res Toxicol, vol.21, issue.1, pp.53-69, 2008.

A. T. Karlberg, Activation of non-sensitizing or low-sensitizing fragrance substances into potent sensitizers -prehaptens and prohaptens, Contact Dermatitis, vol.69, issue.6, pp.323-334, 2013.

T. Kawai and S. Akira, The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors, Nat Immunol, vol.11, issue.5, pp.373-384, 2010.

T. W. Kensler, N. Wakabayashi, and S. Biswal, Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway, Annu Rev Pharmacol Toxicol, vol.47, pp.89-116, 2007.

D. H. Kim, Different characteristics of reactive oxygen species production by human keratinocyte cell line cells in response to allergens and irritants, Exp Dermatol, vol.21, issue.2, pp.99-103, 2012.

D. S. Kim, Immunotherapy of malignant melanoma with tumor lysate-pulsed autologous monocyte-derived dendritic cells, Yonsei Med J, vol.52, issue.6, pp.990-998, 2011.

H. K. Kim, High-level expression of B7-H1 molecules by dendritic cells suppresses the function of activated T cells and desensitizes allergen-primed animals, J Leukoc Biol, vol.79, issue.4, pp.686-695, 2006.

I. Kimber, R. J. Dearman, and D. A. Basketter, Diisocyanates, occupational asthma and IgE antibody: implications for hazard characterization, J Appl Toxicol, vol.34, issue.10, pp.1073-1077, 2014.

I. Kimber, R. J. Dearman, D. A. Basketter, and D. R. Boverhof, Chemical respiratory allergy: reverse engineering an adverse outcome pathway, Toxicology, vol.318, pp.32-39, 2014.

I. Kimber, The local lymph node assay: past, present and future, Contact Dermatitis, vol.47, issue.6, pp.315-328, 2002.

I. Kimber, Allergic contact dermatitis: a commentary on the relationship between T lymphocytes and skin sensitising potency, Toxicology, vol.291, issue.1-3, pp.18-24, 2012.

I. Kimber and M. Pallardy, The use of T cells in hazard characterization of chemical and drug allergens and integration in testin strategies, EXS, vol.104, pp.1-9, 2014.

C. King, New insights into the differentiation and function of T follicular helper cells, Nat Rev Immunol, vol.9, issue.11, pp.757-766, 2009.

Y. Kitagawa and S. Sakaguchi, Molecular control of regulatory T cell development and function, Curr Opin Immunol, vol.49, pp.64-70, 2017.

I. Kryczek, FOXP3 defines regulatory T cells in human tumor and autoimmune disease, Cancer Res, vol.69, issue.9, pp.3995-4000, 2009.

D. A. Kulpa, The immunological synapse: the gateway to the HIV reservoir, Immunol Rev, vol.254, issue.1, pp.305-325, 2013.

C. Lahoute, Z. O.-herbin, A. Mallat, and . Tedgui, Adaptive immunity in atherosclerosis: mechanisms and future therapeutic targets, Nat Rev Cardiol, vol.8, issue.6, pp.348-358, 2011.

J. F. Lalko, Reactivity of chemical respiratory allergens in the Peroxidase Peptide Reactivity Assay, Toxicol In Vitro, vol.27, issue.2, pp.651-661, 2013.

B. N. Lambrecht and H. Hammad, The role of dendritic and epithelial cells as master regulators of allergic airway inflammation, Lancet, vol.376, issue.9743, pp.835-843, 2010.

K. Landsteiner and J. Jacobs, Studies on the Sensitization of Animals with Simple Chemical Compounds, J Exp Med, vol.61, issue.5, pp.643-656, 1935.

K. Landsteiner and J. Jacobs, Studies on the Sensitization of Animals with Simple Chemical Compounds. Ii, J Exp Med, vol.64, issue.4, pp.625-639, 1936.

A. Lanzavecchia, Mechanisms of antigen uptake for presentation, Curr Opin Immunol, vol.8, issue.3, pp.348-354, 1996.

J. P. Lepoittevin, Development of structure-activity relationships (SARs) in allergic contact dermatitis, Cell Biol Toxicol, vol.15, issue.1, pp.47-55, 1999.

J. P. Lepoittevin, Metabolism versus chemical transformation or pro-versus prehaptens?, Contact Dermatitis, vol.54, issue.2, pp.73-74, 2006.

J. P. Lepoittevin, Les bases chimiques de l'allergie de contact, Revue Francaise d'Allergologie, vol.51, pp.208-211, 2011.

B. Leynaert, F. Neukirch, P. Demoly, and J. Bousquet, Epidemiologic evidence for asthma and rhinitis comorbidity, J Allergy Clin Immunol, vol.106, issue.5, pp.201-205, 2000.

Y. J. Li, Nrf2 is closely related to allergic airway inflammatory responses induced by lowdose diesel exhaust particles in mice, Clin Immunol, vol.137, issue.2, pp.234-241, 2010.

C. Louis-dit-sully, Activation of the TCR complex by peptide-MHC and superantigens, EXS, vol.104, pp.9-23, 2014.

S. E. Loveless, Potency values from the local lymph node assay: application to classification, labelling and risk assessment, Regul Toxicol Pharmacol, vol.56, issue.1, pp.54-66, 2010.

U. Luckey, T cell killing by tolerogenic dendritic cells protects mice from allergy, J Clin Invest, vol.121, issue.10, pp.3860-3871, 2011.

A. B. Lyons, S. J. Blake, and K. V. Doherty, Flow Cytometric Analysis of Cell Division by Dilution of CFSE and Related Dyes, Current Protocols in Cytometry, 2001.

Q. Ma and X. He, Molecular basis of electrophilic and oxidative defense: promises and perils of Nrf2, Pharmacol Rev, vol.64, issue.4, pp.1055-1081, 2012.

C. Mackay, M. Davies, V. Summerfield, and G. Maxwell, From pathways to people: applying the adverse outcome pathway (AOP) for skin sensitization to risk assessment, ALTEX, vol.30, issue.4, pp.473-486, 2013.

C. Macri, E. S. Pang, T. Patton, and M. O'keeffe, The three-dimensional structure of peptide-MHC complexes, Semin Cell Dev Biol. Madden, D. R, vol.13, pp.587-622, 1995.

B. Magnusson and A. M. Kligman, The identification of contact allergens by animal assay. The guinea pig maximization test, J Invest Dermatol, vol.52, issue.3, pp.268-276, 1969.

F. Malavasi, Evolution and function of the ADP ribosyl cyclase/CD38 gene family in physiology and pathology, Physiol Rev, vol.88, issue.3, pp.841-886, 2008.

J. L. Malo and O. Vandenplas, Definitions and classification of work-related asthma, Immunol Allergy Clin North Am, vol.31, issue.4, pp.645-662, 2011.

E. Martin, B. O'sullivan, P. Low, and R. Thomas, Antigen-specific suppression of a primed immune response by dendritic cells mediated by regulatory T cells secreting interleukin-10, Immunity, vol.18, issue.1, pp.155-167, 2003.

S. F. Martin, Allergic contact dermatitis: xenoinflammation of the skin, Curr Opin Immunol, vol.24, issue.6, pp.720-729, 2012.

S. F. Martin, Immunological mechanisms in allergic contact dermatitis, Curr Opin Allergy Clin Immunol, vol.15, issue.2, pp.124-130, 2015.

S. F. Martin, New concepts in cutaneous allergy, Contact Dermatitis, vol.72, issue.1, pp.2-10, 2015.

S. F. Martin, T-cell recognition of chemicals, protein allergens and drugs: towards the development of in vitro assays, Cell Mol Life Sci, vol.67, issue.24, pp.4171-4184, 2010.

S. F. Martin and T. Jakob, From innate to adaptive immune responses in contact hypersensitivity, Curr Opin Allergy Clin Immunol, vol.8, issue.4, pp.289-293, 2008.

L. Martinez-pomares, The mannose receptor, J Leukoc Biol, vol.92, issue.6, pp.1177-1186, 2012.

F. Martinon, Signaling by ROS drives inflammasome activation, Eur J Immunol, vol.40, issue.3, pp.616-619, 2010.

P. Matzinger, Tolerance, danger, and the extended family, Annu Rev Immunol, vol.12, pp.991-1045, 1994.

J. C. Mbongue, H. A. Nieves, T. W. Torrez, and W. H. Langridge, The Role of Dendritic Cell Maturation in the Induction of Insulin-Dependent Diabetes Mellitus, Front Immunol, vol.8, p.327, 2017.

R. Mcclure and P. Massari, TLRDependent Human Mucosal Epithelial Cell Responses to Microbial Pathogens, Front Immunol, vol.5, p.386, 2014.

O. Mekenyan, A mechanistic approach to modeling respiratory sensitization, Chem Res Toxicol, vol.27, issue.2, pp.219-239, 2014.

T. R. Mempel, S. E. Henrickson, and U. H. Von-andrian, T-cell priming by dendritic cells in lymph nodes occurs in three distinct phases, Nature, vol.427, issue.6970, pp.154-159, 2004.

M. Merad, F. Ginhoux, and M. Collin, Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells, Nat Rev Immunol, vol.8, issue.12, pp.935-947, 2008.

M. Merad, The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting, Annu Rev Immunol, vol.31, pp.563-604, 2013.

F. Merckel, E. Gimenez-arnau, G. F. Gerberick, and J. P. Lepoittevin, )C)methylphenol in the peroxidase peptide reactivity assay (PPRA) as an alternative to animal testing, Toxicol Lett, vol.218, issue.1, pp.266-272, 2013.

C. Migdal, Reactivity of chemical sensitizers toward amino acids in cellulo plays a role in the activation of the Nrf2-ARE pathway in human monocyte dendritic cells and the THP-1 cell line, Toxicol Sci, 2013.

H. W. Mittrucker, A. Visekruna, and M. Huber, Heterogeneity in the differentiation and function of CD8(+) T cells, Arch Immunol Ther Exp (Warsz), vol.62, issue.6, pp.449-458, 2014.

P. Moi, Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the beta-globin locus control region, Proc Natl Acad Sci U S A, vol.91, issue.21, pp.9926-9930, 1994.

M. E. Monzon, Reactive oxygen species and hyaluronidase 2 regulate airway epithelial hyaluronan fragmentation, J Biol Chem, vol.285, issue.34, pp.26126-26134, 2010.

G. Moscato, EAACI position paper on occupational rhinitis, Respir Res, vol.10, p.16, 2009.

S. N. Mueller, T. Gebhardt, F. R. Carbone, and W. R. Heath, Memory T cell subsets, migration patterns, and tissue residence, Annu Rev Immunol, vol.31, pp.137-161, 2013.

F. Mussotter, Proteomics analysis of dendritic cell activation by contact allergens reveals possible biomarkers regulated by Nrf2, Toxicol Appl Pharmacol, vol.313, pp.170-179, 2016.

J. Muto, Hyaluronan digestion controls DC migration from the skin, J Clin Invest, vol.124, issue.3, pp.1309-1319, 2014.

G. Nace, J. Evankovich, R. Eid, and A. Tsung, Dendritic cells and damage-associated molecular patterns: endogenous danger signals linking innate and adaptive immunity, J Innate Immun, vol.4, issue.1, pp.6-15, 2012.

S. H. Naik, L. M. Corcoran, and L. Wu, Development of murine plasmacytoid dendritic cell subsets, Immunol Cell Biol, vol.83, issue.5, pp.563-570, 2005.

S. H. Naik, Intrasplenic steady-state dendritic cell precursors that are distinct from monocytes, Nat Immunol, vol.7, issue.6, pp.663-671, 2006.

S. H. Naik, CD8+, CD8-, and plasmacytoid dendritic cell generation in vitro using flt3 ligand, Methods Mol Biol, vol.595, pp.167-176, 2010.

T. Nakamura, Covalent modification of lysine residues by allyl isothiocyanate in physiological conditions: plausible transformation of isothiocyanate from thiol to amine, Chem Res Toxicol, vol.22, issue.3, pp.536-542, 2009.

H. Nakano, Blood-derived inflammatory dendritic cells in lymph nodes stimulate acute T helper type 1 immune responses, Nat Immunol, vol.10, issue.4, pp.394-402, 2009.

M. Nakayama, Antigen Presentation by MHC-Dressed Cells, Front Immunol, vol.5, p.672, 2014.

K. Narita, Improvement of human cell line activation test (h-CLAT) using short-time exposure methods for prevention of false-negative results, The Journal of Toxicological Sciences, vol.43, issue.3, pp.229-240, 2018.

A. Natsch, The Nrf2-Keap1-ARE toxicity pathway as a cellular sensor for skin sensitizers--functional relevance and a hypothesis on innate reactions to skin sensitizers, Toxicol Sci, vol.113, issue.2, pp.284-292, 2010.

A. Natsch, The intra-and inter-laboratory reproducibility and predictivity of the KeratinoSens assay to predict skin sensitizers in vitro: results of a ring-study in five laboratories, Toxicol In Vitro, vol.25, issue.3, pp.733-744, 2011.

A. Natsch and R. Emter, Skin sensitizers induce antioxidant response element dependent genes: application to the in vitro testing of the sensitization potential of chemicals, Toxicol Sci, vol.102, issue.1, pp.110-119, 2008.

A. Natsch and R. Emter, Nrf2 activation as a key event triggered by skin sensitisers: The development of the stable KeratinoSens reporter gene assay, Altern Lab Anim, vol.44, issue.5, pp.443-451, 2016.

J. Neefjes, M. L. Jongsma, P. Paul, and O. Bakke, Towards a systems understanding of MHC class I and MHC class II antigen presentation, Nat Rev Immunol, vol.11, issue.12, pp.823-836, 2011.

F. O. Nestle, P. D. Meglio, J. Z. Qin, and B. J. Nickoloff, Skin immune sentinels in health and disease, Nat Rev Immunol, vol.9, issue.10, pp.679-691, 2009.

B. M. Neves, Signal transduction profile of chemical sensitisers in dendritic cells: an endpoint to be included in a cell-based in vitro alternative approach to hazard identification?, Toxicol Appl Pharmacol, vol.250, issue.2, pp.87-95, 2011.

K. Newton and V. M. Dixit, Signaling in innate immunity and inflammation, Cold Spring Harb Perspect Biol, vol.4, issue.3, 2012.

N. Nikolova-pavageau, Les manifestations et mécanismes des allergies professionnelles, Hygiène et Sécurité du Travail HST, vol.233, pp.23-25, 2013.

M. A. Norcross, Abacavir induces loading of novel self-peptides into HLA-B*57: 01: an autoimmune model for HLA-associated drug hypersensitivity, AIDS, vol.26, issue.11, pp.21-29, 2012.

Y. Nukada, Prediction of skin sensitization potency of chemicals by human Cell Line Activation Test (h-CLAT) and an attempt at classifying skin sensitization potency, Toxicol In Vitro, vol.26, issue.7, pp.1150-1160, 2012.

S. Nuriya, S. Enomoto, and M. Azuma, The role of CTLA-4 in murine contact hypersensitivity, J Invest Dermatol, vol.116, issue.5, pp.764-768, 2001.

M. O'keeffe, Mouse plasmacytoid cells: long-lived cells, heterogeneous in surface phenotype and function, that differentiate into CD8(+) dendritic cells only after microbial stimulus, J Exp Med, vol.196, issue.10, pp.1307-1319, 2002.

T. Oakes, The T Cell Response to the Contact Sensitizer Paraphenylenediamine Is Characterized by a Polyclonal Diverse Repertoire of Antigen-Specific Receptors, Frontiers in Immunology, vol.8, p.162, 2017.

. Oecd, Guidelines for Testing of Chemicals. Guideline No.406. Skin sensitisation, 1992.

. Oecd, Guidelines for Testing of Chemicals. Draft Guideline No. 429. Skin sensitisation: The Local Lymph Node Assay, 2001.

. Oecd, Test No. 442C: In Chemico Skin Sensitisation: Direct Peptide Reativity Assay (DPRA), 2015.

. Oecd, Test No. 442E: In Vitro Skin Sensitisation: Human Cell Line Activation Test (h-CLAT), 2016.

. Oecd, Test No. 442E: In Vitro Skin Sensitisation, 2017.

. Oecd, Test No. 442B: Skin Sensitization: local lymph node assay: BrdU-ELISA, 2010.

. Oecd, The Adverse Outcome Pathway for Skin Sensitisation Initiated by Covalent Binding to Proteins, 2014.

. Oecd, Test No. 442D: In Vitro Skin Sensitisation: ARE-Nrf2 Luciferase test method, 2015.

N. Ohkura and S. Sakaguchi, Regulatory T cells: roles of T cell receptor for their development and function, Semin Immunopathol, vol.32, issue.2, pp.95-106, 2010.

I. Okwor, P. Jia, and J. E. Uzonna, Interaction of Macrophage Antigen 1 and CD40 Ligand Leads to IL-12 Production and Resistance in CD40-Deficient Mice Infected with Leishmania major, The Journal of Immunology, vol.195, issue.7, pp.3218-3226, 2015.

E. B. Olasz, J. Linton, and S. I. Katz, Soluble proteins and haptens on bone marrow-derived dendritic cells are presented to host CD4 T cells in an MHC-restricted manner, Int Immunol, vol.14, issue.5, pp.493-502, 2002.

B. , Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12, Immunity, vol.13, issue.5, pp.715-725, 2000.

M. P. Oria and V. A. Stallings, Finding a Path to Safety in Food Allergy: Assessment of the Global Burden, Causes, Prevention, Management, and Public Policy, 2016.

W. O. Osburn, Increased colonic inflammatory injury and formation of aberrant crypt foci in Nrf2-deficient mice upon dextran sulfate treatment, Int J Cancer, vol.121, issue.9, pp.1883-1891, 2007.

D. A. Ostrov, Drug hypersensitivity caused by alteration of the MHC-presented selfpeptide repertoire, Proc Natl Acad Sci U S A, vol.109, issue.25, pp.9959-9964, 2012.

C. Paris, Work-related asthma in France: recent trends for the period, Occupational and Environmental Medicine, vol.69, issue.6, pp.391-397, 2001.
URL : https://hal.archives-ouvertes.fr/hal-01280170

H. Park, A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17, Nat Immunol, vol.6, issue.11, pp.1133-1141, 2005.

S. Patel, Inflammasomes, the cardinal pathology mediators are activated by pathogens, allergens and mutagens: A critical review with focus on NLRP3, Biomed Pharmacother, vol.92, pp.819-825, 2017.

G. Patlewicz, Can currently available non-animal methods detect pre and pro-haptens relevant for skin sensitization?, Regul Toxicol Pharmacol, vol.82, pp.147-155, 2016.

G. Patlewicz, TIMES-SS--a promising tool for the assessment of skin sensitization hazard. A characterization with respect to the OECD validation principles for (Q)SARs and an external evaluation for predictivity, Regul Toxicol Pharmacol, vol.48, issue.2, pp.225-239, 2007.

R. Pavlos, T cell-mediated hypersensitivity reactions to drugs, Annu Rev Med, vol.66, pp.439-454, 2015.

M. Peiser, M. Hitzler, and A. Luch, On the role of co-inhibitory molecules in dendritic cell: T helper cell coculture assays aimed to detect chemical-induced contact allergy, EXS, vol.104, pp.115-135, 2014.

M. Peiser, Allergic contact dermatitis: epidemiology, molecular mechanisms, in vitro methods and regulatory aspects Current knowledge assembled at an international workshop at BfR, Germany, Cellular and Molecular Life Sciences, vol.69, issue.5, pp.763-781, 2012.

V. Pena-cruz, PD-1 on immature and PD-1 ligands on migratory human Langerhans cells regulate antigen-presenting cell activity, J Invest Dermatol, vol.130, issue.9, pp.2222-2230, 2010.

E. Pepin, M. Goutet, and M. Ban, Murine bone marrow-derived dendritic cells as a potential in vitro model for predictive identification of chemical sensitizers, Toxicol Lett, vol.175, issue.1-3, pp.89-101, 2007.

P. Periasamy, J. K. Tan, and H. C. O'neill, Novel splenic antigen-presenting cells derive from a Lin(-) c-kit(lo) progenitor, Journal of Leukocyte Biology, vol.93, issue.1, pp.63-69, 2013.

M. Piccioni, Z. Chen, A. Tsun, and B. Li, Regulatory T-cell differentiation and their function in immune regulation, Adv Exp Med Biol, vol.841, pp.67-97, 2014.

W. J. Pichler, The p-i Concept: Pharmacological Interaction of Drugs With Immune Receptors, World Allergy Organ J, vol.1, issue.6, pp.96-102, 2008.

W. J. Pichler, Drug Hypersensitivity: How Drugs Stimulate T Cells via Pharmacological Interaction with Immune Receptors, Int Arch Allergy Immunol, vol.168, issue.1, pp.13-24, 2015.

W. J. Pichler, Pharmacological interaction of drugs with immune receptors: the p-i concept, Allergol Int, vol.55, issue.1, pp.17-25, 2006.

D. H. Pols, Atopic dermatitis, asthma and allergic rhinitis in general practice and the open population: a systematic review, Scand J Prim Health Care, vol.34, issue.2, pp.143-150, 2016.

J. M. Portnoy, P. B. Williams, and C. S. Barnes, Innate Immune Responses to Fungal Allergens, Curr Allergy Asthma Rep, vol.16, issue.9, p.62, 2016.

H. C. Probst, S. Muth, and H. Schild, Regulation of the tolerogenic function of steady-state DCs, Eur J Immunol, vol.44, issue.4, pp.927-933, 2014.

B. Pulendran, Flt3-ligand and granulocyte colony-stimulating factor mobilize distinct human dendritic cell subsets in vivo, J Immunol, vol.165, issue.1, pp.566-572, 2000.

F. Python, C. Goebel, and P. Aeby, Assessment of the U937 cell line for the detection of contact allergens, Toxicol Appl Pharmacol, vol.220, issue.2, pp.113-124, 2007.

H. Qi, Tfh cell differentiation and their function in promoting B-cell responses, Adv Exp Med Biol, vol.841, pp.153-180, 2014.

C. Raffalli, Editor's Highlight: Fragrance Allergens Linalool and Limonene Allylic Hydroperoxides in Skin Allergy: Mechanisms of Action Focusing on Transcription Factor Nrf2, Toxicological Sciences: An Official Journal of the Society of Toxicology, vol.161, issue.1, pp.139-148, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02322363

T. V. Rajan, The Gell-Coombs classification of hypersensitivity reactions: a re-interpretation, Trends Immunol, vol.24, issue.7, pp.376-379, 2003.

M. Ramos-gomez, Sensitivity to carcinogenesis is increased and chemoprotective efficacy of enzyme inducers is lost in nrf2 transcription factor-deficient mice, Proc Natl Acad Sci U S A, vol.98, issue.6, pp.3410-3415, 2001.

B. Reizis, Plasmacytoid dendritic cells: recent progress and open questions, Annu Rev Immunol, vol.29, pp.163-183, 2011.

S. Remy, Gene expressions changes in bronchial epithelial cells: markers for respiratory sensitizers and exploration of the NRF2 pathway, Toxicol In Vitro, vol.28, issue.2, pp.209-217, 2014.

J. Renaudin, Occupational allergy news, Revue Française d'Allergologie, vol.51, issue.3, pp.265-268, 2011.

J. Rengarajan, Interferon regulatory factor 4 (IRF4) interacts with NFATc2 to modulate interleukin 4 gene expression, J Exp Med, vol.195, issue.8, pp.1003-1012, 2002.

A. Richter, Human T cell priming assay (hTCPA) for the identification of contact allergens based on naive T cells and DC--IFN-gamma and TNF-alpha readout, Toxicol In Vitro, vol.27, issue.3, pp.1180-1185, 2013.

A. Rnv3p, Réseau national des pathologies professionnelles. Rapport scientifique, p.279, 2011.

D. W. Roberts, R. Fraginals, J. P. Lepoittevin, and C. Benezra, Refinement of the relative alkylation index (RAI) model for skin sensitization and application to mouse and guinea-pig test data for alkyl alkanesulphonates, Arch Dermatol Res, vol.283, issue.6, pp.387-394, 1991.

D. W. Roberts and D. L. Williams, The derivation of quantitative correlations between skin sensitisation and physio-chemical parameters for alkylating agents, and their application to experimental data for sultones, J Theor Biol, vol.99, issue.4, pp.807-825, 1982.

P. A. Roche and K. Furuta, The ins and outs of MHC class II-mediated antigen processing and presentation, Nat Rev Immunol, vol.15, issue.4, pp.203-216, 2015.

E. L. Roggen, In vitro approaches for detection of chemical sensitization, Basic Clin Pharmacol Toxicol, vol.115, issue.1, pp.32-40, 2014.

C. Rovida, Management of an integrated project (Sens-it-iv) to develop in vitro tests to assess sensitisation, Altern Lab Anim, vol.35, issue.3, pp.317-322, 2007.

S. Ruiz, P. E. Pergola, R. A. Zager, and N. D. Vaziri, Targeting the transcription factor Nrf2 to ameliorate oxidative stress and inflammation in chronic kidney disease, Kidney Int, vol.83, issue.6, pp.1029-1041, 2013.

W. F. Russell and . Jr, Combined drug treatment of tuberculosis. III. Clinical application of the principles of appropriate and adequate chemotherapy to the treatment of pulmonary tuberculosis, J Clin Invest, vol.38, issue.8, pp.1366-1375, 1959.

A. Saalbach, Fibroblasts support migration of monocyte-derived dendritic cells by secretion of PGE2 and MMP-1, Exp Dermatol, vol.24, issue.8, pp.598-604, 2015.

A. Saei and J. Hadjati, Tolerogenic dendritic cells: key regulators of peripheral tolerance in health and disease, Int Arch Allergy Immunol, vol.161, issue.4, pp.293-303, 2013.

P. Saint-mezard, Allergic contact dermatitis, Eur J Dermatol, vol.14, issue.5, pp.284-295, 2004.

H. Sakaguchi, The relationship between CD86/CD54 expression and THP-1 cell viability in an in vitro skin sensitization test--human cell line activation test (h-CLAT), Cell Biol Toxicol, vol.25, issue.2, pp.109-126, 2009.

H. Sakaguchi, Development of an in vitro skin sensitization test using human cell lines; human Cell Line Activation Test (h-CLAT). II. An inter-laboratory study of the h-CLAT, Toxicol In Vitro, vol.20, issue.5, pp.774-784, 2006.

S. Sakaguchi, Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25), J Immunol, vol.155, issue.3, pp.1151-1164, 1995.

F. Salazar, H. F. Sewell, F. Shakib, and A. M. Ghaemmaghami, The role of lectins in allergic sensitization and allergic disease, J Allergy Clin Immunol, vol.132, issue.1, pp.27-36, 2013.

F. Sallusto, J. Geginat, and A. Lanzavecchia, Central memory and effector memory T cell subsets: function, generation, and maintenance, Annu Rev Immunol, vol.22, pp.745-763, 2004.

F. Sallusto, Two subsets of memory T lymphocytes with distinct homing potentials and effector functions, Nature, vol.401, issue.6754, pp.708-712, 1999.

D. M. Sanderson and C. G. Earnshaw, Computer prediction of possible toxic action from chemical structure; the DEREK system, Hum Exp Toxicol, vol.10, issue.4, pp.261-273, 1991.

I. Sandquist and J. Kolls, Update on regulation and effector functions of Th17 cells, F1000Res, vol.7, p.205, 2018.

L. F. Santamaria-babi, CLA(+) T cells in cutaneous diseases, Eur J Dermatol, vol.14, issue.1, pp.13-18, 2004.

A. Savarino, F. Bottarel, F. Malavasi, and U. Dianzani, Role of CD38 in HIV-1 infection: an epiphenomenon of T-cell activation or an active player in virus/host interactions, AIDS, vol.14, issue.9, pp.1079-1089, 2000.

L. Schaefer, Complexity of danger: the diverse nature of damage-associated molecular patterns, J Biol Chem, vol.289, issue.51, pp.35237-35245, 2014.

A. Schlitzer and F. Ginhoux, Organization of the mouse and human DC network, Curr Opin Immunol, vol.26, pp.90-99, 2014.

A. Schlitzer, N. Mcgovern, and F. Ginhoux, Dendritic cells and monocyte-derived cells: Two complementary and integrated functional systems, Semin Cell Dev Biol, vol.41, pp.9-22, 2015.

T. Scholzen and J. Gerdes, The Ki-67 protein: from the known and the unknown, J Cell Physiol, vol.182, issue.3, pp.311-322, 2000.

M. Schott, Immunesurveillance by dendritic cells: potential implication for immunotherapy of endocrine cancers, Endocr Relat Cancer, vol.13, issue.3, pp.779-795, 2006.

S. Schulke, Induction of Interleukin-10 Producing Dendritic Cells As a Tool to Suppress Allergen-Specific T Helper 2 Responses, Front Immunol, vol.9, p.455, 2018.

D. H. Schuurhuis, N. Fu, F. Ossendorp, and C. J. Melief, Ins and outs of dendritic cells, International Archives of Allergy and Immunology, vol.140, issue.1, pp.53-72, 2006.

N. V. Serbina, TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection, Immunity, vol.19, issue.1, pp.59-70, 2003.

A. H. Sharpe and G. J. Freeman, The B7-CD28 superfamily, Nat Rev Immunol, vol.2, issue.2, pp.116-126, 2002.

D. Shik, IL-9-producing cells in the development of IgE-mediated food allergy, Semin Immunopathol, vol.39, issue.1, pp.69-77, 2017.

K. Shortman, Burnet oration: dendritic cells: multiple subtypes, multiple origins, multiple functions, Immunol Cell Biol, vol.78, issue.2, pp.161-165, 2000.

D. Sichien, B. N. Lambrecht, M. Guilliams, and C. L. Scott, Development of conventional dendritic cells: from common bone marrow progenitors to multiple subsets in peripheral tissues, Mucosal Immunol, vol.10, issue.4, pp.831-844, 2017.

F. P. Siegal, The nature of the principal type 1 interferon-producing cells in human blood, Science, vol.284, issue.5421, pp.1835-1837, 1999.

A. Silva, Respiratory sensitizer hexamethylene diisocyanate inhibits SOD 1 and induces ERK-dependent detoxifying and maturation pathways in dendritic-like cells, Free Radic Biol Med, vol.72, pp.238-246, 2014.

C. Silvia and G. Claudius, EURL ECVAM Recommendation on the Direct Peptide Reactivity Assay (DPRA) for Skin Sensitisation Testing, Publications Office of the European Union, pp.1-36, 2013.

B. J. Smith, J. F. Curtis, and T. E. Eling, Bioactivation of xenobiotics by prostaglandin H synthase, Chem Biol Interact, vol.79, issue.3, pp.245-264, 1991.

C. K. Smith, Human skin absorption and metabolism of the contact allergens, cinnamic aldehyde, and cinnamic alcohol, Toxicol Appl Pharmacol, vol.168, issue.3, pp.189-199, 2000.

C. K. Smith-pease, D. A. Basketter, and G. Y. Patlewicz, Contact allergy: the role of skin chemistry and metabolism, Clin Exp Dermatol, vol.28, issue.2, pp.177-183, 2003.

S. Sozzani, Dendritic cell trafficking: more than just chemokines, Cytokine Growth Factor Rev, vol.16, issue.6, pp.581-592, 2005.

R. M. Steinman, The dendritic cell system and its role in immunogenicity, Annu Rev Immunol, vol.9, pp.271-296, 1991.

R. M. Steinman, J. C. Adams, and Z. A. Cohn, Identification of a novel cell type in peripheral lymphoid organs of mice. IV. Identification and distribution in mouse spleen, J Exp Med, vol.141, issue.4, pp.804-820, 1975.

R. M. Steinman and Z. A. Cohn, Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution, J Exp Med, vol.137, issue.5, pp.1142-1162, 1973.

R. M. Steinman and Z. A. Cohn, Identification of a novel cell type in peripheral lymphoid organs of mice. II. Functional properties in vitro, J Exp Med, vol.139, issue.2, pp.380-397, 1974.

R. M. Steinman and K. Inaba, Myeloid dendritic cells, J Leukoc Biol, vol.66, issue.2, pp.205-208, 1999.

R. M. Steinman and M. C. Nussenzweig, Avoiding horror autotoxicus: the importance of dendritic cells in peripheral T cell tolerance, Proc Natl Acad Sci U S A, vol.99, issue.1, pp.351-358, 2002.

R. M. Steinman, M. Pack, and K. Inaba, Dendritic cells in the T-cell areas of lymphoid organs, Immunol Rev, vol.156, pp.25-37, 1997.

R. M. Steinman and J. Swanson, The endocytic activity of dendritic cells, J Exp Med, vol.182, issue.2, pp.283-288, 1995.

R. Stoop, Trafficking of CD44-deficient murine lymphocytes under normal and inflammatory conditions, Eur J Immunol, vol.32, issue.9, pp.2532-2574, 2002.

J. Strickland, Integrated decision strategies for skin sensitization hazard, J Appl Toxicol, vol.36, issue.9, pp.1150-1162, 2016.

L. Summers-deluca and J. L. Gommerman, Fine-tuning of dendritic cell biology by the TNF superfamily, Nat Rev Immunol, vol.12, issue.5, pp.339-351, 2012.

B. Sun and Y. Zhang, Overview of orchestration of CD4+ T cell subsets in immune responses, Adv Exp Med Biol, vol.841, pp.1-13, 2014.

S. L. Swain, K. K. Mckinstry, and T. M. Strutt, Expanding roles for CD4(+) T cells in immunity to viruses, Nat Rev Immunol, vol.12, issue.2, pp.136-148, 2012.

H. Tanaka, Human monocyte-derived dendritic cells induce naive T cell differentiation into T helper cell type 2 (Th2) or Th1/Th2 effectors. Role of stimulator/responder ratio, J Exp Med, vol.192, issue.3, pp.405-412, 2000.

A. Tang, T. A. Judge, B. J. Nickoloff, and L. A. Turka, Suppression of murine allergic contact dermatitis by CTLA4Ig. Tolerance induction of Th2 responses requires additional blockade of CD40-ligand, J Immunol, vol.157, issue.1, pp.117-125, 1996.

R. Testi, J. H. Phillips, and L. L. Lanier, T cell activation via Leu-23 (CD69), J Immunol, vol.143, issue.4, pp.1123-1128, 1989.

H. J. Thierse, T cell receptor (TCR) interaction with haptens: metal ions as non-classical haptens, Toxicology, vol.209, issue.2, pp.101-107, 2005.

R. K. Thimmulappa, Nrf2 is a critical regulator of the innate immune response and survival during experimental sepsis, J Clin Invest, vol.116, issue.4, pp.984-995, 2006.

L. Tiberio, Chemokine and chemotactic signals in dendritic cell migration, Cell Mol Immunol, 2018.

M. J. Toebak, Dendritic cells: biology of the skin, Contact Dermatitis, vol.60, issue.1, pp.2-20, 2009.

S. Trifari, Identification of a human helper T cell population that has abundant production of interleukin 22 and is distinct from T(H)-17, T(H)1 and T(H)2 cells, Nat Immunol, vol.10, issue.8, pp.864-871, 2009.

J. A. Troutman, The incorporation of lysine into the peroxidase peptide reactivity assay for skin sensitization assessments, Toxicol Sci, vol.122, issue.2, pp.422-436, 2011.

F. Tsushima, Preferential contribution of B7-H1 to programmed death-1-mediated regulation of hapten-specific allergic inflammatory responses, Eur J Immunol, vol.33, issue.10, pp.2773-2782, 2003.

D. J. Tumes, Epigenetic regulation of T-helper cell differentiation, memory, and plasticity in allergic asthma, Immunol Rev, vol.278, issue.1, pp.8-19, 2017.

D. Urbisch, Assessment of Pre-and Pro-haptens Using Nonanimal Test Methods for Skin Sensitization, Chem Res Toxicol, vol.29, issue.5, pp.901-913, 2016.

H. H. Van-acker, A. Capsomidis, E. L. Smits, and V. F. Van-tendeloo, CD56 in the Immune System: More Than a Marker for Cytotoxicity, vol.8, p.892, 2017.

L. T. Van-den-broeke, Direct Ni2+ antigen formation on cultured human dendritic cells, Immunology, vol.96, issue.4, pp.578-585, 1999.

J. W. Van-der-veen, The involvement of the Toll-like receptor signaling and Nrf2-Keap1 pathways in the in vitro regulation of IL-8 and HMOX1 for skin sensitization, Journal of Immunotoxicology, vol.13, issue.1, pp.1-6, 2016.

E. Van-vliet, State-of-the-art and new options to assess T cell activation by skin sensitizers: Cosmetics Europe Workshop, ALTEX, 2017.

R. J. Vandebriel and H. Van-loveren, Non-animal sensitization testing: state-of-the-art, Crit Rev Toxicol, vol.40, issue.5, pp.389-404, 2010.

M. Vigan, Clinical patterns of allergic contact dermatitis to fragrances, Revue Francaise D'Allergologie, vol.49, issue.4, pp.356-359, 2009.

M. Vocanson, In vitro sensitization and primary T cell responses to contact allergens: Towards the development of human T cell priming assays, Toxicology Letters, vol.229, pp.28-29, 2014.

M. Vocanson, Human T cell priming assay: depletion of peripheral blood lymphocytes in CD25(+) cells improves the in vitro detection of weak allergen-specific T cells, EXS, vol.104, pp.89-100, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01911350

M. Vocanson, Depletion of human peripheral blood lymphocytes in CD25+ cells allows for the sensitive in vitro screening of contact allergens, J Invest Dermatol, vol.128, issue.8, pp.2119-2122, 2008.

M. Vocanson, Effector and regulatory mechanisms in allergic contact dermatitis, Allergy, vol.64, issue.12, pp.1699-1714, 2009.

M. Vocanson, Effector and regulatory mechanisms in allergic contact dermatitis, Allergy, vol.64, issue.12, pp.1699-1714, 2009.

M. Vocanson, Contact hypersensitivity: T-cell based assay, Curr Opinion in Toxicology, vol.5, pp.39-45, 2017.

M. Vocanson, J. Nicolas, and D. Basketter, In vitro approaches to the identification and characterization of skin sensitizers, Expert Review of Dermatology, vol.8, issue.4, pp.395-405, 2013.

M. Vocanson, Inducible costimulator (ICOS) is a marker for highly suppressive antigenspecific T cells sharing features of TH17/TH1 and regulatory T cells, J Allergy Clin Immunol, vol.126, issue.2, pp.281-287, 2010.

D. Vremec, CD4 and CD8 expression by dendritic cell subtypes in mouse thymus and spleen, J Immunol, vol.164, issue.6, pp.2978-2986, 2000.

S. Vukmanovic and N. Sadrieh, Skin sensitizers in cosmetics and beyond: potential multiple mechanisms of action and importance of T-cell assays for in vitro screening, Crit Rev Toxicol, vol.47, issue.5, pp.415-432, 2017.

T. Walzer, Cutting edge: immediate RANTES secretion by resting memory CD8 T cells following antigenic stimulation, J Immunol, vol.170, issue.4, pp.1615-1619, 2003.

H. Watanabe, Activation of the IL-1beta-processing inflammasome is involved in contact hypersensitivity, J Invest Dermatol, vol.127, issue.8, pp.1956-1963, 2007.

R. S. Welner, Interferon-producing killer dendritic cells (IKDCs) arise via a unique differentiation pathway from primitive c-kitHiCD62L+ lymphoid progenitors, Blood, vol.109, issue.11, pp.4825-4931, 2007.

G. S. Whitehead, Allergen-induced airway disease is mouse strain dependent, American Journal of Physiology. Lung Cellular and Molecular Physiology, vol.285, issue.1, pp.32-42, 2003.

, Global, regional, and national comparative risk assessment of 84 behavioural, environmental and occupational, and metabolic risks or clusters of risks, 1990-2016: a systematic analysis for the Global Burden of Disease Study, Lancet, vol.390, pp.1345-1422, 2016.

M. A. Willart and B. N. Lambrecht, The danger within: endogenous danger signals, atopy and asthma, Clinical and Experimental Allergy, vol.39, issue.1, pp.12-19, 2009.

M. A. Williams and M. J. Bevan, Effector and memory CTL differentiation, Annu Rev Immunol, vol.25, pp.171-192, 2007.

E. J. Witsch, ICOS and CD28 reversely regulate IL-10 on re-activation of human effector T cells with mature dendritic cells, Eur J Immunol, vol.32, issue.9, pp.2680-2686, 2002.

C. L. Wong, S. Ghassabian, M. T. Smith, and A. L. Lam, In vitro methods for hazard assessment of industrial chemicals -opportunities and challenges, Front Pharmacol, vol.6, p.94, 2015.

C. Yang and P. , The phenotype and survival of antigen-stimulated transgenic CD4 T cells in vivo: the influence of persisting antigen, Int Immunol, vol.18, issue.4, pp.515-538, 2006.

B. Yang, B. K. Sundquist, and M. A. Pasha, Determining the clinical relevance of positive patch testing to gold in evaluation of contact dermatitis, J Allergy Clin Immunol Pract, 2018.

T. Yokosuka and T. Saito, The immunological synapse, TCR microclusters, and T cell activation, Curr Top Microbiol Immunol, vol.340, pp.81-107, 2010.

W. M. Yokoyama, Characterization of a cell surface-expressed disulfide-linked dimer involved in murine T cell activation, J Immunol, vol.141, issue.2, pp.369-376, 1988.

Y. Yoshida, Evaluation of the skin sensitization potential of chemicals using expression of co-stimulatory molecules, CD54 and CD86, on the naive THP-1 cell line, Toxicol In Vitro, vol.17, issue.2, pp.221-228, 2003.

J. Yun, J. Adam, D. Yerly, and W. J. Pichler, Human leukocyte antigens (HLA) associated drug hypersensitivity: consequences of drug binding to HLA, Allergy, vol.67, issue.11, pp.1338-1346, 2012.

Z. Zas?ona, Prostaglandin E? suppresses allergic sensitization and lung inflammation by targeting the E prostanoid 2 receptor on T cells, The Journal of Allergy and Clinical Immunology, vol.133, issue.2, pp.379-387, 2014.

Y. Zhang, W. Gu, L. He, and B. Sun, Th1/Th2 cell's function in immune system, Adv Exp Med Biol, vol.841, pp.45-65, 2014.

Y. Zhang, W. Gu, and B. Sun, TH1/TH2 cell differentiation and molecular signals, Adv Exp Med Biol, vol.841, pp.15-44, 2014.

L. J. Zhou and T. F. Tedder, Human blood dendritic cells selectively express CD83, a member of the immunoglobulin superfamily, J Immunol, vol.154, issue.8, pp.3821-3835, 1995.

L. Ziegler-heitbrock and T. P. Hofer, Toward a refined definition of monocyte subsets, Front Immunol, vol.4, p.23, 2013.

S. F. Ziegler, F. Ramsdell, and M. R. Alderson, The activation antigen CD69, Stem Cells, vol.12, issue.5, pp.456-465, 1994.

C. Wong, S. Ghassabian, M. Smith, and A. L. Lam, In vitro methods for hazard assessment of industrial chemicals -opportunities and challenges, Front Pharmacol, issue.6, p.94, 2015.

M. Fernandez-nieto, S. Quirce, J. Carnes, and J. Sastre, Occupational asthma due to chromium and nickel salts, Int Arch Occup Environ Health, vol.79, pp.483-486, 2006.

I. Spoerri, K. Scherer, and S. Michel, Detection of nickel and palladium contact hypersensitivity by a flow cytometric lymphocyte proliferation test, Allergy, vol.70, pp.323-327, 2015.

D. Basketter, S. Lemoine, and J. P. Mcfadden, Skin sensitisation to fragrance ingredients: is there a role for household cleaning/maintenance products?, Eur J Dermatol, vol.25, pp.7-13, 2015.

O. Yamamoto and Y. Tokura, Photocontact dermatitis and chloracne: two major occupational and environmental skin diseases induced by different actions of halogenated chemicals, J Dermatol Sci, vol.32, pp.85-94, 2003.

J. Cahill, J. Williams, and M. Matheson, Occupational contact dermatitis: a review of 18 years of data from an occupational dermatology clinic in Australia, Research Reports, 2012.

S. Cochrane, J. Arts, and C. Ehnes, Thresholds in chemical respiratory sensitisation, Toxicology, vol.333, pp.179-194, 2015.

I. Kimber, R. Dearman, D. Basketter, and D. R. Boverhof, Chemical respiratory allergy: reverse engineering an adverse outcome pathway, Toxicology, vol.318, pp.32-39, 2014.

, Guidelines for testing of chemicals. Draft Guideline No. 429. Skin sensitisation: the local lymph node assay, 2001.

. Onu)-nu, Système général harmonisé de classification et d'étiquetage des produits chimiques (SGH)5th revised edition: New York and Geneva, United Nations, 2013.

, on classification, labelling and packaging of substances and mixtures, amending and repealing Directives 67/548/EEC and 1999/45/EC, and amending Regulation (EC) No. 1907, European Commission, p.353, 2006.

, Regulation (EC) No. 1907/2006 of the European Parliament and of the Council, European Commission, vol.18

, Authorisation and Restriction of Chemicals (REACH), establishing a European Chemicals Agency, amending Directive 1999/45/EC and repealing Council Regulation (EEC) No. 793/93 and Commission Regulation (EC) No. 1488/94 as well as Council Directive 76/769/EEC and Commission Directives 91/155/EEC, 93/67/EEC, 93/105/EC and 2000/21/EC, Off J Eur Union, pp.136-133, 2006.

G. Santos, J. Reinders, and K. Ouwehand, Progress on the development of human in vitro dendritic cell based assays for assessment of the sensitizing potential of a compound, Toxicol Appl Pharmacol, vol.236, pp.372-382, 2009.

J. Ezendam, H. Braakhuis, and R. J. Vandebriel, State of the art in non-animal approaches for skin sensitization testing: from individual test methods towards testing strategies, Arch Toxicol, vol.90, pp.2861-2883, 2016.

C. Mackay, M. Davies, V. Summerfield, and G. Maxwell, From pathways to people: applying the adverse outcome pathway (AOP) for skin sensitization to risk assessment, ALTEX, vol.2013, pp.473-486

J. Hennen and B. Blömeke, Keratinocytes improve prediction of sensitization potential and potency of chemicals with THP-1 cells, ALTEX, vol.2017, pp.279-288

A. Mehling, T. Eriksson, and T. Eltze, Non-animal test methods for predicting skin sensitization potentials, Arch Toxicol, vol.86, pp.1273-1295, 2012.

N. Delrue, M. Sachana, and Y. Sakuratani, The adverse outcome pathway concept: a basis for developing regulatory decision-making tools, Altern Lab Anim, vol.44, pp.417-429, 2016.

, The Adverse Outcome Pathway for Skin Sensitisation Initiated by Covalent Binding to Proteins: Paris, 2014.

I. Kimber, D. Basketter, and G. Gerberick, Chemical allergy: translating biology into hazard characterization, Toxicol Sci, pp.238-268, 2011.

M. Toebak, S. Gibbs, and D. Bruynzeel, Dendritic cells: biology of the skin, Contact Dermatitis, vol.60, pp.2-20, 2009.

S. Casati, A. Worth, A. Sven, and P. , EURL ECVAM Strategy for Replacement of Animal Testing for Skin Sensitisation Hazard Identification and Classification. Publications Office of the European Union, 2013.

, Chemico Skin Sensitisation: Direct Peptide Reativity Assay (DPRA), vol.442, 2015.

, In Vitro Skin Sensitisation: Human Cell Line Activation Test (h-CLAT, vol.442, 2016.

, Vitro Skin Sensitisation: ARE-Nrf2 Luciferase Test Method, vol.442, 2015.

. Eurl-ecvam, Status report on the development, validation and regulatory acceptance of alternative methods and approaches, 2016.

E. Pepin, M. Goutet, and M. Ban, Murine bone marrow-derived dendritic cells as a potential in vitro model for predictive identification of chemical sensitizers, Toxicol Lett, vol.175, pp.89-101, 2007.

S. Casati, P. Aeby, and K. I. , Selection of chemicals for the development and evaluation of in vitro methods for skin sensitisation testing, Altern Lab Anim, vol.37, pp.305-312, 2009.

, Contact Sensitisation: Classification According to Potency, European Centre for Ecotoxicology and Toxicology of Chemicals, vol.87, 2003.

C. Rovida, D. Basketter, and S. Casati, Management of an integrated project (Sens-it-iv) to develop in vitro tests to assess sensitisation, Altern Lab Anim, vol.35, pp.317-322, 2007.

K. Inaba, W. Swiggard, and R. Steinman, Isolation of dendritic cells, Curr Protoc Immunol, vol.3, p.7, 2009.

P. Azam, J. Peiffer, and D. Chamousset, The cytokine-dependent MUTZ-3 cell line as an in vitro model for the screening of contact sensitizers, Toxicol Appl Pharmacol, vol.212, pp.14-23, 2006.

D. Basketter, I. White, J. Mcfadden, and K. I. , Skin sensitization: implications for integration of clinical data into hazard identification and risk assessment, Hum Exp Toxicol, vol.34, pp.1222-1230, 2015.

D. Basketter, K. Andersen, and C. Lidén, Evaluation of the skin sensitizing potency of chemicals by using the existing methods and considerations of relevance for elicitation, Contact Dermatitis, vol.52, pp.39-43, 2005.

P. Griem, C. Goebel, and H. Scheffler, Proposal for a risk assessment methodology for skin sensitization based on sensitization potency data, Regul Toxicol Pharmacol, vol.38, pp.269-290, 2003.

K. Schneider and Z. Akkan, Quantitative relationship between the local lymph node assay and human skin sensitization IN VITRO DETECTION OF CHEMICAL ALLERGENS ? BATTAIS ET AL. assays, Regul Toxicol Pharmacol, vol.39, pp.245-255, 2004.

S. Loveless, A. Api, and R. Crevel, Potency values from the local lymph node assay: application to classification, labelling and risk assessment, Regul Toxicol Pharmacol, vol.56, pp.54-66, 2010.

K. Saito, M. Miyazawa, and Y. Nukada, Development of an in vitro skin sensitization test based on ROS production in THP-1 cells, Toxicol In Vitro, vol.27, pp.857-863, 2013.

N. Lambrechts, H. Vanheel, and I. Nelissen, Assessment of chemical skin-sensitizing potency by an in vitro assay based on human dendritic cells, Toxicol Sci, vol.116, pp.122-129, 2010.

G. Gerberick, C. Ryan, and P. Kern, Compilation of historical local lymph node data for evaluation of skin sensitization alternative methods, Dermatitis, vol.16, pp.157-202, 2005.

R. Emter, G. Ellis, and A. Natsch, Performance of a novel keratinocyte-based reporter cell line to screen skin sensitizers in vitro, Toxicol Appl Pharmacol, vol.245, pp.281-290, 2010.

C. Kuper, M. Radonjic, and J. Van-triel, Oxazolone (OXA) is a respiratory allergen in Brown Norway rats, Toxicology, pp.290-59, 2011.

J. Thyssen, W. Uter, and J. Mcfadden, The EU Nickel Directive revisited -future steps towards better protection against nickel allergy, Contact Dermatitis, vol.64, pp.121-125, 2011.

M. Schmidt, B. Raghavan, and V. Müller, Crucial role for human Toll-like receptor 4 in the development of contact allergy to nickel, Nat Immunol, vol.11, pp.814-819, 2010.

M. Cruz, M. Gonçalo, and A. Paiva, Contact sensitizers downregulate the expression of the chemokine receptors CCR6 and CXCR4 in a skin dendritic cell line, Arch Dermatol Res, vol.297, pp.43-47, 2005.

M. Watanabe, N. Ishimaru, and M. Ashrin, A novel DC therapy with manipulation of MKK6 gene on nickel allergy in mice, PLoS ONE, pp.6-19017, 2011.

E. Corsini, V. Galbiati, and M. Mitjans, NCTC 2544 and IL-18 production: a tool for the identification of contact allergens, Toxicol In Vitro, vol.27, pp.1127-1134, 2013.

K. Saito, Y. Nukada, and O. Takenouchi, Development of a new in vitro skin sensitization assay (Epidermal Sensitization Assay; EpiSensA) using reconstructed human epidermis, Toxicol In Vitro, vol.27, pp.2213-2224, 2013.

A. Cortial, A. Nosbaum, and A. Rozieres, Encapsulation of hydrophobic allergens into nanoparticles improves the in vitro immunological diagnosis of allergic contact dermatitis, Nanomedicine, vol.11, pp.1029-1033, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01911058

A. Natsch, C. Ryan, and L. Foertsch, A dataset on 145 chemicals tested in alternative assays for skin sensitization undergoing prevalidation, J Appl Toxicol, vol.33, pp.1337-1352, 2013.

D. Basketter and K. I. , Skin irritation, false positives and the local lymph node assay: a guideline issue, Regul Toxicol Pharmacol, pp.61-137, 2011.

N. Ball, S. Cagen, and J. Carrillo, Evaluating the sensitization potential of surfactants: integrating data from the local lymph node assay, guinea pig maximization test, and in vitro methods in a weight-of-evidence approach, Regul Toxicol Pharmacol, vol.60, pp.389-400, 2011.

F. Python, C. Goebel, and P. Aeby, Assessment of the U937 cell line for the detection of contact allergens, Toxicol Appl Pharmacol, vol.220, pp.113-124, 2007.

H. Reuter, S. Gerlach, and J. Spieker, Evaluation of an optimized protocol using human peripheral blood monocyte derived dendritic cells for the in vitro detection of sensitizers: results of a ring study in five laboratories, Toxicol In Vitro, vol.29, pp.976-986, 2015.

R. Dearman, D. Basketter, and K. I. , Inter-relationships between different classes of chemical allergens, J Appl Toxicol, vol.33, pp.558-565, 2013.

L. Plitnick, S. Loveless, and G. Ladics, Cytokine mRNA profiles for isocyanates with known and unknown potential to induce respiratory sensitization, Toxicology, vol.207, pp.487-499, 2005.

D. Basketter, M. Marriott, N. Gilmour, and I. R. White, Strong irritants masquerading as skin allergens: the case of benzalkonium chloride, Contact Dermatitis, vol.50, pp.213-217, 2004.

Y. Cao, P. Ma, and W. Liu, Evaluation of the skin sensitization potential of chemicals in THP-1/keratinocyte co-cultures, Immunopharmacol Immunotoxicol, vol.34, pp.196-204, 2012.

D. Basketter and K. I. , Skin sensitization, false positives and false negatives: experience with guinea pig assays, J Appl Toxicol, vol.30, pp.381-386, 2010.

H. Dao, C. Fricker, and S. T. Nedorost, Sensitization prevalence for benzalkonium chloride and benzethonium chloride, Dermatitis, vol.23, pp.162-166, 2012.

T. Suneja and D. Belsito, Occupational dermatoses in health care workers evaluated for suspected allergic contact dermatitis, Contact Dermatitis, vol.58, pp.285-290, 2008.

W. Uter, H. Lessmann, J. Geier, and A. Schnuch, Is the irritant benzalkonium chloride a contact allergen? A contribution to the ongoing debate from a clinical perspective, Contact Dermatitis, vol.58, pp.359-363, 2008.

W. Uter, C. Rämsch, and W. Aberer, The European baseline series in 10 European Countries, -results of the European Surveillance System on Contact Allergies (ESSCA), vol.61, pp.31-38, 2005.

A. Wentworth, J. Yiannias, M. Davis, and J. Killian, Benzalkonium chloride: a known irritant and novel allergen, Dermatitis, vol.27, pp.14-20, 2016.

M. Bellier, C. Barnig, and J. Renaudin, Importance of specific inhalation challenge in the diagnosis of occupational asthma induced by quaternary ammonium compounds, J Allergy Clin Immunol Pract, vol.3, pp.819-820, 2015.

B. Marple, P. Roland, and M. Benninger, Safety review of benzalkonium chloride used as a preservative in intranasal solutions: an overview of conflicting data and opinions, Otolaryngol Head Neck Surg, vol.130, pp.131-141, 2004.

A. Purohit, M. Kopferschmitt-kubler, and C. Moreau, Quaternary ammonium compounds and occupational asthma, Int Arch Occup Environ Health, vol.73, pp.423-427, 2000.

I. Kimber, D. A. Basketter, G. F. Gerberick, C. A. Ryan, and R. J. Dearman, Chemical allergy: translating biology into hazard characterization, Toxicol Sci, vol.120, issue.1, pp.238-268, 2011.

E. Mamessier, F. Milhe, and C. Guillot, T-cell activation in occupational asthma and rhinitis, Allergy, vol.62, pp.162-169, 2007.

C. L. Wong, S. Ghassabian, M. T. Smith, and A. L. Lam, In vitro methods for hazard assessment of industrial chemicals-opportunities and challenges, Front Pharmacol, vol.6, p.94, 2015.

S. A. Cochrane, J. H. Arts, and C. Ehnes, Thresholds in chemical respiratory sensitisation, Toxicology, vol.333, pp.179-194, 2015.

. European, Regulation (EC) No 1272/2008 of the European Parliament and of the Council of 16 December 2008 on classification, labelling and packaging of substances and mixtures, amending and repealing Directives 67/548/EEC and 1999/45/EC, and amending Regulation (EC) No, Official Journal of the European Union, p.353, 1907.

, Chemico Skin Sensitisation: Direct Peptide Reativity Assay (DPRA), vol.442, 2015.

, Vitro Skin Sensitisation: ARE-Nrf2 Luciferase test method, vol.442, 2015.

, Vitro Skin Sensitisation, vol.442, 2017.

, The Adverse Outcome Pathway for Skin Sensitisation Initiated by Covalent Binding to Proteins, 2014.

S. F. Martin, P. R. Esser, and S. Schmucker, T-cell recognition of chemicals, protein allergens and drugs: towards the development of in vitro assays, Cell Mol Life Sci, vol.67, pp.4171-4184, 2010.

M. Vocanson, A. Hennino, A. Rozieres, G. Poyet, and J. F. Nicolas, Effector and regulatory mechanisms in allergic contact dermatitis, Allergy, vol.64, pp.1699-1714, 2009.

H. Reuter, J. Spieker, and S. Gerlach, In vitro detection of contact allergens: development of an optimized protocol using human peripheral blood monocyte-derived dendritic cells, Toxicol In Vitro, vol.25, pp.315-323, 2011.

M. J. Toebak, S. Gibbs, D. P. Bruynzeel, R. J. Scheper, and T. Rustemeyer, Dendritic cells: biology of the skin, Contact Dermatitis, vol.60, pp.2-20, 2009.

M. Krasteva, J. Peguet-navarro, C. Moulon, P. Courtellemont, G. Redziniak et al., In vitro primary sensitization of hapten-specific T cells by cultured human epidermal Langerhans cells-a screening predictive assay for contact sensitizers, Clin Exp Allergy, vol.26, pp.563-570, 1996.

N. Rougier, G. Redziniak, D. Mougin, D. Schmitt, and C. Vincent, In vitro evaluation of the sensitization potential of weak contact allergens using langerhans-like dendritic cells and autologous T cells, Toxicology, vol.145, pp.73-82, 2000.

M. Vocanson, A. Achachi, and V. Mutez, Human T cell priming assay: depletion of peripheral blood lymphocytes in CD25(+) cells improves the in vitro detection of weak allergen-specific T cells, EXS, vol.104, pp.89-100, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01911350

G. Guironnet, C. Dalbiez-gauthier, F. Rousset, D. Schmitt, and J. Péguet-navarro, In vitro human T cell sensitization to haptens by monocyte-derived dendritic cells, Toxicol In Vitro, vol.14, pp.517-522, 2000.

D. Masopust, C. P. Sivula, and S. C. Jameson, Of mice, dirty mice, and men: using mice to understand human immunology, J Immunol, vol.199, pp.383-388, 2017.

F. Battais, C. Huppert, and I. Langonne, In vitro detection of chemical allergens: an optimized assay using mouse bone marrow-derived dendritic cells, Contact Dermatitis, vol.77, issue.5, pp.311-322, 2017.

E. Pepin, M. Goutet, and M. Ban, Murine bone marrow-derived dendritic cells as a potential in vitro model for predictive identification of chemical sensitizers, Toxicol Lett, vol.175, pp.89-101, 2007.

G. G. Santos, J. Reinders, K. Ouwehand, T. Rustemeyer, R. J. Scheper et al., Progress on the development of human in vitro dendritic cell based assays for assessment of the sensitizing potential of a compound, Toxicol Appl Pharmacol, vol.236, pp.372-382, 2009.

R. J. Dearman, D. A. Basketter, and K. I. , Inter-relationships between different classes of chemical allergens, J Appl Toxicol, vol.33, pp.558-565, 2013.

. Ecetoc, Skin and Respiratory Sensitisers: Reference Chemical Data Bank

, European Center of Ecotoxicology and Toxicology of Chemicals, p.92, 1999.

I. Kimber, M. A. Travis, S. F. Martin, and R. J. Dearman, Immunoregulation of skin sensitization and regulatory T cells, Contact Dermatitis, vol.67, pp.179-183, 2012.

K. Inaba, W. J. Swiggard, R. M. Steinman, N. Romani, and G. Schuler, Isolation of dendritic cells, Curr Protoc Immunol, 2009.

G. F. Gerberick, L. W. Cruse, C. M. Miller, E. E. Sikorski, and G. M. Ridder, Selective modulation of T cell memory markers CD62L and CD44 on murine draining lymph node cells following allergen and irritant treatment, Toxicol Appl Pharmacol, vol.146, pp.1-10, 1997.

D. Cibrian and F. Sanchez-madrid, CD69: from activation marker to metabolic gatekeeper, Eur J Immunol, vol.47, pp.946-953, 2017.

R. Gonzalez-amaro, J. R. Cortes, F. Sanchez-madrid, and P. Martin, Is CD69 an effective brake to control inflammatory diseases?, Trends Mol Med, vol.19, pp.625-632, 2013.

M. Moniuszko, K. Kowal, M. Jeznach, M. Rusak, M. Dabrowska et al., Phenotypic correlations between monocytes and CD4+ T cells in allergic patients, Int Arch Allergy Immunol, vol.161, pp.131-141, 2013.

M. Ban, I. Langonne, M. Goutet, N. Huguet, and E. Pépin, Simultaneous analysis of the local and systemic immune responses in mice to study the occupational asthma mechanisms induced by chromium and platinum, Toxicology, vol.277, pp.29-37, 2010.

C. Tan, A. A. Taylor, M. Z. Coburn, J. H. Marino, C. J. Van-de-wiele et al., Ten-color flow cytometry reveals distinct patterns of expression of CD124 and CD126 by developing thymocytes, BMC Immunol, vol.12, p.36, 2011.

M. Berker, L. J. Frank, and A. L. Gessner, Allergies-a T cells perspective in the era beyond the TH1/TH2 paradigm, Clin Immunol, vol.174, pp.73-83, 2017.

M. Goutet, E. Pepin, I. Langonne, N. Huguet, and M. Ban, Identification of contact and respiratory sensitizers according to IL-4 receptor alpha expression and IL-2 production, Toxicol Appl Pharmacol, vol.260, pp.95-104, 2012.

M. Vocanson, J. Nicolas, and D. Basketter, In vitro approaches to the identification and characterization of skin sensitizers, Expert Rev Dermatol, vol.8, pp.395-405, 2013.

T. Rustemeyer, S. De-ligter, V. Blomberg, B. M. Frosch, P. J. Scheper et al., Human T lymphocyte priming in vitro by haptenated autologous dendritic cells, Clin Exp Immunol, vol.117, pp.209-216, 1999.

J. T. White, E. W. Cross, and R. M. Kedl, Antigen-inexperienced memory CD8+ T cells: where they come from and why we need them, Nat Rev Immunol, vol.17, pp.391-400, 2017.

J. M. Curtsinger, P. Agarwal, D. C. Lins, and M. F. Mescher, Autocrine IFN-gamma promotes naive CD8 T cell differentiation and synergizes with IFN-alpha to stimulate strong function, J Immunol, vol.189, pp.659-668, 2012.

N. Zhang and M. J. Bevan, CD8(+) T cells: foot soldiers of the immune system, Immunity, vol.35, pp.161-168, 2011.

Y. Takeda, Y. Suto, and K. Ito, TRAV7-2*02 Expressing CD8(+) T Cells Are Responsible for Palladium Allergy, Int J Mol Sci, vol.18, 2017.

M. Kawano, M. Nakayama, and Y. Aoshima, NKG2D(+) IFN-gamma(+) CD8(+) T cells are responsible for palladium allergy, PLoS One, vol.9, p.86810, 2014.

S. F. Martin, J. C. Dudda, and V. Delattre, Fas-mediated inhibition of CD4+ T cell priming results in dominance of type 1 CD8+ T cells in the immune response to the contact sensitizer trinitrophenyl, J Immunol, vol.173, pp.3178-3185, 2004.

S. Stone, L. Flamme, and A. C. , Type II activation of macrophages and microglia by immune complexes enhances Th17 biasing in an IL-6-independent manner, PLoS ONE, vol.11, p.164454, 2016.

Y. Garfias, E. Rojas-ramos, J. Mdel, and C. , Comparative analysis of mononuclear cell surface markers in atopic processes-a preliminary study, Immunol Invest, vol.32, pp.95-104, 2003.

D. Gioacchino, M. , D. Giampaolo, L. Verna, and N. , In vitro effects of platinum compounds on lymphocyte proliferation and cytokine release, Ann Clin Lab Sci, vol.34, pp.195-202, 2004.

C. Paolucci, J. Ponti, M. V. Fabbri, D. Breda, E. Sabbioni et al., Platinum group elements enhance the allergic immune response by acting on dendritic cells, Int Arch Allergy Immunol, vol.143, pp.225-236, 2007.

, SCOEL/SUM/150. Recommendation from the Scientific Committee on Occupational Exposure Limits for Platinum and Platinum compounds, 2011.

K. Nelms, A. D. Keegan, J. Zamorano, J. J. Ryan, and W. E. Paul, The IL-4 receptor: signaling mechanisms and biologic functions, Annu Rev Immunol, vol.17, pp.701-738, 1999.

H. Kotanides and N. C. Reich, Interleukin-4-induced STAT6 recognizes and activates a target site in the promoter of the interleukin-4 receptor gene, J Biol Chem, vol.271, pp.25555-25561, 1996.

J. Ohara and W. E. Paul, Up-regulation of interleukin 4/B-cell stimulatory factor 1 receptor expression, Proc Natl Acad Sci U S A, vol.85, pp.8221-8225, 1988.

R. Nishino, T. Fukuyama, Y. Watanabe, Y. Kurosawa, H. Ueda et al., Effect of mouse strain in a model of chemical-induced respiratory allergy, Exp Anim, vol.63, pp.435-445, 2014.

J. C. Wang and A. M. Livingstone, Cutting edge: CD4+ T cell help can be essential for primary CD8+ T cell responses in vivo, J Immunol, vol.171, pp.6339-6343, 2003.

A. M. Livingstone, E. B. Wilson, F. Ontiveros, J. Wang, C. Huppert et al., Activation of T cells by dendritic cells exposed to a reference sensitizer: Towards a promising model to assess the allergenic potential of chemicals, Contact Dermatitis, vol.45, pp.209-217, 2009.

,