J. H. Lee, J. W. Kim, D. K. Kim, H. S. Kim, H. J. Park et al., The Src family kinase Fgr is critical for activation of mast cells and IgE-mediated anaphylaxis in mice, J Immunol, vol.187, pp.1807-1815, 2011.

S. G. Brown, K. E. Blackman, and R. J. Heddle, Can serum mast cell tryptase help diagnose anaphylaxis, Emerg Med Australas, vol.16, pp.120-124, 2004.

A. Cheifetz, M. Smedley, S. Martin, M. Reiter, G. Leone et al., The incidence and management of infusion reactions to infliximab: a large center experience, Am J Gastroenterol, vol.98, pp.1315-1324, 2003.

A. P. Schmidt, H. F. Taswell, and G. J. Gleich, Anaphylactic transfusion reactions associated with anti-IgA antibody, N Engl J Med, vol.280, pp.188-193, 1969.

H. Hedin, W. Richter, K. Messmer, H. Renck, K. G. Ljungstrom et al., Laubenthal, Incidence, pathomechanism and prevention of dextran-induced anaphylactoid / anaphylactic reactions in man, Dev Biol Stand, vol.48, pp.179-189, 1980.

L. Bergamaschini, P. M. Mannucci, A. B. Federici, R. Coppola, S. Guzzoni et al., Posttransfusion anaphylactic reactions in a patient with severe von Willebrand disease: role of complement and alloantibodies to von Willebrand factor, J Lab Clin Med, vol.125, pp.348-355, 1995.

C. Steenholdt, M. Svenson, K. Bendtzen, O. O. Thomsen, J. Brynskov et al., Acute and delayed hypersensitivity reactions to infliximab and adalimumab in a patient with Crohn's disease, J Crohns Colitis, vol.6, pp.108-111, 2012.

M. E. Weiss, D. Nyhan, Z. K. Peng, J. C. Horrow, E. Lowenstein et al., Association of protamine IgE and IgG antibodies with life-threatening reactions to intravenous protamine, N Engl J Med, vol.320, pp.886-892, 1989.

F. D. Finkelman, M. V. Khodoun, and R. Strait, Human IgE-independent systemic anaphylaxis, J Allergy Clin Immunol, vol.137, pp.1674-1680, 2016.

H. C. Oettgen, T. R. Martin, A. Wynshaw-boris, C. Deng, J. M. Drazen et al., Active anaphylaxis in IgE-deficient mice, Nature, vol.370, pp.367-370, 1994.

D. Dombrowicz, V. Flamand, I. Miyajima, J. V. Ravetch, S. J. Galli et al., Absence of Fc epsilonRI alpha chain results in upregulation of Fc gammaRIII-dependent mast cell degranulation and anaphylaxis. Evidence of competition between Fc epsilonRI and Fc gammaRIII for limiting amounts of FcR beta and gamma chains, J Clin Invest, vol.99, pp.915-925, 1997.

I. Miyajima, D. Dombrowicz, T. R. Martin, J. V. Ravetch, J. P. Kinet et al., Systemic anaphylaxis in the mouse can be mediated largely through IgG1 and Fc gammaRIII. Assessment of the cardiopulmonary changes, mast cell degranulation, and death associated with active or IgE-or IgG1-dependent passive anaphylaxis, J Clin Invest, vol.99, pp.901-914, 1997.

I. H. Choi, Y. M. Shin, J. S. Park, M. S. Lee, E. H. Han et al., Immunoglobulin E-dependent active fatal anaphylaxis in mast cell-deficient mice, J Exp Med, vol.188, pp.1587-1592, 1998.

F. D. Finkelman, Anaphylaxis: lessons from mouse models, J Allergy Clin Immunol, vol.120, pp.516-507, 2007.

H. Beutier, C. M. Gillis, B. Iannascoli, O. Godon, P. England et al., IgG subclasses determine pathways of anaphylaxis in mice, J Allergy Clin Immunol, vol.139, p.267, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-01388338

M. V. Khodoun, Z. Y. Kucuk, R. T. Strait, D. Krishnamurthy, K. Janek et al., Rapid desensitization of mice with antiFcgammaRIIb/FcgammaRIII mAb safely prevents IgG-mediated anaphylaxis, J Allergy Clin Immunol, vol.132, pp.1375-1387, 2013.

R. T. Strait, S. C. Morris, M. Yang, X. W. Qu, and F. D. Finkelman, Pathways of anaphylaxis in the mouse, J Allergy Clin Immunol, vol.109, pp.658-668, 2002.

D. Jiao, Y. Liu, X. Lu, B. Liu, Q. Pan et al., Macrophages are the dominant effector cells responsible for IgG-mediated passive systemic anaphylaxis challenged by natural protein antigen in BALB/c and C57BL/6 mice, Cell Immunol, vol.289, pp.97-105, 2014.

F. Jönsson, D. A. Mancardi, Y. Kita, H. Karasuyama, B. Iannascoli et al., Mouse and human neutrophils induce anaphylaxis, J Clin Invest, vol.121, pp.1484-1496, 2011.

Y. Tsujimura, K. Obata, K. Mukai, H. Shindou, M. Yoshida et al., Basophils play a pivotal role in immunoglobulin-G-mediated but not immunoglobulin-E-mediated systemic anaphylaxis, Immunity, vol.28, pp.581-589, 2008.

P. Bruhns and F. Jonsson, Mouse and human FcR effector functions, Immunol Rev, vol.268, pp.25-51, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01281740

R. Molfetta, F. Gasparrini, A. Santoni, and R. Paolini, Ubiquitination and endocytosis of the high affinity receptor for IgE, Mol Immunol, vol.47, pp.2427-2434, 2010.

M. V. Khodoun, R. Strait, L. Armstrong, N. Yanase, and F. D. Finkelman, Identification of markers that distinguish IgE-from IgG-mediated anaphylaxis, Proc Natl Acad Sci U S A, vol.108, pp.12413-12418, 2011.

C. M. Gillis, F. Jonsson, D. A. Mancardi, N. Tu, H. Beutier et al., Mechanisms of anaphylaxis in human low-affinity IgG receptor locus knock-in mice, J Allergy Clin Immunol, vol.139, p.1214, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-01397927

F. Jönsson, D. A. Mancardi, W. Zhao, Y. Kita, B. Iannascoli et al., Human FcgammaRIIA induces anaphylactic and allergic reactions, Blood, vol.119, pp.2533-2544, 2012.

V. Papayannopoulos, Neutrophil extracellular traps in immunity and disease, Nat Rev Immunol, vol.18, pp.134-147, 2018.

G. Z. Lotner, J. M. Lynch, S. J. Betz, and P. M. Henson, Human neutrophil-derived platelet activating factor, J Immunol, vol.124, pp.676-684, 1980.

E. Jouvin-marche, E. Ninio, G. Beaurain, M. Tence, P. Niaudet et al., Biosynthesis of Paf-acether (platelet-activating factor). VII. Precursors of Paf-acether and acetyl-transferase activity in human leukocytes, J Immunol, vol.133, pp.892-898, 1984.

S. Ishii, T. Kuwaki, T. Nagase, K. Maki, F. Tashiro et al., Impaired anaphylactic responses with intact sensitivity to endotoxin in mice lacking a platelet-activating factor receptor, J Exp Med, vol.187, pp.1779-1788, 1998.

K. Arias, M. Baig, M. Colangelo, D. Chu, T. Walker et al., Concurrent blockade of platelet-activating factor and histamine prevents life-threatening peanut-induced anaphylactic reactions, J Allergy Clin Immunol, vol.124, pp.301-302, 2009.

P. Vadas, M. Gold, B. Perelman, G. M. Liss, G. Lack et al., Platelet-activating factor, PAF acetylhydrolase, and severe anaphylaxis, N Engl J Med, vol.358, pp.28-35, 2008.

P. Vadas, B. Perelman, and G. Liss, Platelet-activating factor, histamine, and tryptase levels in human anaphylaxis, J Allergy Clin Immunol, vol.131, pp.144-149, 2013.

A. Gouel-cheron, L. De-chaisemartin, F. Jonsson, P. Nicaise-roland, V. Granger et al., Longrois, N. s. group, Low end-tidal CO2 as a real-time severity marker of intra-anaesthetic acute hypersensitivity reactions, Br J Anaesth, vol.119, pp.908-917, 2017.

H. Chou, M. F. Tam, S. S. Lee, R. B. Tang, T. H. Lin et al., Asp159 is a critical core amino acid of an IgE-binding and cross-reactive epitope of a dust mite allergen Der f 7, Mol Immunol, vol.48, pp.2130-2134, 2011.

D. Laroche, S. Chollet-martin, P. Leturgie, L. Malzac, M. C. Vergnaud et al., Evaluation of a new routine diagnostic test for immunoglobulin e sensitization to neuromuscular blocking agents, Anesthesiology, vol.114, pp.91-97, 2011.

B. A. Baldo, M. M. Fisher, and N. H. Pham, On the origin and specificity of antibodies to neuromuscular blocking (muscle relaxant) drugs: an immunochemical perspective, Clin Exp Allergy, vol.39, pp.325-344, 2009.

J. J. Roy and F. Varin, Physicochemical properties of neuromuscular blocking agents and their impact on the pharmacokinetic-pharmacodynamic relationship, Br J Anaesth, vol.93, pp.241-248, 2004.

L. Bergamaschini, T. Santangelo, A. Faricciotti, N. Ciavarella, P. M. Mannucci et al., Study of complement-mediated anaphylaxis in humans. The role of IgG subclasses (IgG1 and/or IgG4) in the complement-activating capacity of immune complexes, J Immunol, vol.156, pp.1256-1261, 1996.

F. Baert, M. Noman, S. Vermeire, G. Van-assche, D. H. et al., Influence of immunogenicity on the long-term efficacy of infliximab in Crohn's disease, N Engl J Med, vol.348, pp.601-608, 2003.

H. Subramanian, K. Gupta, and H. Ali, Roles of Mas-related G protein-coupled receptor X2 on mast cell-mediated host defense, pseudoallergic drug reactions, and chronic inflammatory diseases, J Allergy Clin Immunol, vol.138, pp.700-710, 2016.

B. D. Mcneil, P. Pundir, S. Meeker, L. Han, B. J. Undem et al., Identification of a mast-cell-specific receptor crucial for pseudo-allergic drug reactions, Nature, vol.519, pp.237-241, 2015.

E. Florvaag and S. G. Johansson, The Pholcodine Case. Cough Medicines, IgE-Sensitization, and Anaphylaxis: A Devious Connection, World Allergy Organ J, vol.5, pp.73-78, 2012.

L. K. James and S. J. Till, Potential Mechanisms for IgG4 Inhibition of Immediate Hypersensitivity Reactions, Curr Allergy Asthma Rep, vol.16, p.23, 2016.

H. Z. Movat, T. Uriuhara, N. S. Taichman, H. C. Rowsell, and J. F. Mustard, The role of PMN-leucocyte lysosomes in tissue injury, inflammation and hypersensitivity. VI. The participation of the PMN-leucocyte and the blood platelet in systemic aggregate anaphylaxis, Immunology, vol.14, pp.637-648, 1968.

B. Revenas, G. Smedegard, and T. Saldeen, The pulmonary reaction in aggregate anaphylaxis in the monkey, Acta Chir Scand Suppl, vol.499, pp.161-170, 1980.

G. Smedegard, B. Revenas, and T. Saldeen, Aggregate anaphylaxis in the monkey: haematological and histological findings, Int Arch Allergy Appl Immunol, vol.61, pp.117-124, 1980.

D. Spoerl, H. Nigolian, C. Czarnetzki, and T. Harr, Reclassifying Anaphylaxis to Neuromuscular Blocking Agents Based on the Presumed Patho-Mechanism: IgEMediated, Pharmacological Adverse Reaction or "Innate Hypersensitivity, Int J Mol Sci, vol.18, 2017.

S. E. Mckenzie, S. M. Taylor, P. Malladi, H. Yuhan, D. L. Cassel et al., The role of the human Fc receptor Fc gamma RIIA in the immune clearance of platelets: a transgenic mouse model, J Immunol, vol.162, pp.4311-4318, 1999.

N. Tsuboi, K. Asano, M. Lauterbach, and T. N. Mayadas, Human neutrophil Fcgamma receptors initiate and play specialized nonredundant roles in antibody-mediated inflammatory diseases, Immunity, vol.28, pp.833-846, 2008.

K. Chen, H. Nishi, R. Travers, N. Tsuboi, K. Martinod et al., Endocytosis of soluble immune complexes leads to their clearance by FcgammaRIIIB but induces neutrophil extracellular traps via FcgammaRIIA in vivo, Blood, vol.120, pp.4421-4431, 2012.

B. Balbino, R. Sibilano, P. Starkl, T. Marichal, N. Gaudenzio et al., Pathways of immediate hypothermia and leukocyte infiltration in an adjuvant-free mouse model of anaphylaxis, J Allergy Clin Immunol, vol.139, p.510, 2017.

R. Ahrens, H. Osterfeld, D. Wu, C. Y. Chen, M. Arumugam et al., Intestinal mast cell levels control severity of oral antigen-induced anaphylaxis in mice, Am J Pathol, vol.180, pp.1535-1546, 2012.

L. L. Reber, T. Marichal, K. Mukai, Y. Kita, S. M. Tokuoka et al., Selective ablation of mast cells or basophils reduces peanut-induced anaphylaxis in mice, J Allergy Clin Immunol, vol.132, pp.881-888, 2013.

S. G. Brown, S. F. Stone, D. M. Fatovich, S. A. Burrows, A. Holdgate et al., Anaphylaxis: clinical patterns, mediator release, and severity, J Allergy Clin Immunol, vol.132, p.1145, 2013.

Y. Makabe-kobayashi, Y. Hori, T. Adachi, S. Ishigaki-suzuki, Y. Kikuchi et al., The control effect of histamine on body temperature and respiratory function in IgE-dependent systemic anaphylaxis, J Allergy Clin Immunol, vol.110, pp.298-303, 2002.

P. Gill, N. L. Jindal, A. Jagdis, and P. Vadas, Platelets in the immune response: Revisiting platelet-activating factor in anaphylaxis, J Allergy Clin Immunol, vol.135, pp.1424-1432, 2015.

J. M. Mencia-huerta, R. A. Lewis, E. Razin, and K. F. Austen, Antigen-initiated release of platelet-activating factor (PAF-acether) from mouse bone marrow-derived mast cells sensitized with monoclonal IgE, J Immunol, vol.131, pp.2958-2964, 1983.

L. B. Schwartz, Mediators of human mast cells and human mast cell subsets, Ann Allergy, vol.58, pp.226-235, 1987.

J. Szebeni, F. M. Muggia, and C. R. Alving, Complement activation by Cremophor EL as a possible contributor to hypersensitivity to paclitaxel: an in vitro study, J Natl Cancer Inst, vol.90, pp.300-306, 1998.

M. R. Woolhiser, K. Brockow, and D. D. Metcalfe, Activation of human mast cells by aggregated IgG through FcgammaRI: additive effects of C3a, Clin Immunol, vol.110, pp.172-180, 2004.

M. Khodoun, R. Strait, T. Orekov, S. Hogan, H. Karasuyama et al., Peanuts can contribute to anaphylactic shock by activating complement, J Allergy Clin Immunol, vol.123, pp.342-351, 2009.

R. E. Lewis, J. M. Cruse, and J. V. Richey, Effects of anesthesia and operation on the classical pathway of complement activation, Clin Immunol Immunopathol, vol.23, pp.666-671, 1982.

M. Toussaint, D. J. Jackson, D. Swieboda, A. Guedan, T. D. Tsourouktsoglou et al.,

C. Ching, H. Radermecker, J. Makrinioti, M. R. Aniscenko, R. Edwards et al., Host DNA released by NETosis promotes rhinovirus-induced type-2 allergic asthma exacerbation, Nat Med, vol.23, pp.681-691, 2017.

M. Saffarzadeh and K. T. Preissner, Fighting against the dark side of neutrophil extracellular traps in disease: manoeuvres for host protection, Curr Opin Hematol, vol.20, pp.3-9, 2013.

S. Oehmcke, M. Morgelin, and H. Herwald, Activation of the human contact system on neutrophil extracellular traps, J Innate Immun, vol.1, pp.225-230, 2009.

A. Sala-cunill, J. Bjorkqvist, R. Senter, M. Guilarte, V. Cardona et al., Plasma contact system activation drives anaphylaxis in severe mast cell-mediated allergic reactions, J Allergy Clin Immunol, vol.135, pp.1031-1043, 2015.

R. Munoz-cano, M. Pascal, J. Bartra, C. Picado, A. Valero et al., Distinct transcriptome profiles differentiate nonsteroidal anti-inflammatory drug-dependent from nonsteroidal antiinflammatory drug-independent food-induced anaphylaxis, J Allergy Clin Immunol, vol.137, pp.137-146, 2016.

C. Uermosi, F. Zabel, V. Manolova, M. Bauer, R. R. Beerli et al., IgG-mediated down-regulation of IgE bound to mast cells: a potential novel mechanism of allergen-specific desensitization, Allergy, vol.69, pp.338-347, 2014.

D. Macglashan, M. Mogowski, and L. M. Lichtenstein, Continued expression of antigen-specific IgE during antigeninduced desensitization, Studies of antigen binding on human basophils. II, vol.130, pp.2337-2342, 1983.

C. Kitzmuller, B. Nagl, S. Deifl, C. Walterskirchen, B. Jahn-schmid et al., Human blood basophils do not act as antigen-presenting cells for the major birch pollen allergen Bet v 1, Allergy, vol.67, pp.593-600, 2012.

M. P. Hosking, R. L. Lennon, and G. A. Gronert, Combined H1 and H2 receptor blockade attenuates the cardiovascular effects of high-dose atracurium for rapid sequence endotracheal intubation, Anesth Analg, vol.67, pp.1089-1092, 1988.

S. G. Johansson, J. O. Hourihane, J. Bousquet, C. Bruijnzeel-koomen, S. Dreborg et al., force, A revised nomenclature for allergy. An EAACI position statement from the EAACI nomenclature task force, Allergy, vol.56, pp.813-824, 2001.

J. Ring and K. Messmer, Incidence and severity of anaphylactoid reactions to colloid volume substitutes, Lancet, vol.1, pp.466-469, 1977.

J. Sprung, T. N. Weingarten, and L. B. Schwartz, Presence or absence of elevated acute total serum tryptase by itself is not a definitive marker for an allergic reaction, Anesthesiology, vol.122, pp.713-714, 2015.

A. P. Uyttebroek, V. Sabato, C. H. Bridts, L. S. De-clerck, and D. G. Ebo, Immunoglobulin E antibodies to atracurium: a new diagnostic tool?, Clin Exp Allergy, vol.45, pp.485-487, 2015.

D. G. Ebo, L. Venemalm, C. H. Bridts, F. Degerbeck, H. Hagberg et al., Immunoglobulin E antibodies to rocuronium: a new diagnostic tool, vol.107, pp.253-259, 2007.

S. G. Johansson, H. Oman, F. Degerbeck, J. Tunelli, E. Florvaag et al., Anaphylaxis to atracurium -a non-QAI-dependent reaction?, Acta Anaesthesiol Scand, vol.56, pp.262-263, 2012.

V. Pravettoni, M. Piantanida, L. Primavesi, S. Forti, and E. A. Pastorello, Basal plateletactivating factor acetylhydrolase: prognostic marker of severe Hymenoptera venom anaphylaxis, J Allergy Clin Immunol, vol.133, pp.1218-1220, 2014.

G. Doge, R. Pohloudek-fabini, and D. Kottke, Analysis and stability of suxamethonium chloride. 1. Detection and quantitative determination of the intact active agent with its degradation products, Pharmazie, vol.37, pp.708-711, 1982.

I. Recherche-du-comité-d'interface, CMG was supported partly by a stipend from the Pasteur-Paris University (PPU) International PhD program and by the Institut Carnot Pasteur Maladies Infectieuses. PB benefited from an additional support from AP-HP through a, Contral Local, 2012.

, The sponsor was the Direction de la Rercherche Clinique et de l'Innovation de l'AP-HP (France)

L. Fj, . Ft, and D. L. Ma,

. Hupnvs,

, Inflammation Chimiokines et Immunopathologie, INSERM UMRS996, Faculté de

U. Pharmacie and . Paris-sud, Hôpital Bichat, Département d'Anesthésie-Réanimation, vol.5

H. Aphp,

. Bichat, INSERM

D. Pasteur, . Biologie-structurale, and . Chimie, Unité de Chimie des Biomolécules

, CNRS UMR, vol.3523

, Département d'Anesthésie-Réanimation, Hôpital

A. Beaujon, F. Clichy, and U. Diderot,

L. Mourier, A. Colombes, F. , and U. Paris-diderot,

S. Paris and C. , EA Recherche Clinique coordonnée ville-hôpital, Méthodologies et Société (REMES)

, Département d'Anesthésie-Réanimation, Hôpital Lariboisière

, Service d'Anesthésie-Réanimation, Hôpital Européen Georges Pompidou

F. Paris, U. Paris-descartes, and S. P. Cité, 14 Department of Anesthesia and Critical Care, Institut Pasteur Human Histopathology and Animal Models Unit

, Département d'Anesthésie-Réanimation, Hôpital Saint Louis, AP-HP

H. Service-d'anesthésie and . Foch,

H. Aphp and . Bichat, Service de Pneumologie A

H. Foch, Service de médecine interne, vol.19

S. Université, H. Ap-hp, and . Pitié-salpêtrière,

, Biostatistique Santé Publique et Information Médicale, Unité de Recherche Clinique PSL-CFX , Centre de Pharmacoépidémiologie (Cephepi), INSERM, UMR 1123, p.1421

, 21 Département d'anesthésie-réanimation, Hôpital Cochin, AP-HP