E. E. Perez, J. S. Orange, F. Bonilla, J. Chinen, I. K. Chinn et al., Update on the use of immunoglobulin in human disease: a review of evidence, J Allergy Clin Immunol, vol.139, issue.3S, pp.1-46, 2017.

M. Buttmann, S. Kaveri, and H. P. Hartung, Polyclonal immunoglobulin G for autoimmune demyelinating nervous system disorders, Trends Pharmacol Sci, vol.34, pp.445-57, 2013.

J. D. Lunemann, F. Nimmerjahn, and M. C. Dalakas, Intravenous immunoglobulin in neurology-mode of action and clinical efficacy, Nat Rev Neurol, vol.11, pp.80-89, 2015.

J. Bayry, H. P. Hartung, and S. V. Kaveri, IVIg for relapsing-remitting multiple sclerosis: promises and uncertainties, Trends Pharmacol Sci, vol.36, issue.7, pp.419-440, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01155485

T. Tha-in, J. Bayry, H. J. Metselaar, S. V. Kaveri, and J. Kwekkeboom, Modulation of the cellular immune system by intravenous immunoglobulin, Trends Immunol, vol.29, pp.608-623, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-02455606

I. Schwab and F. Nimmerjahn, Intravenous immunoglobulin therapy: how does IgG modulate the immune system?, Nat Rev Immunol, vol.13, pp.176-89, 2013.

J. F. Seite, Y. Shoenfeld, P. Youinou, and S. Hillion, What is the contents of the magic draft IVIG?, Autoimmun Rev, vol.7, pp.435-444, 2008.

K. Z. Lok, M. Basta, S. Manzanero, and T. V. Arumugam, Intravenous immunoglobulin (IVIG) dampens neuronal toll-like receptor-mediated responses in ischemia, J Neuroinflammation, vol.12, p.73, 2015.

M. S. Maddur, S. Othy, P. Hegde, J. Vani, S. Lacroix-desmazes et al., Immunomodulation by intravenous immunoglobulin: role of regulatory T cells, J Clin Immunol, vol.30, issue.1, pp.4-8, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-02455581

M. S. Maddur, J. Vani, P. Hegde, S. Lacroix-desmazes, S. V. Kaveri et al., Inhibition of differentiation, amplification, and function of human TH17 cells by intravenous immunoglobulin, J Allergy Clin Immunol, vol.127, pp.823-853, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-02455567

C. Galeotti, S. V. Kaveri, and J. Bayry, Molecular and immunological biomarkers to predict IVIg response, Trends Mol Med, vol.21, pp.145-152, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01117084

C. Ritter, I. Bobylev, and H. C. Lehmann, Chronic inflammatory demyelinating polyneuropathy (CIDP): change of serum IgG dimer levels during treatment with intravenous immunoglobulins, J Neuroinflammation, vol.12, p.148, 2015.

S. Sakaguchi, M. Miyara, C. M. Costantino, and D. A. Hafler, FOXP3+ regulatory T cells in the human immune system, Nat Rev Immunol, vol.10, pp.490-500, 2010.

J. Bayry, L. Mouthon, and S. V. Kaveri, Intravenous immunoglobulin expands regulatory T cells in autoimmune rheumatic disease, J Rheumatol, vol.39, pp.450-451, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-02455584

J. Trinath, P. Hegde, M. Sharma, M. S. Maddur, M. Rabin et al., Intravenous immunoglobulin expands regulatory T cells via induction of cyclooxygenase-2-dependent prostaglandin E2 in human dendritic cells, Blood, vol.122, pp.1419-1446, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-02455532

A. Kessel, H. Ammuri, R. Peri, E. R. Pavlotzky, M. Blank et al., Intravenous immunoglobulin therapy affects T regulatory cells by increasing their suppressive function, J Immunol, vol.179, pp.5571-5576, 2007.

L. P. Cousens, R. Tassone, B. D. Mazer, V. Ramachandiran, D. W. Scott et al., Tregitope update: mechanism of action parallels IVIG, Autoimmun Rev, vol.12, pp.436-479, 2013.

A. H. Massoud, M. Yona, D. Xue, F. Chouiali, H. Alturaihi et al., Dendritic cell immunoreceptor: a novel receptor for intravenous immunoglobulin mediates induction of regulatory T cells, J Allergy Clin Immunol, vol.133, pp.853-63, 2014.

R. M. Anthony, T. Kobayashi, F. Wermeling, and J. V. Ravetch, Intravenous gammaglobulin suppresses inflammation through a novel T(H)2 pathway, Nature, vol.475, pp.110-113, 2011.

B. M. Fiebiger, J. Maamary, A. Pincetic, and J. V. Ravetch, Protection in antibody-and T cell-mediated autoimmune diseases by antiinflammatory IgG Fcs requires type II FcRs, Proc Natl Acad Sci U S A, vol.112, pp.2385-94, 2015.

H. C. Lehmann and H. P. Hartung, Plasma exchange and intravenous immunoglobulins: mechanism of action in immune-mediated neuropathies, J Neuroimmunol, vol.231, pp.61-70, 2011.

R. A. Hughes, A. V. Swan, and P. A. Van-doorn, Intravenous immunoglobulin for Guillain-Barre syndrome, Cochrane Database Syst Rev, vol.7, p.2063, 2012.

P. A. Van-doorn, L. Ruts, and B. C. Jacobs, Clinical features, pathogenesis, and treatment of Guillain-Barre syndrome, Lancet Neurol, vol.7, pp.939-50, 2008.

L. J. Chi, H. B. Wang, Y. Zhang, and W. Z. Wang, Abnormality of circulating CD4 + CD25+ regulatory T cell in patients with Guillain-Barre syndrome, J Neuroimmunol, vol.192, pp.206-220, 2007.

X. L. Li, Y. C. Dou, Y. Liu, C. W. Shi, L. L. Cao et al., Atorvastatin ameliorates experimental autoimmune neuritis by decreased Th1/Th17 cytokines and up-regulated T regulatory cells, Cell Immunol, vol.271, pp.455-61, 2011.

L. Gilardin, J. Bayry, and S. V. Kaveri, Intravenous immunoglobulin as clinical immune-modulating therapy, CMAJ, vol.187, pp.257-64, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02455495

G. Zhang, C. A. Massaad, T. Gao, L. Pillai, N. Bogdanova et al., Sialylated intravenous immunoglobulin suppress anti-ganglioside antibody mediated nerve injury, Exp Neurol, vol.282, pp.49-55, 2016.

M. Sudo, Y. Yamaguchi, P. J. Spath, K. Matsumoto-morita, B. K. Ong et al., Different IVIG glycoforms affect in vitro inhibition of anti-ganglioside antibody-mediated complement deposition, PLoS One, vol.9, issue.9, p.107772, 2014.

M. S. Maddur, M. Rabin, P. Hegde, F. Bolgert, M. Guy et al., Intravenous immunoglobulin exerts reciprocal regulation of Th1/ Th17 cells and regulatory T cells in Guillain-Barre syndrome patients, Immunol Res, vol.60, pp.320-329, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01102873

M. Sharma, Y. Schoindre, P. Hegde, C. Saha, M. S. Maddur et al., Intravenous immunoglobulin-induced IL-33 is insufficient to mediate basophil expansion in autoimmune patients, Sci Rep, vol.4, p.5672, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01358914

A. Ephrem, S. Chamat, C. Miquel, S. Fisson, L. Mouthon et al., Expansion of CD4 + CD25+ regulatory T cells by intravenous immunoglobulin: a critical factor in controlling experimental autoimmune encephalomyelitis, Blood, vol.111, pp.715-737, 2008.

F. Y. Liew, J. P. Girard, and H. R. Turnquist, Interleukin-33 in health and disease, Nat Rev Immunol, vol.16, pp.676-89, 2016.

M. S. Maddur, J. Trinath, M. Rabin, F. Bolgert, M. Guy et al., Intravenous immunoglobulin-mediated expansion of regulatory T cells in autoimmune patients is associated with increased prostaglandin E2 levels in the circulation, Cell Mol Immunol, vol.12, pp.650-652, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02455500

H. C. Kuo, C. L. Wang, K. D. Yang, M. H. Lo, K. S. Hsieh et al., Plasma prostaglandin E2 levels correlated with the prevention of intravenous immunoglobulin resistance and coronary artery lesions formation via CD40L in Kawasaki disease, PLoS One, vol.11, p.161265, 2016.

O. E. Savenije, M. John, J. M. Granell, R. Kerkhof, M. Dijk et al., Association of IL33-IL-1 receptor-like 1 (IL1RL1) pathway polymorphisms with wheezing phenotypes and asthma in childhood, J Allergy Clin Immunol, vol.134, pp.170-177, 2014.

S. Liu, Z. Zhou, C. Wang, M. Guo, N. Chu et al., Associations between interleukin and interleukin receptor gene polymorphisms and risk of gout, Sci Rep, vol.5, p.13887, 2015.

A. K. Fu, K. W. Hung, M. Y. Yuen, X. Zhou, D. S. Mak et al., IL-33 ameliorates Alzheimer's disease-like pathology and cognitive decline, Proc Natl Acad Sci, vol.113, pp.2705-2718, 2016.

P. Korhonen, K. M. Kanninen, S. Lehtonen, S. Lemarchant, K. A. Puttonen et al., Immunomodulation by interleukin-33 is protective in stroke through modulation of inflammation, Brain Behav Immun, vol.49, pp.322-358, 2015.

A. G. Besnard, R. Guabiraba, W. Niedbala, J. Palomo, F. Reverchon et al., IL-33-mediated protection against experimental cerebral malaria is linked to induction of type 2 innate lymphoid cells, M2 macrophages and regulatory T cells, PLoS Pathog, vol.11, issue.2, p.1004607, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01190147

C. Dai, F. N. Lu, J. N. Yang, B. Gao, C. Zhao et al., Recombinant IL-33 prolongs leflunomide-mediated graft survival by reducing IFN-gamma and expanding CD4 + Foxp3+ T cells in concordant heart transplantation, Lab Invest, vol.96, issue.8, pp.820-829, 2016.

R. F. Franca, R. S. Costa, J. R. Silva, R. S. Peres, L. R. Mendonca et al., IL-33 signaling is essential to attenuate viral-induced encephalitis development by downregulating iNOS expression in the central nervous system, J Neuroinflammation, vol.13, issue.1, p.159, 2016.