IgG subclasses determine pathways of anaphylaxis in mice

Background: Animal models have demonstrated that allergen‐specific IgG confers sensitivity to systemic anaphylaxis that relies on IgG Fc receptors (Fc&ggr;Rs). Mouse IgG2a and IgG2b bind activating Fc&ggr;RI, Fc&ggr;RIII, and Fc&ggr;RIV and inhibitory Fc&ggr;RIIB; mouse IgG1 binds only Fc&ggr;RIII and Fc&ggr;RIIB. Although these interactions are of strikingly different affinities, these 3 IgG subclasses have been shown to enable induction of systemic anaphylaxis. Objective: We sought to determine which pathways control the induction of IgG1‐, IgG2a‐, and IgG2b‐dependent passive systemic anaphylaxis. Methods: Mice were sensitized with IgG1, IgG2a, or IgG2b anti‐trinitrophenyl mAbs and challenged with trinitrophenyl‐BSA intravenously to induce systemic anaphylaxis that was monitored by using rectal temperature. Anaphylaxis was evaluated in mice deficient for Fc&ggr;Rs injected with mediator antagonists or in which basophils, monocytes/macrophages, or neutrophils had been depleted. Fc&ggr;R expression was evaluated on these cells before and after anaphylaxis. Results: Activating Fc&ggr;RIII is the receptor primarily responsible for all 3 models of anaphylaxis, and subsequent downregulation of this receptor was observed. These models differentially relied on histamine release and the contribution of mast cells, basophils, macrophages, and neutrophils. Strikingly, basophil contribution and histamine predominance in mice with IgG1‐ and IgG2b‐induced anaphylaxis correlated with the ability of inhibitory Fc&ggr;RIIB to negatively regulate these models of anaphylaxis. Conclusion: We propose that the differential expression of inhibitory Fc&ggr;RIIB on myeloid cells and its differential binding of IgG subclasses controls the contributions of mast cells, basophils, neutrophils, and macrophages to IgG subclass–dependent anaphylaxis. Collectively, our results unravel novel complexities in the involvement and regulation of cell populations in IgG‐dependent reactions in vivo.

[1]  M. Capron Eosinophils: receptors and mediators in hypersensitivity. , 1989, Clinical and experimental allergy : journal of the British Society for Allergy and Clinical Immunology.

[2]  S. Akilesh,et al.  FcRn: the neonatal Fc receptor comes of age , 2007, Nature Reviews Immunology.

[3]  P. Bruhns,et al.  Regulation of allergy by Fc receptors. , 2005, Current opinion in immunology.

[4]  C. Chen,et al.  Reduced mast cell and basophil numbers and function in Cpa3-Cre; Mcl-1fl/fl mice. , 2011, Blood.

[5]  J. Stavenhagen,et al.  Monoclonal antibodies capable of discriminating the human inhibitory Fcγ‐receptor IIB (CD32B) from the activating Fcγ‐receptor IIA (CD32A): biochemical, biological and functional characterization , 2007, Immunology.

[6]  Calman Prussin,et al.  IgE, mast cells, basophils, and eosinophils. , 2003, The Journal of allergy and clinical immunology.

[7]  F. Finkelman,et al.  Molecular mechanisms of anaphylaxis: lessons from studies with murine models. , 2005, The Journal of allergy and clinical immunology.

[8]  J. Ravetch,et al.  FcγRIV deletion reveals its central role for IgG2a and IgG2b activity in vivo , 2010, Proceedings of the National Academy of Sciences.

[9]  L. Cassard,et al.  Inhibit Mouse and Human Basophil Activation , 2012 .

[10]  D. Voehringer,et al.  Basophils orchestrate chronic allergic dermatitis and protective immunity against helminths. , 2010, Immunity.

[11]  F. Finkelman,et al.  Pathways of anaphylaxis in the mouse. , 2002, The Journal of allergy and clinical immunology.

[12]  F. Finkelman,et al.  Identification of markers that distinguish IgE- from IgG-mediated anaphylaxis , 2011, Proceedings of the National Academy of Sciences.

[13]  Takao Shimizu,et al.  Basophils play a pivotal role in immunoglobulin-G-mediated but not immunoglobulin-E-mediated systemic anaphylaxis. , 2008, Immunity.

[14]  B. Nieswandt,et al.  Both FcγRIV and FcγRIII are essential receptors mediating type II and type III autoimmune responses via FcRγ‐LAT‐dependent generation of C5a , 2009, European journal of immunology.

[15]  P. Hogarth,et al.  Identification of the mouse IgG3 receptor: implications for antibody effector function at the interface between innate and adaptive immunity. , 1998, Journal of immunology.

[16]  P. Bruhns,et al.  FcgammaRIV is a mouse IgE receptor that resembles macrophage FcepsilonRI in humans and promotes IgE-induced lung inflammation. , 2008, The Journal of clinical investigation.

[17]  L. Buéno,et al.  Effects of neuropeptide FF on intestinal motility and temperature changes induced by endotoxin and platelet-activating factor. , 1997, European journal of pharmacology.

[18]  L. Cassard,et al.  Fcγ Receptors Inhibit Mouse and Human Basophil Activation , 2012, The Journal of Immunology.

[19]  A. Nagy,et al.  The control effect of histamine on body temperature and respiratory function in IgE-dependent systemic anaphylaxis. , 2002, The Journal of allergy and clinical immunology.

[20]  S. Galli,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. , 1997, The Journal of clinical investigation.

[21]  T. Yoshino,et al.  Modulation of Immunoglobulin (Ig)E-mediated Systemic Anaphylaxis by Low-Affinity Fc Receptors for IgG , 1999, The Journal of experimental medicine.

[22]  M. Daëron,et al.  Mast Cells and Company , 2012, Front. Immun..

[23]  D. Sears,et al.  Unique Monoclonal Antibodies Define Expression of FcγRI on Macrophages and Mast Cell Lines and Demonstrate Heterogeneity Among Subcutaneous and Other Dendritic Cells 1 , 2003, The Journal of Immunology.

[24]  G. Camussi,et al.  Release of platelet-activating factor (PAF) and histamine. II. The cellular origin of human PAF: monocytes, polymorphonuclear neutrophils and basophils. , 1981, Immunology.

[25]  J. Wedemeyer,et al.  Roles of mast cells and basophils in innate and acquired immunity. , 2000, Current opinion in immunology.

[26]  M. Cragg,et al.  Development and characterisation of monoclonal antibodies specific for the murine inhibitory FcγRIIB (CD32B) , 2012, European journal of immunology.

[27]  P. Bruhns,et al.  Cutting Edge: The Murine High-Affinity IgG Receptor FcγRIV Is Sufficient for Autoantibody-Induced Arthritis , 2011, The Journal of Immunology.

[28]  H. Eisen,et al.  Binding of monomeric immunoglobulins to Fc receptors of mouse macrophages , 1975, The Journal of experimental medicine.

[29]  Wei Zhao,et al.  Human FcγRIIA induces anaphylactic and allergic reactions. , 2012, Blood.

[30]  P. Zhu,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. , 2014, Cellular immunology.

[31]  P. Bruhns Properties of mouse and human IgG receptors and their contribution to disease models. , 2012, Blood.

[32]  Kenneth G. C. Smith,et al.  FcγRIIB in autoimmunity and infection: evolutionary and therapeutic implications , 2010, Nature Reviews Immunology.

[33]  M. Biburger,et al.  Low level of FcγRIII expression on murine natural killer cells. , 2012, Immunology letters.

[34]  Y. Saeys,et al.  The function of Fcγ receptors in dendritic cells and macrophages , 2014, Nature Reviews Immunology.

[35]  N. Van Rooijen,et al.  Liposome mediated depletion of macrophages: mechanism of action, preparation of liposomes and applications. , 1994, Journal of immunological methods.

[36]  P. Bruhns,et al.  Specificity and affinity of human Fcgamma receptors and their polymorphic variants for human IgG subclasses. , 2009, Blood.

[37]  M. Cragg,et al.  Anti-mouse FcγRIV antibody 9E9 also blocks FcγRIII in vivo. , 2015, Blood.

[38]  D. Dudziak,et al.  Monocyte subsets responsible for immunoglobulin G-dependent effector functions in vivo. , 2011, Immunity.

[39]  P. Bruhns,et al.  The high-affinity human IgG receptor FcγRI (CD64) promotes IgG-mediated inflammation, anaphylaxis, and antitumor immunotherapy. , 2013, Blood.

[40]  A. Casadevall,et al.  Murine IgG1 and IgG3 Isotype Switch Variants Promote Phagocytosis of Cryptococcus neoformans through Different Receptors , 2009, The Journal of Immunology.

[41]  P. Bruhns,et al.  Neutrophils in local and systemic antibody‐dependent inflammatory and anaphylactic reactions , 2013, Journal of leukocyte biology.

[42]  K. Nadeau,et al.  Selective ablation of mast cells or basophils reduces peanut-induced anaphylaxis in mice. , 2013, The Journal of allergy and clinical immunology.

[43]  F. Finkelman,et al.  Rapid desensitization of mice with anti-FcγRIIb/FcγRIII mAb safely prevents IgG-mediated anaphylaxis. , 2013, The Journal of allergy and clinical immunology.

[44]  P. Bruhns,et al.  FcγRIV : A novel FcR with distinct IgG subclass specificity , 2005 .

[45]  J. Ravetch,et al.  Divergent Immunoglobulin G Subclass Activity Through Selective Fc Receptor Binding , 2005, Science.

[46]  Takao Shimizu,et al.  Mouse and human neutrophils induce anaphylaxis. , 2011, The Journal of clinical investigation.

[47]  T. Möröy,et al.  Gfi1:Green Fluorescent Protein Knock-in Mutant Reveals Differential Expression and Autoregulation of the Growth Factor Independence 1 (Gfi1) Gene during Lymphocyte Development* , 2004, Journal of Biological Chemistry.

[48]  P. Bruhns,et al.  FcgammaRIV: a novel FcR with distinct IgG subclass specificity. , 2005, Immunity.

[49]  G. Isbister,et al.  Anaphylaxis: clinical patterns, mediator release, and severity. , 2013, The Journal of allergy and clinical immunology.