Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Eur J Immunol ; 52(5): 753-759, 2022 05.
Article in English | MEDLINE | ID: mdl-35133670

ABSTRACT

IgG is the predominant antibody class generated during infections and used for the generation of therapeutic antibodies. Antibodies are mainly characterized in or generated from animal models that support particular infections, respond to particular antigens or allow the generation of hybridomas. Due to the availability of numerous transgenic mouse models and the ease of performing bioassays with human blood cells in vitro, most antibodies from species other than mice and humans are tested in vitro using human cells and/or in vivo using mice. In this process, it is expected, but not yet systematically documented, that IgG from these species interact with human or mouse IgG receptors (FcγRs). In this study, we undertook a systematic assessment of binding specificities of IgG from various species to the families of mouse and human FcγRs including their polymorphic variants. Our results document the specific binding patterns for each of these IgG (sub)classes, reveal possible caveats of antibody-based immunoassays, and will be a useful reference for the transition from one animal model to preclinical mouse models or human cell-based bioassays.


Subject(s)
Immunoglobulin G , Receptors, IgG , Animals , Carrier Proteins/metabolism , Humans , Hybridomas , Mice , Mice, Transgenic
2.
J Allergy Clin Immunol ; 139(4): 1253-1265.e14, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27568081

ABSTRACT

BACKGROUND: Anaphylaxis can proceed through distinct IgE- or IgG-dependent pathways, which have been investigated in various mouse models. We developed a novel mouse strain in which the human low-affinity IgG receptor locus, comprising both activating (hFcγRIIA, hFcγRIIIA, and hFcγRIIIB) and inhibitory (hFcγRIIB) hFcγR genes, has been inserted into the equivalent murine locus, corresponding to a locus swap. OBJECTIVE: We sought to determine the capabilities of hFcγRs to induce systemic anaphylaxis and identify the cell types and mediators involved. METHODS: hFcγR expression on mouse and human cells was compared to validate the model. Passive systemic anaphylaxis was induced by injection of heat-aggregated human intravenous immunoglobulin and active systemic anaphylaxis after immunization and challenge. Anaphylaxis severity was evaluated based on hypothermia and mortality. The contribution of receptors, mediators, or cell types was assessed based on receptor blockade or depletion. RESULTS: The human-to-mouse low-affinity FcγR locus swap engendered hFcγRIIA/IIB/IIIA/IIIB expression in mice comparable with that seen in human subjects. Knock-in mice were susceptible to passive and active anaphylaxis, accompanied by downregulation of both activating and inhibitory hFcγR expression on specific myeloid cells. The contribution of hFcγRIIA was predominant. Depletion of neutrophils protected against hypothermia and mortality. Basophils contributed to a lesser extent. Anaphylaxis was inhibited by platelet-activating factor receptor or histamine receptor 1 blockade. CONCLUSION: Low-affinity FcγR locus-switched mice represent an unprecedented model of cognate hFcγR expression. Importantly, IgG-related anaphylaxis proceeds within a native context of activating and inhibitory hFcγRs, indicating that, despite robust hFcγRIIB expression, activating signals can dominate to initiate a severe anaphylactic reaction.


Subject(s)
Anaphylaxis/immunology , Receptors, IgG/immunology , Animals , Disease Models, Animal , GPI-Linked Proteins/immunology , Gene Knock-In Techniques , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL
3.
J Allergy Clin Immunol ; 139(1): 269-280.e7, 2017 01.
Article in English | MEDLINE | ID: mdl-27246523

ABSTRACT

BACKGROUND: Animal models have demonstrated that allergen-specific IgG confers sensitivity to systemic anaphylaxis that relies on IgG Fc receptors (FcγRs). Mouse IgG2a and IgG2b bind activating FcγRI, FcγRIII, and FcγRIV and inhibitory FcγRIIB; mouse IgG1 binds only FcγRIII and FcγRIIB. 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 FcγRs injected with mediator antagonists or in which basophils, monocytes/macrophages, or neutrophils had been depleted. FcγR expression was evaluated on these cells before and after anaphylaxis. RESULTS: Activating FcγRIII 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 FcγRIIB to negatively regulate these models of anaphylaxis. CONCLUSION: We propose that the differential expression of inhibitory FcγRIIB 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.


Subject(s)
Anaphylaxis/immunology , Immunoglobulin G/immunology , Protein Subunits/immunology , Animals , Antibodies, Monoclonal/immunology , Female , Haptens/immunology , Histamine/immunology , Immunoglobulin E/immunology , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology , Serum Albumin, Bovine/immunology
4.
J Autoimmun ; 80: 95-102, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27745779

ABSTRACT

Two activating mouse IgG receptors (FcγRs) have the ability to bind monomeric IgG, the high-affinity mouse FcγRI and FcγRIV. Despite high circulating levels of IgG, reports using FcγRI-/- or FcγRIV-/- mice or FcγRIV-blocking antibodies implicate these receptors in IgG-induced disease severity or therapeutic Ab efficacy. From these studies, however, one cannot conclude on the effector capabilities of a given receptor, because different activating FcγRs possess redundant properties in vivo, and cooperation between FcγRs may occur, or priming phenomena. To help resolve these uncertainties, we used mice expressing only FcγRI to determine its intrinsic properties in vivo. FcγRIonly mice were sensitive to IgG-induced autoimmune thrombocytopenia and anti-CD20 and anti-tumour immunotherapy, but resistant to IgG-induced autoimmune arthritis, anaphylaxis and airway inflammation. Our results show that the in vivo roles of FcγRI are more restricted than initially reported using FcγRI-/- mice, but confirm effector capabilities for this high-affinity IgG receptor in vivo.


Subject(s)
Antibodies, Blocking/therapeutic use , B-Lymphocytes/immunology , Immunotherapy/methods , Purpura, Thrombocytopenic, Idiopathic/immunology , Receptors, IgG/metabolism , Animals , Antibody Affinity , Disease Models, Animal , Hepatectomy , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Purpura, Thrombocytopenic, Idiopathic/therapy , Receptors, IgG/genetics , Splenectomy
5.
Blood ; 121(9): 1563-73, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23293080

ABSTRACT

Receptors for the Fc portion of IgG (FcγRs) are mandatory for the induction of various IgG-dependent models of autoimmunity, inflammation, anaphylaxis, and cancer immunotherapy. A few FcγRs have the ability to bind monomeric IgG: high-affinity mouse mFcγRI, mFcγRIV, and human hFcγRI. All others bind IgG only when aggregated in complexes or bound to cells or surfaces: low-affinity mouse mFcγRIIB and mFcγRIII and human hFcγRIIA/B/C and hFcγRIIIA/B. Although it has been proposed that high-affinity FcγRs are occupied by circulating IgG, multiple roles for mFcγRI and mFcγRIV have been reported in vivo. However, the potential roles of hFcγRI that is expressed on monocytes, macrophages, and neutrophils have not been reported. In the present study, we therefore investigated the role of hFcγRI in antibody-mediated models of disease and therapy by generating hFcγRI-transgenic mice deficient for multiple endogenous FcRs. hFcγRI was sufficient to trigger autoimmune arthritis and thrombocytopenia, immune complex-induced airway inflammation, and active and passive systemic anaphylaxis. We found monocyte/macrophages to be responsible for thrombocytopenia, neutrophils to be responsible for systemic anaphylaxis, and both cell types to be responsible for arthritis induction. Finally, hFcγRI was capable of mediating antibody-induced immunotherapy of metastatic melanoma. Our results unravel novel capabilities of human FcγRI that confirm the role of high-affinity IgG receptors in vivo.


Subject(s)
Anaphylaxis/genetics , Immunoglobulin G/physiology , Immunotherapy , Inflammation/genetics , Neoplasms/therapy , Receptors, IgG/physiology , Anaphylaxis/immunology , Animals , Antineoplastic Agents/pharmacology , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Humans , Immunoglobulin G/pharmacology , Immunotherapy/methods , Inflammation/chemically induced , Inflammation/immunology , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Receptors, IgG/genetics , Xenograft Model Antitumor Assays
6.
Blood ; 122(18): 3160-4, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-23980063

ABSTRACT

Tumor engraftment followed by monoclonal antibody (mAb) therapy targeting tumor antigens represents a gold standard for assessing the efficiency of mAbs to eliminate tumor cells. Mouse models have demonstrated that receptors for the Fc portion of immunoglobulin G (FcγRs) are critical determinants of mAb therapeutic efficacy, but the FcγR-expressing cell populations responsible remain elusive. We show that neutrophils are responsible for mAb-induced therapy of both subcutaneous syngeneic melanoma and human breast cancer xenografts. mAb-induced tumor reduction, abolished in neutropenic mice, could be restored in FcγR-deficient hosts upon transfer of FcγR+ neutrophils or upon human FcγRIIA/CD32A transgenic expression. Finally, conditional knockout mice unable to perform FcγR-mediated activation and phagocytosis specifically in neutrophils were resistant to mAb-induced therapy. Our work suggests that neutrophils are necessary and sufficient for mAb-induced therapy of subcutaneous tumors, and represent a new and critical focal point for optimizing mAb-induced immunotherapies that will impact on human cancer treatment.


Subject(s)
Antibodies, Monoclonal/immunology , Breast Neoplasms/immunology , Melanoma, Experimental/immunology , Neutrophils/immunology , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/genetics , Antibody-Dependent Cell Cytotoxicity/immunology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Mice, Transgenic , Neutrophils/drug effects , Neutrophils/metabolism , Receptors, IgG/genetics , Receptors, IgG/immunology , Receptors, IgG/metabolism , Rituximab , Trastuzumab , Tumor Burden/drug effects , Tumor Burden/immunology
7.
Blood ; 119(11): 2533-44, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22138510

ABSTRACT

IgE and IgE receptors (FcεRI) are well-known inducers of allergy. We recently found in mice that active systemic anaphylaxis depends on IgG and IgG receptors (FcγRIIIA and FcγRIV) expressed by neutrophils, rather than on IgE and FcεRI expressed by mast cells and basophils. In humans, neutrophils, mast cells, basophils, and eosinophils do not express FcγRIIIA or FcγRIV, but FcγRIIA. We therefore investigated the possible role of FcγRIIA in allergy by generating novel FcγRIIA-transgenic mice, in which various models of allergic reactions induced by IgG could be studied. In mice, FcγRIIA was sufficient to trigger active and passive anaphylaxis, and airway inflammation in vivo. Blocking FcγRIIA in vivo abolished these reactions. We identified mast cells to be responsible for FcγRIIA-dependent passive cutaneous anaphylaxis, and monocytes/macrophages and neutrophils to be responsible for FcγRIIA-dependent passive systemic anaphylaxis. Supporting these findings, human mast cells, monocytes and neutrophils produced anaphylactogenic mediators after FcγRIIA engagement. IgG and FcγRIIA may therefore contribute to allergic and anaphylactic reactions in humans.


Subject(s)
Hypersensitivity/etiology , Hypersensitivity/pathology , Inflammation/etiology , Inflammation/pathology , Passive Cutaneous Anaphylaxis , Receptors, IgG/physiology , Respiratory System/pathology , Animals , Cells, Cultured , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , Humans , Hypersensitivity/metabolism , Inflammation/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Respiratory System/metabolism
8.
J Immunol ; 189(12): 5513-7, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23150715

ABSTRACT

mAb therapy for experimental metastatic melanoma relies on activating receptors for the Fc portion of IgG (FcγR). Opposing results on the respective contribution of mouse FcγRI, FcγRIII, and FcγRIV have been reported using the gp75-expressing B16 melanoma and the protective anti-gp75 mAb TA99. We analyzed the contribution of FcγRs to this therapy model using bioluminescent measurement of lung metastases loads, novel mouse strains, and anti-FcγR blocking mAbs. We found that the TA99 mAb-mediated effects in a combination therapy using cyclophosphamide relied on activating FcγRs. The combination therapy, however, was not more efficient than mAb therapy alone. We demonstrate that FcγRI and, unexpectedly, FcγRIII contributed to TA99 mAb therapeutic effects, whereas FcγRIV did not. Therefore, FcγRIII and FcγRI are, together, responsible for anti-gp75 mAb therapy of B16 lung metastases. Our finding that mouse FcγRIII contributes to Ab-induced tumor reduction correlates with clinical data on its human functional equivalent human FcγRIIIA (CD16A).


Subject(s)
Antibodies, Monoclonal/therapeutic use , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Receptors, IgG/therapeutic use , Viral Proteins/immunology , Animals , Antibodies, Blocking/therapeutic use , Arboviruses/immunology , Hybridomas , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, IgG/deficiency , Receptors, IgG/genetics
9.
Eur J Immunol ; 42(9): 2395-408, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22684987

ABSTRACT

Using N-ethyl-N-nitrosourea-induced mutagenesis, we established a mouse model with a novel form of neutropenia resulting from a point mutation in the transcriptional repressor Growth Factor Independence 1 (Gfi1). These mice, called Genista, had normal viability and no weight loss, in contrast to mice expressing null alleles of the Gfi1 gene. Furthermore, the Genista mutation had a very limited impact on lymphopoiesis or on T- and B-cell function. Within the bone marrow (BM), the Genista mutation resulted in a slight increase of monopoiesis and in a block of terminal granulopoiesis. This block occurred just after the metamyelocytic stage and resulted in the generation of small numbers of atypical CD11b(+) Ly-6G(int) neutrophils, the nuclear morphology of which resembled that of mature WT neutrophils. Unexpectedly, once released from the BM, these atypical neutrophils contributed to induce mild forms of autoantibody-induced arthritis and of immune complex-mediated lung alveolitis. They additionally failed to provide resistance to acute bacterial infection. Our study demonstrates that a hypomorphic mutation in the Gfi1 transcriptional repressor results in a novel form of neutropenia characterized by a split pattern of functional responses, reflecting the distinct thresholds required for eliciting neutrophil-mediated inflammatory and anti-infectious responses.


Subject(s)
DNA-Binding Proteins/genetics , Neutropenia/genetics , Point Mutation , Repressor Proteins/genetics , Transcription Factors/genetics , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Arthritis/genetics , Arthritis/metabolism , B-Lymphocytes/metabolism , Bone Marrow/metabolism , CD11b Antigen/genetics , CD11b Antigen/metabolism , DNA-Binding Proteins/metabolism , Ethylnitrosourea , Female , Inflammation/genetics , Inflammation/metabolism , Lymphocytes/metabolism , Lymphopoiesis/genetics , Male , Mice , Mice, Inbred C57BL , Neutropenia/chemically induced , Neutrophils/metabolism , Repressor Proteins/metabolism , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Transcription, Genetic
10.
J Immunol ; 186(4): 1899-903, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21248252

ABSTRACT

K/BxN serum-induced passive arthritis was reported to depend on the activation of mast cells, triggered by the activating IgG receptor FcγRIIIA, when engaged by IgG1 autoantibodies present in K/BxN serum. This view is challenged by the fact that FcγRIIIA-deficient mice still develop K/BxN arthritis and because FcγRIIIA is the only activating IgG receptor expressed by mast cells. We investigated the contribution of IgG receptors, IgG subclasses, and cells in K/BxN arthritis. We found that the activating IgG2 receptor FcγRIV, expressed only by monocytes/macrophages and neutrophils, was sufficient to induce disease. K/BxN arthritis occurred not only in mast cell-deficient W(sh) mice, but also in mice whose mast cells express no activating IgG receptors. We propose that at least two autoantibody isotypes, IgG1 and IgG2, and two activating IgG receptors, FcγRIIIA and FcγRIV, contribute to K/BxN arthritis, which requires at least two cell types other than mast cells, monocytes/macrophages, and neutrophils.


Subject(s)
Arthritis, Experimental/immunology , Autoantibodies/physiology , Binding Sites, Antibody , Receptors, IgG/physiology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Autoantibodies/blood , Immunoglobulin G/metabolism , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Receptors, IgG/deficiency , Receptors, IgG/genetics , Serum/physiology
11.
J Immunol ; 187(5): 2646-55, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21810608

ABSTRACT

Some nonpathogenic bacteria were found to have protective effects in mouse models of allergic and autoimmune diseases. These "probiotics" are thought to interact with dendritic cells during Ag presentation, at the initiation of adaptive immune responses. Many other myeloid cells are the effector cells of immune responses. They are responsible for inflammation that accounts for symptoms in allergic and autoimmune diseases. We investigated in this study whether probiotics might affect allergic and autoimmune inflammation by acting at the effector phase of adaptive immune responses. The effects of one strain of Lactobacillus casei were investigated in vivo on IgE-induced passive systemic anaphylaxis and IgG-induced passive arthritis, two murine models of acute allergic and autoimmune inflammation, respectively, which bypass the induction phase of immune responses, in vitro on IgE- and IgG-induced mouse mast cell activation and ex vivo on IgE-dependent human basophil activation. L. casei protected from anaphylaxis and arthritis, and inhibited mouse mast cell and human basophil activation. Inhibition required contact between mast cells and bacteria, was reversible, and selectively affected the Lyn/Syk/linker for activation of T cells pathway induced on engagement of IgE receptors, leading to decreased MAPK activation, Ca(2+) mobilization, degranulation, and cytokine secretion. Also, adoptive anaphylaxis induced on Ag challenge in mice injected with IgE-sensitized mast cells was abrogated in mice injected with IgE-sensitized mast cells exposed to bacteria. These results demonstrate that probiotics can influence the effector phase of adaptive immunity in allergic and autoimmune diseases. They might, therefore, prevent inflammation in patients who have already synthesized specific IgE or autoantibodies.


Subject(s)
Autoimmunity/immunology , Hypersensitivity/immunology , Inflammation/immunology , Lacticaseibacillus casei/immunology , Probiotics/pharmacology , Adaptive Immunity/drug effects , Adaptive Immunity/immunology , Anaphylaxis/immunology , Animals , Arthritis, Experimental/immunology , Basophils/drug effects , Basophils/immunology , Blotting, Western , Humans , Hypersensitivity/prevention & control , Immunoglobulin E/immunology , Inflammation/prevention & control , Mast Cells/drug effects , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
12.
J Clin Invest ; 118(11): 3738-50, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18949059

ABSTRACT

FcgammaRIV is a recently identified mouse activating receptor for IgG2a and IgG2b that is expressed on monocytes, macrophages, and neutrophils; herein it is referred to as mFcgammaRIV. Although little is known about mFcgammaRIV, it has been proposed to be the mouse homolog of human FcgammaRIIIA (hFcgammaRIIIA) because of high sequence homology. Our work, however, has revealed what we believe to be new properties of mFcgammaRIV that endow this receptor with a previously unsuspected biological significance; we have shown that it is a low-affinity IgE receptor for all IgE allotypes. Although mFcgammaRIV functioned as a high-affinity IgG receptor, mFcgammaRIV-bound monomeric IgGs were readily displaced by IgE immune complexes. Engagement of mFcgammaRIV by IgE immune complexes induced bronchoalveolar and peritoneal macrophages to secrete cytokines, suggesting that mFcgammaRIV may be an equivalent of human FceRI(alphagamma), which is expressed by macrophages and neutrophils and especially in atopic individuals, rather than an equivalent of hFcgammaRIIIA, which has no affinity for IgE. Using mice lacking 3 FcgammaRs and 2 FceRs and expressing mFcgammaRIV only, we further demonstrated that mFcgammaRIV promotes IgE-induced lung inflammation. These data lead us to propose a mouse model of IgE-induced lung inflammation in which cooperation exists between mast cells and mFcgammaRIV-expressing lung cells. We therefore suggest that a similar cooperation may occur between mast cells and hFceRI-expressing lung cells in human allergic asthma.


Subject(s)
Immunoglobulin E/physiology , Inflammation , Lung/immunology , Macrophages/immunology , Receptors, IgE/physiology , Receptors, IgG/physiology , Animals , Bone Marrow Cells/cytology , Cell Line , Humans , Immunoglobulin E/immunology , Kidney/cytology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, IgE/immunology , Receptors, IgG/immunology
13.
Blood ; 113(16): 3716-25, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19018092

ABSTRACT

Distinct genes encode 6 human receptors for IgG (hFcgammaRs), 3 of which have 2 or 3 polymorphic variants. The specificity and affinity of individual hFcgammaRs for the 4 human IgG subclasses is unknown. This information is critical for antibody-based immunotherapy which has been increasingly used in the clinics. We investigated the binding of polyclonal and monoclonal IgG1, IgG2, IgG3, and IgG4 to FcgammaRI; FcgammaRIIA, IIB, and IIC; FcgammaRIIIA and IIIB; and all known polymorphic variants. Wild-type and low-fucosylated IgG1 anti-CD20 and anti-RhD mAbs were also examined. We found that (1) IgG1 and IgG3 bind to all hFcgammaRs; (2) IgG2 bind not only to FcgammaRIIA(H131), but also, with a lower affinity, to FcgammaRIIA(R131) and FcgammaRIIIA(V158); (3) IgG4 bind to FcgammaRI, FcgammaRIIA, IIB and IIC and FcgammaRIIIA(V158); and (4) the inhibitory receptor FcgammaRIIB has a lower affinity for IgG1, IgG2, and IgG3 than all other hFcgammaRs. We also identified parameters that determine the specificity and affinity of hFcgammaRs for IgG subclasses. These results document how hFcgammaR specificity and affinity may account for the biological activities of antibodies. They therefore highlight the role of specific hFcgammaRs in the therapeutic and pathogenic effects of antibodies in disease.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin G/immunology , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal/genetics , CHO Cells , Cricetinae , Cricetulus , Humans , Immunoglobulin G/genetics , Mice , Protein Binding/genetics , Protein Binding/immunology , Receptors, IgG/genetics
15.
J Leukoc Biol ; 94(4): 643-56, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23532517

ABSTRACT

Neutrophils are notorious for their efficacy in microbial killing. Various mechanisms, such as phagocytosis, production of ROS, cytokines/chemokines and lipid mediators, degranulation of antimicrobials and enzymes, as well as NETosis contribute to this capacity. However, every incidence of neutrophil activation bears a risk to cause damage to the host. Several distinct steps, i.e., adhesion to endothelial cells, transmigration, chemotaxis, cytokine stimulation, and TLR signaling, are thought to control the extent of neutrophil activation. In the absence of a microbial stimulus, other pathways can induce neutrophil activation, among which FcR-induced activation when neutrophils encounter ICs. In these situations (inflammation, autoimmunity, allergy), neutrophils may act as primary or secondary effectors of immune reactions. In the presence of circulating ICs, neutrophils can indeed get stimulated directly in the bloodstream and trigger an immune response. Upon deposition of antibody complexes inside of tissues, neutrophils are first recruited and primed before being highly activated to amplify the ongoing inflammation. This review focuses on the engagement, activation, and responses of neutrophils to antibody ICs, inside of tissues or in the vasculature.


Subject(s)
Anaphylaxis/immunology , Anaphylaxis/pathology , Antibodies/immunology , Inflammation/immunology , Inflammation/pathology , Neutrophils/immunology , Animals , Antigen-Antibody Complex/metabolism , Humans , Organ Specificity/immunology
16.
J Clin Invest ; 121(4): 1484-96, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21436586

ABSTRACT

Anaphylaxis is a life-threatening hyperacute immediate hypersensitivity reaction. Classically, it depends on IgE, FcεRI, mast cells, and histamine. However, anaphylaxis can also be induced by IgG antibodies, and an IgG1-induced passive type of systemic anaphylaxis has been reported to depend on basophils. In addition, it was found that neither mast cells nor basophils were required in mouse models of active systemic anaphylaxis. Therefore, we investigated what antibodies, receptors, and cells are involved in active systemic anaphylaxis in mice. We found that IgG antibodies, FcγRIIIA and FcγRIV, platelet-activating factor, neutrophils, and, to a lesser extent, basophils were involved. Neutrophil activation could be monitored in vivo during anaphylaxis. Neutrophil depletion inhibited active, and also passive, systemic anaphylaxis. Importantly, mouse and human neutrophils each restored anaphylaxis in anaphylaxis-resistant mice, demonstrating that neutrophils are sufficient to induce anaphylaxis in mice and suggesting that neutrophils can contribute to anaphylaxis in humans. Our results therefore reveal an unexpected role for IgG, IgG receptors, and neutrophils in anaphylaxis in mice. These molecules and cells could be potential new targets for the development of anaphylaxis therapeutics if the same mechanism is responsible for anaphylaxis in humans.


Subject(s)
Anaphylaxis/etiology , Anaphylaxis/immunology , Neutrophils/immunology , Animals , Basophils/immunology , Humans , Immunoglobulin G/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Neutrophil Activation , Platelet Activating Factor/metabolism , Receptors, IgG/deficiency , Receptors, IgG/genetics , Receptors, IgG/metabolism , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL