Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Nature ; 576(7785): E3, 2019 12.
Article in English | MEDLINE | ID: mdl-31745371

ABSTRACT

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Nature ; 574(7776): 45-56, 2019 10.
Article in English | MEDLINE | ID: mdl-31578484

ABSTRACT

New therapies that promote antitumour immunity have been recently developed. Most of these immunomodulatory approaches have focused on enhancing T-cell responses, either by targeting inhibitory pathways with immune checkpoint inhibitors, or by targeting activating pathways, as with chimeric antigen receptor T cells or bispecific antibodies. Although these therapies have led to unprecedented successes, only a minority of patients with cancer benefit from these treatments, highlighting the need to identify new cells and molecules that could be exploited in the next generation of immunotherapy. Given the crucial role of innate immune responses in immunity, harnessing these responses opens up new possibilities for long-lasting, multilayered tumour control.


Subject(s)
Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunotherapy/methods , Immunotherapy/trends , Neoplasms/immunology , Neoplasms/therapy , Animals , Humans , Neoplasms/drug therapy , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
4.
J Allergy Clin Immunol ; 142(5): 1548-1557, 2018 11.
Article in English | MEDLINE | ID: mdl-29366701

ABSTRACT

BACKGROUND: Basophil activation contributes to inflammatory reactions, especially in allergy. It is controlled, both positively and negatively, by several mechanisms. High-affinity IgE receptors (FcεRI) generate a mixture of activation and inhibition signals when aggregated, the ratio of which depends on the concentration of allergen recognized by receptor-bound IgE. Low-affinity IgG receptors (FcγRIIA/B) generate inhibition signals when coengaged with FcεRI by allergen-antibody immune complexes. Commensal and probiotic bacteria, such as Lactobacillus paracasei, generate inhibition signals through still unclear mechanisms. OBJECTIVE: We sought to investigate whether mechanisms that control, both positively and negatively, basophil activation, which were unraveled and studied in basophils from healthy donors, are functional in allergic patients. METHODS: FcεRI and FcγRIIA/B expression, FcεRI-dependent activation, FcεRI-dependent inhibition, and FcγRIIB-dependent inhibition were examined in blood basophils incubated overnight with or without L paracasei and challenged under 10 experimental conditions. Basophils from healthy donors were compared with basophils from patients who consulted an allergology outpatient clinic over a period of 3 months with respiratory allergy, anaphylaxis antecedents, chronic urticaria, and/or atopic dermatitis. RESULTS: Patients' basophils expressed neither more FcεRI nor less FcγRIIB than basophils from healthy donors. They were neither hyperreactive to positive regulation nor hyporeactive to negative regulation, irrespective of the receptors or mechanisms involved and the allergic manifestations of the patients. CONCLUSION: Regulatory mechanisms that control basophil activation are fully functional in allergic patients. Intrinsic defects in these mechanisms do not explain allergic manifestations. Based on these mechanisms, immune checkpoint modifiers can be developed as novel therapeutic tools for allergy.


Subject(s)
Basophils/immunology , Hypersensitivity/immunology , Adolescent , Adult , Aged , Female , Humans , Lacticaseibacillus paracasei/immunology , Male , Middle Aged , Receptors, IgE/immunology , Receptors, IgG/immunology , Young Adult
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.
Curr Top Microbiol Immunol ; 382: 131-64, 2014.
Article in English | MEDLINE | ID: mdl-25116099

ABSTRACT

Most biological activities of antibodies depend on their ability to engage Receptors for the Fc portion of immunoglobulins (FcRs) on a variety of cell types. As FcRs can trigger positive and negative signals, as these signals control several biological activities in individual cells, as FcRs are expressed by many cells of hematopoietic origin, mostly of the myeloid lineage, as these cells express various combinations of FcRs, and as FcR-expressing cells have different functional repertoires, antibodies can exert a wide spectrum of biological activities. Like B and T Cell Receptors (BCRs and TCRs), FcRs are bona fide immunoreceptors. Unlike BCRs and TCRs, however, FcRs are immunoreceptors with an adaptive specificity for antigen, with an adaptive affinity for antibodies, with an adaptive structure and with an adaptive signaling. They induce adaptive biological responses that depend on their tissue distribution and on FcR-expressing cells that are selected locally by antibodies. They critically determine health and disease. They are thus exquisitely adaptive therapeutic tools.


Subject(s)
Adaptive Immunity , Receptors, Fc/physiology , Animals , Antibody Affinity , Autoimmunity , Humans , Hypersensitivity/immunology , Infections/immunology , Signal Transduction
7.
Curr Top Microbiol Immunol ; 382: 69-93, 2014.
Article in English | MEDLINE | ID: mdl-25116096

ABSTRACT

A global and rigorous understanding of the signaling pathways and cross-regulatory processes involved in mast cell activation requires the integration of published information with novel functional datasets into a comprehensive computational model. Based on an exhaustive curation of the existing literature and using the software CellDesigner, we have built and annotated a comprehensive molecular map for the FcεRI signaling network. This map can be used to visualize and interpret high-throughput expression data. Furthermore, leaning on this map and using the logical modeling software GINsim, we have derived a qualitative dynamical model, which recapitulates the most salient features of mast cell activation. The resulting logical model can be used to explore the dynamical properties of the system and its responses to different stimuli, in normal or mutant conditions.


Subject(s)
Computer Simulation , Mast Cells/physiology , Signal Transduction/physiology , Animals , Humans , Receptors, Fc/physiology , Software
8.
J Immunol ; 190(1): 106-14, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23209322

ABSTRACT

Maintenance of plasma IgM levels is critical for immune system function and homeostasis in humans and mice. However, the mechanisms that control homeostasis of the activated IgM-secreting B cells are unknown. After adoptive transfer into immune-deficient hosts, B lymphocytes expand poorly, but fully reconstitute the pool of natural IgM-secreting cells and circulating IgM levels. By using sequential cell transfers and B cell populations from several mutant mice, we were able to identify novel mechanisms regulating the size of the IgM-secreting B cell pool. Contrary to previous mechanisms described regulating homeostasis, which involve competition for the same niche by cells having overlapping survival requirements, homeostasis of the innate IgM-secreting B cell pool is also achieved when B cell populations are able to monitor the number of activated B cells by detecting their secreted products. Notably, B cell populations are able to assess the density of activated B cells by sensing their secreted IgG. This process involves the FcγRIIB, a low-affinity IgG receptor that is expressed on B cells and acts as a negative regulator of B cell activation, and its intracellular effector the inositol phosphatase SHIP. As a result of the engagement of this inhibitory pathway, the number of activated IgM-secreting B cells is kept under control. We hypothesize that malfunction of this quorum-sensing mechanism may lead to uncontrolled B cell activation and autoimmunity.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Homeostasis/immunology , Immunoglobulin M/metabolism , Lymphocyte Activation/immunology , Quorum Sensing/immunology , Adoptive Transfer , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , B-Lymphocyte Subsets/transplantation , Cell Differentiation/genetics , Cell Differentiation/immunology , Homeostasis/genetics , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Quorum Sensing/genetics
9.
Mol Cell Proteomics ; 12(10): 2874-89, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23820730

ABSTRACT

We report the first proteomic analysis of the SLP76 interactome in resting and activated primary mouse mast cells. This was made possible by a novel genetic approach used for the first time here. It consists in generating knock-in mice that express signaling molecules bearing a C-terminal tag that has a high affinity for a streptavidin analog. Tagged molecules can be used as molecular baits to affinity-purify the molecular complex in which they are engaged, which can then be studied by mass spectrometry. We examined first SLP76 because, although this cytosolic adapter is critical for both T cell and mast cell activation, its role is well known in T cells but not in mast cells. Tagged SLP76 was expressed in physiological amounts and fully functional in mast cells. We unexpectedly found that SLP76 is exquisitely sensitive to mast cell granular proteases, that Zn(2+)-dependent metalloproteases are especially abundant in mast cells and that they were responsible for SLP76 degradation. Adding a Zn(2+) chelator fully protected SLP76 in mast cell lysates, thereby enabling an efficient affinity-purification of this adapter with its partners. Label-free quantitative mass spectrometry analysis of affinity-purified SLP76 interactomes uncovered both partners already described in T cells and novel partners seen in mast cells only. Noticeably, molecules inducibly recruited in both cell types primarily concur to activation signals, whereas molecules recruited in activated mast cells only are mostly associated with inhibition signals. The transmembrane adapter LAT2, and the serine/threonine kinase with an exchange factor activity Bcr were the most recruited molecules. Biochemical and functional validations established the unexpected finding that Bcr is recruited by SLP76 and positively regulates antigen-induced mast cell activation. Knock-in mice expressing tagged molecules with a normal tissue distribution and expression therefore provide potent novel tools to investigate signalosomes and to uncover novel signaling molecules in mast cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Mast Cells/metabolism , Phosphoproteins/metabolism , Receptors, IgE/metabolism , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Interaction Maps , Proteomics , Proto-Oncogene Proteins c-bcr/genetics , Proto-Oncogene Proteins c-bcr/metabolism
10.
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
11.
Virol J ; 11: 82, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24885320

ABSTRACT

BACKGROUND: Public health risks associated to infection by human coronaviruses remain considerable and vaccination is a key option for preventing the resurgence of severe acute respiratory syndrome coronavirus (SARS-CoV). We have previously reported that antibodies elicited by a SARS-CoV vaccine candidate based on recombinant, full-length SARS-CoV Spike-protein trimers, trigger infection of immune cell lines. These observations prompted us to investigate the molecular mechanisms and responses to antibody-mediated infection in human macrophages. METHODS: We have used primary human immune cells to evaluate their susceptibility to infection by SARS-CoV in the presence of anti-Spike antibodies. Fluorescence microscopy and real-time quantitative reverse transcriptase polymerase chain reaction (RT-PCR) were utilized to assess occurrence and consequences of infection. To gain insight into the underlying molecular mechanism, we performed mutational analysis with a series of truncated and chimeric constructs of fragment crystallizable γ receptors (FcγR), which bind antibody-coated pathogens. RESULTS: We show here that anti-Spike immune serum increased infection of human monocyte-derived macrophages by replication-competent SARS-CoV as well as Spike-pseudotyped lentiviral particles (SARS-CoVpp). Macrophages infected with SARS-CoV, however, did not support productive replication of the virus. Purified anti-viral IgGs, but not other soluble factor(s) from heat-inactivated mouse immune serum, were sufficient to enhance infection. Antibody-mediated infection was dependent on signaling-competent members of the human FcγRII family, which were shown to confer susceptibility to otherwise naïve ST486 cells, as binding of immune complexes to cell surface FcγRII was necessary but not sufficient to trigger antibody-dependent enhancement (ADE) of infection. Furthermore, only FcγRII with intact cytoplasmic signaling domains were competent to sustain ADE of SARS-CoVpp infection, thus providing additional information on the role of downstream signaling by FcγRII. CONCLUSIONS: These results demonstrate that human macrophages can be infected by SARS-CoV as a result of IgG-mediated ADE and indicate that this infection route requires signaling pathways activated downstream of binding to FcγRII receptors.


Subject(s)
Antibodies, Viral/immunology , Endocytosis , Macrophages/virology , Severe acute respiratory syndrome-related coronavirus/physiology , Cells, Cultured , Humans , Microscopy, Fluorescence , Real-Time Polymerase Chain Reaction
12.
J Immunol ; 189(6): 2995-3006, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22908332

ABSTRACT

Besides high-affinity IgE receptors (FcεRI), human basophils express activating (FcγRIIA) and inhibitory (FcγRIIB) low-affinity IgG receptors. IgG receptors (FcγR) were also found on mouse basophils, but not identified. We investigated in this study FcγR and the biological consequences of their engagement in basophils of the two species. We found the following: (1) that mouse basophils also express activating (FcγRIIIA) and inhibitory (FcγRIIB) low-affinity FcγR; (2) that activating FcγR can activate both human and mouse basophils, albeit with different efficacies; (3) that negative signals triggered by inhibitory FcγR are dominant over positive signals triggered by activating FcγR, thus preventing both human and mouse basophils from being activated by IgG immune complexes; (4) that the coengagement of FcεRI with inhibitory and activating FcγR results in a FcγRIIB-dependent inhibition of IgE-induced responses of both human and mouse basophils; (5) that FcγRIIB has a similar dominant inhibitory effect in basophils from virtually all normal donors; and (6) that IL-3 upregulates the expression of both activating and inhibitory FcγR on human basophils from normal donors, but further enhances FcγRIIB-dependent inhibition. FcγR therefore function as a regulatory module, made of two subunits with antagonistic properties, that prevents IgG-induced and controls IgE-induced basophil activation in both mice and humans.


Subject(s)
Basophils/immunology , Basophils/metabolism , Down-Regulation/immunology , Receptors, IgG/physiology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , GPI-Linked Proteins/physiology , Humans , Immunoglobulin E/biosynthesis , Immunoglobulin E/blood , Immunoglobulin G/blood , Immunoglobulin G/physiology , Mice , Mice, Transgenic , Receptors, IgG/biosynthesis , Receptors, IgG/blood
13.
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
14.
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
15.
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
16.
J Immunol ; 186(6): 3289-93, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21317388

ABSTRACT

Despite their increasing use in autoimmune, inflammatory, and allergic conditions, the mechanism of action of i.v. Igs (IVIg) is poorly understood. On the basis of the critical role of invariant NKT (iNKT) cells in allergic airway inflammation (AAI) and their constitutive expression of the low-affinity IgG receptor FcγRIIIA, we surmised that IVIg targets iNKT cells to exert their anti-inflammatory effect. We found that IVIg treatment significantly inhibited AAI in OVA-sensitized C57BL/6 mice and downregulated α-galactosylceramide-induced iNKT cell activation and cytokine production. Allergic responses were restored in iNKT cell-deficient mice by transferring iNKT cells from PBS- but not from IVIg-treated mice, suggesting that IVIg acts directly on activated iNKT cells that have a critical role in AAI. The inhibitory effects of IVIg on both iNKT cell activation/function and OVA-driven AAI were lost in FcγRIIIA(-/-) mice. Our data unravel an FcγRIIIA-dependent inhibitory effect of IVIg on activated iNKT cells that confers protection in AAI.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Immunoglobulins, Intravenous/physiology , Inflammation Mediators/physiology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/pathology , Receptors, IgG/physiology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/prevention & control , Adoptive Transfer , Allergens/immunology , Allergens/toxicity , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Bronchial Hyperreactivity/therapy , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Galactosylceramides/antagonists & inhibitors , Galactosylceramides/pharmacology , Immunoglobulins, Intravenous/therapeutic use , Inflammation Mediators/therapeutic use , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/metabolism , Ovalbumin/immunology , Ovalbumin/toxicity , Receptors, IgG/therapeutic use , Respiratory Hypersensitivity/pathology , Spleen/immunology , Spleen/pathology , Spleen/transplantation
17.
J Virol ; 85(20): 10582-97, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21775467

ABSTRACT

Public health measures successfully contained outbreaks of the severe acute respiratory syndrome coronavirus (SARS-CoV) infection. However, the precursor of the SARS-CoV remains in its natural bat reservoir, and reemergence of a human-adapted SARS-like coronavirus remains a plausible public health concern. Vaccination is a major strategy for containing resurgence of SARS in humans, and a number of vaccine candidates have been tested in experimental animal models. We previously reported that antibody elicited by a SARS-CoV vaccine candidate based on recombinant full-length Spike-protein trimers potentiated infection of human B cell lines despite eliciting in vivo a neutralizing and protective immune response in rodents. These observations prompted us to investigate the mechanisms underlying antibody-dependent enhancement (ADE) of SARS-CoV infection in vitro. We demonstrate here that anti-Spike immune serum, while inhibiting viral entry in a permissive cell line, potentiated infection of immune cells by SARS-CoV Spike-pseudotyped lentiviral particles, as well as replication-competent SARS coronavirus. Antibody-mediated infection was dependent on Fcγ receptor II but did not use the endosomal/lysosomal pathway utilized by angiotensin I converting enzyme 2 (ACE2), the accepted receptor for SARS-CoV. This suggests that ADE of SARS-CoV utilizes a novel cell entry mechanism into immune cells. Different SARS vaccine candidates elicit sera that differ in their capacity to induce ADE in immune cells despite their comparable potency to neutralize infection in ACE2-bearing cells. Our results suggest a novel mechanism by which SARS-CoV can enter target cells and illustrate the potential pitfalls associated with immunization against it. These findings should prompt further investigations into SARS pathogenesis.


Subject(s)
Antibodies, Viral/metabolism , Antibody-Dependent Enhancement , Lymphocytes/virology , Membrane Glycoproteins/metabolism , Receptors, IgG/metabolism , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Viral Envelope Proteins/metabolism , Virus Internalization , Animals , Antibodies, Neutralizing/metabolism , Cells, Cultured , Chlorocebus aethiops , Cysteine Proteases , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Severe acute respiratory syndrome-related coronavirus/immunology , Spike Glycoprotein, Coronavirus
18.
Immunol Rev ; 224: 11-43, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18759918

ABSTRACT

Since an immunoreceptor tyrosine-based inhibition motif (ITIM) was first identified in the intracytoplasmic domain of Fc gammaRIIB, ITIMs have been found in a large number of inhibitory molecules that were shown to negatively regulate cell activation. Due to their wide tissue distribution and to the variety of their extracellular ligands, ITIM-containing molecules are involved in the control of a large spectrum of biological functions, mostly but not exclusively related to immunity. On the basis of sequence comparison, ITIMs were structurally defined as 6-amino acid sequences containing a tyrosine (Y) with loosely conserved N-terminal (Y-2) and C-terminal (Y+3) residues. Molecular analysis of signaling events demonstrated that when coaggregated with activating receptors, ITIMs are phosphorylated by Src-family tyrosine kinases, which enables them to recruit Src homology 2 domain-containing phosphatases that antagonize activation signals. Because ITIM-dependent negative regulation seems to be a fundamental regulatory mechanism, both in rodents and in humans, and because it can be used either as a target or as a powerful tool in various diseases, we undertook (i) a genome-wide search of potential novel ITIM-containing molecules in humans, mice, frogs, birds, and flies and (ii) a comparative analysis of potential ITIMs in major animal phyla, from mammals to protozoa. We found a surprisingly high number of potential ITIM-containing molecules, having a great diversity of extracellular domains, and being expressed by a variety of immune and non-immune cells. ITIMs could be traced back to the most primitive metazoa. The genes that encode ITIM-containing molecules that belong to the immunoglobulin superfamily or to the C-lectin family seem to derive from a common set of ancestor genes and to have dramatically expanded and diverged in Gnathostomata (from fish to mammals).


Subject(s)
Amino Acid Motifs/immunology , Receptors, Immunologic/immunology , Signal Transduction/immunology , src Homology Domains/immunology , src-Family Kinases/immunology , Animals , Evolution, Molecular , Feedback, Physiological , Humans , Receptors, IgG/genetics , Receptors, IgG/immunology , Receptors, IgG/metabolism , Receptors, Immunologic/genetics
19.
Front Immunol ; 13: 984678, 2022.
Article in English | MEDLINE | ID: mdl-36177051

ABSTRACT

Progress in neuroimmunology established that the nervous and the immune systems are two functionally related physiological systems. Unique sensory and immune receptors enable them to control interactions of the organism with the inner and the outer worlds. Both systems undergo an experience-driven selection process during their ontogeny. They share the same mediators/neurotransmitters and use synapses for intercellular communication. They keep a memory of previous experiences. Immune cells can affect nervous cells, nervous cells can affect immune cells, and they regulate each other. I however argue that the two systems differ by three major points: 1) Unlike the nervous system, the immune system has a loose anatomical structure, in which molecular and cellular events mostly occur at random; 2) The immune system can respond to molecules of the living world whereas the nervous system can respond to phenomena of the physical world; 3) Responses of the immune system act both on the organism and on the stimulus that triggered the response, whereas responses of the nervous system act on the organism only. The nervous and the immune systems therefore appear as two complementary systems of relations that closely work together, and whose reactivities are well-suited to deal with physical and biological stimuli, respectively. Its ability both to adapt the organism to the living world and to adapt the living world to the organism endows the immune system with powerful adaptive properties that enable the organism to live in peace with itself and with other living beings, whether pathogens or commensals.


Subject(s)
Immune System , Neuroimmunomodulation , Cell Communication , Immune System/physiology , Nervous System , Neuroimmunomodulation/physiology , Neurotransmitter Agents/physiology
20.
J Exp Med ; 202(12): 1635-41, 2005 Dec 19.
Article in English | MEDLINE | ID: mdl-16352739

ABSTRACT

Mastocytosis is a rare neoplastic disease characterized by a pathologic accumulation of tissue mast cells (MCs). Mastocytosis is often associated with a somatic point mutation in the Kit protooncogene leading to an Asp/Val substitution at position 816 in the kinase domain of this receptor. The contribution of this mutation to mastocytosis development remains unclear. In addition, the clinical heterogeneity presented by mastocytosis patients carrying the same mutation is unexplained. We report that a disease with striking similarities to human mastocytosis develops spontaneously in transgenic mice expressing the human Asp816Val mutant Kit protooncogene specifically in MCs. This disease is characterized by clinical signs ranging from a localized and indolent MC hyperplasia to an invasive MC tumor. In addition, bone marrow-derived MCs from transgenic animals can be maintained in culture for >24 mo and acquire growth factor independency for proliferation. These results demonstrate a causal link in vivo between the Asp816Val Kit mutation and MC neoplasia and suggest a basis for the clinical heterogeneity of human mastocytosis.


Subject(s)
Mast Cells/metabolism , Mastocytosis/genetics , Mutation, Missense/genetics , Proto-Oncogene Proteins c-kit/genetics , Animals , Blotting, Western , Cell Culture Techniques , DNA Primers , Humans , Mastocytosis/pathology , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL