Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Clin Transl Allergy ; 10: 32, 2020.
Article in English | MEDLINE | ID: mdl-32695309

ABSTRACT

Elevated serum IgE levels are associated with allergic disorders, parasitosis and specific immunologic abnormalities. In addition, epidemiological and mechanistic evidence indicates an association between IgE-mediated immune surveillance and protection from tumour growth. Intriguingly, recent studies reveal a correlation between IgE deficiency and increased malignancy risk. This is the first review discussing IgE levels and links to pathological conditions, with special focus on the potential clinical significance of ultra-low serum IgE levels and risk of malignancy. In this Position Paper we discuss: (a) the utility of measuring total IgE levels in the management of allergies, parasitosis, and immunodeficiencies, (b) factors that may influence serum IgE levels, (c) IgE as a marker of different disorders, and d) the relationship between ultra-low IgE levels and malignancy susceptibility. While elevated serum IgE is generally associated with allergic/atopic conditions, very low or absent IgE may hamper anti-tumour surveillance, indicating the importance of a balanced IgE-mediated immune function. Ultra-low IgE may prove to be an unexpected biomarker for cancer risk. Nevertheless, given the early stage of investigations conducted mostly in patients with diseases that influence IgE levels, in-depth mechanistic studies and stratification of malignancy risk based on associated demographic, immunological and clinical co-factors are warranted.

2.
Allergy ; 73(12): 2328-2341, 2018 12.
Article in English | MEDLINE | ID: mdl-29654623

ABSTRACT

BACKGROUND: Designing biologically informative models for assessing the safety of novel agents, especially for cancer immunotherapy, carries substantial challenges. The choice of an in vivo system for studies on IgE antibodies represents a major impediment to their clinical translation, especially with respect to class-specific immunological functions and safety. Fcε receptor expression and structure are different in humans and mice, so that the murine system is not informative when studying human IgE biology. By contrast, FcεRI expression and cellular distribution in rats mirror that of humans. METHODS: We are developing MOv18 IgE, a human chimeric antibody recognizing the tumour-associated antigen folate receptor alpha. We created an immunologically congruent surrogate rat model likely to recapitulate human IgE-FcεR interactions and engineered a surrogate rat IgE equivalent to MOv18. Employing this model, we examined in vivo safety and efficacy of antitumour IgE antibodies. RESULTS: In immunocompetent rats, rodent IgE restricted growth of syngeneic tumours in the absence of clinical, histopathological or metabolic signs associated with obvious toxicity. No physiological or immunological evidence of a "cytokine storm" or allergic response was seen, even at 50 mg/kg weekly doses. IgE treatment was associated with elevated serum concentrations of TNFα, a mediator previously linked with IgE-mediated antitumour and antiparasitic functions, alongside evidence of substantially elevated tumoural immune cell infiltration and immunological pathway activation in tumour-bearing lungs. CONCLUSION: Our findings indicate safety of MOv18 IgE, in conjunction with efficacy and immune activation, supporting the translation of this therapeutic approach to the clinical arena.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/adverse effects , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Immunoglobulin E/adverse effects , Immunoglobulin E/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Receptors, IgE/metabolism , Animals , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/metabolism , Cell Line, Tumor , Folate Receptor 1/immunology , Humans , Immunoglobulin E/administration & dosage , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Mice , Models, Animal , Neoplasms/pathology , Protein Binding , Rats , Statistics, Nonparametric , Treatment Outcome , Tumor Necrosis Factor-alpha/blood
3.
Allergy ; 73(2): 328-340, 2018 02.
Article in English | MEDLINE | ID: mdl-28921585

ABSTRACT

While desired for the cure of allergy, regulatory immune cell subsets and nonclassical Th2-biased inflammatory mediators in the tumour microenvironment can contribute to immune suppression and escape of tumours from immunological detection and clearance. A key aim in the cancer field is therefore to design interventions that can break immunological tolerance and halt cancer progression, whereas on the contrary allergen immunotherapy exactly aims to induce tolerance. In this position paper, we review insights on immune tolerance derived from allergy and from cancer inflammation, focusing on what is known about the roles of key immune cells and mediators. We propose that research in the field of AllergoOncology that aims to delineate these immunological mechanisms with juxtaposed clinical consequences in allergy and cancer may point to novel avenues for therapeutic interventions that stand to benefit both disciplines.


Subject(s)
Hypersensitivity/immunology , Hypersensitivity/therapy , Immune Tolerance/immunology , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Desensitization, Immunologic/methods , Humans
4.
Allergy ; 72(8): 1212-1221, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28052336

ABSTRACT

BACKGROUND: Ivermectin (IVM) is widely used in both human and veterinary medicine to treat parasitic infections. Recent reports have suggested that IVM could also have anti-inflammatory properties. METHODS: Here, we investigated the activity of IVM in a murine model of atopic dermatitis (AD) induced by repeated exposure to the allergen Dermatophagoides farinae, and in standard cellular immunological assays. RESULTS: Our results show that topical IVM improved allergic skin inflammation by reducing the priming and activation of allergen-specific T cells, as well as the production of inflammatory cytokines. While IVM had no major impact on the functions of dendritic cells in vivo and in vitro, IVM impaired T-cell activation, proliferation, and cytokine production following polyclonal and antigen-specific stimulation. CONCLUSION: Altogether, our results show that IVM is endowed with topical anti-inflammatory properties that could have important applications for the treatment of T-cell-mediated skin inflammatory diseases.


Subject(s)
Allergens/immunology , Dermatitis, Atopic/immunology , Ivermectin/administration & dosage , Administration, Topical , Animals , Antigens, Dermatophagoides/immunology , Cell Line , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Disease Models, Animal , Enzyme-Linked Immunospot Assay , Fragile X Mental Retardation Protein/metabolism , Humans , Inflammation Mediators/metabolism , Lymphocyte Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Receptors, Purinergic P2X4/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
5.
Allergy ; 72(6): 866-887, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28032353

ABSTRACT

Th2 immunity and allergic immune surveillance play critical roles in host responses to pathogens, parasites and allergens. Numerous studies have reported significant links between Th2 responses and cancer, including insights into the functions of IgE antibodies and associated effector cells in both antitumour immune surveillance and therapy. The interdisciplinary field of AllergoOncology was given Task Force status by the European Academy of Allergy and Clinical Immunology in 2014. Affiliated expert groups focus on the interface between allergic responses and cancer, applied to immune surveillance, immunomodulation and the functions of IgE-mediated immune responses against cancer, to derive novel insights into more effective treatments. Coincident with rapid expansion in clinical application of cancer immunotherapies, here we review the current state-of-the-art and future translational opportunities, as well as challenges in this relatively new field. Recent developments include improved understanding of Th2 antibodies, intratumoral innate allergy effector cells and mediators, IgE-mediated tumour antigen cross-presentation by dendritic cells, as well as immunotherapeutic strategies such as vaccines and recombinant antibodies, and finally, the management of allergy in daily clinical oncology. Shedding light on the crosstalk between allergic response and cancer is paving the way for new avenues of treatment.


Subject(s)
Hypersensitivity/immunology , Immunotherapy/methods , Neoplasms/immunology , Antibodies , Humans , Immunoglobulin E/immunology , Immunologic Surveillance , Immunotherapy/trends , Neoplasms/therapy , Th2 Cells/immunology
6.
Immunol Cell Biol ; 93(2): 198-212, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25385067

ABSTRACT

Interleukin-17A (IL-17A) is a pro-inflammatory cytokine that has an important role at mucosal sites in a wide range of immune responses including infection, allergy and auto-immunity. γδ T cells are recognized as IL-17 producers, but based on the level of CD3 expression, we now define the remarkable ability of a CD3(bright) γδ T-cell subset with an effector memory phenotype to rapidly produce IL-17A, but not interferon-γ. CD3(bright) γδ T cells uniformly express the canonical germline encoded Vγ6/Vδ1(+) T-cell receptor. They are widely distributed with a preferential representation in the lungs and skin are negatively impacted in the absence of retinoic acid receptor-related orphan receptor gammat expression or endogenous flora. This population responded rapidly to various stimuli in a mechanism involving IL-23 and NOD-like receptor family, pyrin domain containing 3 (NLRP3)-inflammasome-dependent IL-1ß. Finally, we demonstrated that IL-17-producing CD3(bright) γδ T cells responded promptly and strongly to pneumococcal infection and during skin inflammation. Here, we propose a new way to specifically analyze IL-17-producing Vγ6/Vδ1(+) T cells based on the level of CD3 signals. Using this gating strategy, our data reinforce the crucial role of this γδ T-cell subset in respiratory and skin disorders.


Subject(s)
CD3 Complex/metabolism , Interleukin-17/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/immunology , Amino Acid Sequence , Aminoquinolines/pharmacology , Animals , CD3 Complex/chemistry , Carrier Proteins/metabolism , Germ Cells/drug effects , Homeostasis/drug effects , Imiquimod , Immunity , Inflammasomes/drug effects , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Interleukin-23 , Lung/drug effects , Lung/immunology , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Male , Mice, Inbred C57BL , Molecular Sequence Data , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype , Skin/drug effects , Skin/immunology , T-Lymphocytes/drug effects
7.
Mucosal Immunol ; 8(4): 841-51, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25425267

ABSTRACT

Allergic asthma is a chronic inflammatory disease characterized by airway hyperresponsiveness (AHR), lung infiltration of Th2 cells, and high levels of IgE. To date, allergen-specific immunotherapy (SIT) is the only treatment that effectively alleviates clinical symptoms and has a long-term effect after termination. Unfortunately, SIT is unsuitable for plurisensitized patients, and highly immunogenic allergens cannot be used. To overcome these hurdles, we sought to induce regulatory CD4(+) T cells (Treg) specific to an exogenous antigen that could be later activated as needed in vivo to control allergic responses. We have established an experimental approach in which mice tolerized to ovalbumin (OVA) were sensitized to the Leishmania homolog of receptors for activated c kinase (LACK) antigen, and subsequently challenged with aerosols of LACK alone or LACK and OVA together. Upon OVA administration, AHR and allergic airway responses were strongly reduced. OVA-induced suppression was mediated by CD25(+) Treg, required CTLA-4 and ICOS signaling and resulted in decreased numbers of migrating airway dendritic cells leading to a strong impairment in the proliferation of allergen-specific Th2 cells. Therefore, inducing Treg specific to a therapeutic antigen that could be further activated in vivo may represent a safe and novel curative approach for allergic asthma.


Subject(s)
Allergens/immunology , Desensitization, Immunologic , Respiratory Hypersensitivity/immunology , Allergens/administration & dosage , Animals , Antigens, Protozoan/immunology , Asthma/immunology , Asthma/metabolism , Asthma/therapy , Bronchoalveolar Lavage Fluid/immunology , CTLA-4 Antigen/metabolism , Desensitization, Immunologic/methods , Disease Models, Animal , Immunoglobulin E/immunology , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Protozoan Proteins/immunology , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/therapy , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
8.
Mucosal Immunol ; 4(1): 53-65, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20811345

ABSTRACT

The prevalence of asthma has steadily increased during the last decade, probably as the result of changes in the environment, including reduced microbial exposure during infancy. Accordingly, experimental studies have shown that deliberate infections with live pathogens prevent the development of allergic airway diseases in mice. Bacterial extracts are currently used in children suffering from repeated upper respiratory tract infections. In the present study, we have investigated whether bacterial extracts, commercially available as Broncho-Vaxom (BV), could prevent allergic airway disease in mice. Oral treatment with BV suppressed airway inflammation through interleukin-10 (IL-10)-dependent and MyD88 (myeloid differentiation primary response gene (88))-dependent mechanisms and induced the conversion of FoxP3 (forkhead box P3)(-) T cells into FoxP3(+) regulatory T cells. Furthermore, CD4(+) T cells purified from the trachea of BV-treated mice conferred protection against airway inflammation when adoptively transferred into sensitized mice. Therefore, treatment with BV could possibly be a safe and efficient strategy to prevent the development of allergic diseases in children.


Subject(s)
Asthma , Bacteria , Respiratory System , T-Lymphocytes, Regulatory , Animals , Mice , Administration, Oral , Adoptive Transfer , Asthma/immunology , Asthma/prevention & control , Bacteria/cytology , Bacteria/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Interleukin-10/metabolism , Mice, Inbred BALB C , Myeloid Differentiation Factor 88/metabolism , Respiratory System/immunology , T-Lymphocytes, Regulatory/immunology
9.
Mucosal Immunol ; 4(3): 343-53, 2011 May.
Article in English | MEDLINE | ID: mdl-21048704

ABSTRACT

Allergic asthma is a T cell-dependent inflammatory lung disease that results from complex interactions between genetic predisposition and environmental factors, including exposure to lipopolysaccharide (LPS). In this study, we have shown that airway LPS exposure was sufficient to induce airway hyperreactivity (AHR) and eosinophil recruitment in mice that had previously experienced an acute episode of allergic asthma. LPS-induced disease reactivation depended on the activation of allergen-specific CD4(+) T cells by a subset of lung langerin(+) dendritic cells (DCs) that retained the allergen. Upon LPS exposure, migration of langerin(+) DCs from lungs to draining lymph nodes increased and LPS-exposed langerin(+) DCs instructed CD4(+) T cells toward a T helper (Th) 2 response. Selective depletion of langerin(+) DCs prevented LPS-induced eosinophil recruitment and T-cell activation, further demonstrating a critical role for langerin(+) DCs in disease reactivation. This finding provides a possible explanation for the subclinical worsening of asthmatics following exposure to low-dose LPS.


Subject(s)
Asthma/immunology , Dendritic Cells/metabolism , Th2 Cells/metabolism , Allergens/immunology , Animals , Antigens, Surface/biosynthesis , Cell Movement , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/pathology , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , Humans , Lectins, C-Type/biosynthesis , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Lung/pathology , Lymphocyte Activation , Mannose-Binding Lectins/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Th2 Cells/immunology , Th2 Cells/pathology
10.
Mucosal Immunol ; 3(5): 461-74, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20485331

ABSTRACT

Allergic asthma is a chronic lung disease resulting from an inappropriate T helper (Th)-2 response to environmental antigens. Early tolerance induction is an attractive approach for primary prevention of asthma. Here, we found that breastfeeding by antigen-sensitized mothers exposed to antigen aerosols during lactation induced a robust and long-lasting antigen-specific protection from asthma. Protection was more profound and persistent than the one induced by antigen-exposed non-sensitized mothers. Milk from antigen-exposed sensitized mothers contained antigen-immunoglobulin (Ig) G immune complexes that were transferred to the newborn through the neonatal Fc receptor resulting in the induction of antigen-specific FoxP3(+) CD25(+) regulatory T cells. The induction of oral tolerance by milk immune complexes did not require the presence of transforming growth factor-beta in milk in contrast to tolerance induced by milk-borne free antigen. Furthermore, neither the presence of IgA in milk nor the expression of the inhibitory FcgammaRIIb in the newborn was required for tolerance induction. This study provides new insights on the mechanisms of tolerance induction in neonates and highlights that IgG immune complexes found in breast milk are potent inducers of oral tolerance. These observations may pave the way for the identification of key factors for primary prevention of immune-mediated diseases such as asthma.


Subject(s)
Antigen-Antibody Complex/metabolism , Asthma/immunology , Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/metabolism , Milk, Human/metabolism , Receptors, Fc/metabolism , Administration, Oral , Allergens/administration & dosage , Allergens/adverse effects , Animals , Animals, Newborn , Antigen-Antibody Complex/immunology , Asthma/chemically induced , Breast Feeding , Female , Forkhead Transcription Factors/biosynthesis , Histocompatibility Antigens Class I/genetics , Immune Tolerance , Immunity, Maternally-Acquired , Immunoglobulin G/immunology , Interleukin-2 Receptor alpha Subunit/biosynthesis , Male , Maternal Exposure , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage , Ovalbumin/immunology , Pregnancy , Receptors, Fc/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
11.
Curr Opin Immunol ; 13(6): 716-20, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11677095

ABSTRACT

Recently, known eosinophil functions have been extended considerably: previously the cells were thought to have an exclusive role in the release of cytotoxic mediators; now they are known to have roles in antigen presentation and immunoregulation through the release of cytokines. Although questionable, animal models indicate a rather beneficial role of eosinophils in parasitic infections but a detrimental one, together with other cells, in allergy.


Subject(s)
Eosinophils/immunology , Hypersensitivity, Immediate/immunology , Parasitic Diseases/immunology , Animals , Anti-Inflammatory Agents/therapeutic use , Antibodies/therapeutic use , Cell Degranulation , Cytokines/biosynthesis , Disease Models, Animal , Eosinophilia/immunology , Humans , Hypersensitivity, Immediate/therapy , Mice
12.
J Immunol ; 167(2): 995-1003, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11441108

ABSTRACT

FcepsilonRI expressed by human eosinophils is involved in IgE-mediated cytotoxicity reactions toward the parasite Schistosoma mansoni in vitro. However, because receptor expression is low on these cells, its functional role is still controversial. In this study, we have measured surface and intracellular expression of FcepsilonRI by blood eosinophils from hypereosinophilic patients and normal donors. The number of unoccupied receptors corresponded to approximately 4,500 Ab binding sites per cell, whereas 50,000 Ab binding sites per cell were detected intracellularly. Eosinophils from patients displayed significantly more unoccupied receptors than cells from normal donors. This number correlated to both serum IgE concentrations and to membrane-bound IgE. The lack of FcepsilonRI expression by mouse eosinophils has hampered further studies. To overcome this fact and experimentally confirm our findings on human eosinophils, we engineered IL-5 x hFcepsilonRIalpha double-transgenic mice, whose bone marrow, blood, spleen, and peritoneal eosinophils expressed FcepsilonRI levels similar to levels of human eosinophils, after 4 days culture with IgE in the presence of IL-5. Both human and mouse eosinophils were able to secrete IL-10 upon FcepsilonRI engagement. Thus, comparative analysis of cells from patients and from a relevant animal model allowed us to clearly demonstrate that FcepsilonRI-mediated eosinophil activation leads to IL-10 secretion. Through FcepsilonRI expression, these cells are able to contribute to both the regulation of the immune response and to its effector mechanisms.


Subject(s)
Eosinophils/immunology , Eosinophils/metabolism , Interleukin-10/metabolism , Mice, Transgenic/immunology , Receptors, IgE/biosynthesis , Receptors, IgE/genetics , Animals , Cells, Cultured , Eosinophils/parasitology , Humans , Interleukin-5/genetics , Mice , Mice, Transgenic/genetics , Receptors, IgE/metabolism , Schistosoma mansoni/immunology
13.
Int Arch Allergy Immunol ; 124(1-3): 9-15, 2001.
Article in English | MEDLINE | ID: mdl-11306914

ABSTRACT

Schistosomiasis, the second major parasitic disease in the world after malaria, affects 200 million people. Vaccine strategies represent an essential component of the control of this chronic debilitating disease where the deposition of millions of eggs in the tissues is the main cause of pathology. Research developed in our laboratory over the last 20 years has led to the identification of novel effector mechanisms, pointing for the first time to the protective role of Th2 responses and of IgE antibodies now supported by seven studies in human populations. The identification and molecular cloning of a target antigen, a glutathione S-transferase (GST), has made it possible to demonstrate its vaccine potential in several animal species (rodents, cattle, primates) and to establish consistently the capacity of vaccination to reduce female worm fecundity and egg viability through the production of neutralizing antibodies (IgA and IgG). Following promising preclinical studies, clinical trials (phase I and II) have been undertaken using Schistosoma haematobium GST, Sh28GST. High titers of neutralizing antibodies were produced (IgG3 and IgA) together with Th2 cytokines, consistently with the concepts developed from experimental models. With these results we are on the way towards a feasible approach of vaccine development against a major human parasitic disease.


Subject(s)
Protozoan Vaccines , Schistosoma haematobium/immunology , Schistosomiasis/prevention & control , Animals , Antibodies, Helminth/biosynthesis , Antigens, Helminth/therapeutic use , Clinical Trials as Topic , Cytokines/biosynthesis , Glutathione Transferase/therapeutic use , Humans , Immunity, Mucosal , Immunoglobulin A/biosynthesis , Immunoglobulin E/biosynthesis , Mice , Rats , Schistosoma haematobium/enzymology , Schistosomiasis/immunology , Th2 Cells/immunology
15.
J Exp Med ; 193(1): 25-34, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11136818

ABSTRACT

A role for immunoglobulin E and its high affinity receptor (Fc epsilon RI) in the control of bacterial pathogenicity and intestinal inflammation has been suggested, but relevant animal models are lacking. Here we compare transgenic mice expressing a humanized Fc epsilon RI (hFc epsilon RI), with a cell distribution similar to that in humans, to Fc epsilon RI-deficient animals. In hFc epsilon RI transgenic mice, levels of colonic interleukin 4 were higher, the composition of fecal flora was greatly modified, and bacterial translocation towards mesenteric lymph nodes was increased. In hFc epsilon RI transgenic mice, 2,4,6-tri-nitrobenzenesulfonic acid (TNBS)-induced colitis was also more pronounced, whereas Fc epsilon RI-deficient animals were protected from colitis, demonstrating that Fc epsilon RI can affect the onset of intestinal inflammation.


Subject(s)
Colitis/immunology , Colitis/microbiology , Receptors, IgE/metabolism , Animals , Bacteria/isolation & purification , Bacteria/pathogenicity , Base Sequence , Colitis/pathology , DNA Primers/genetics , Disease Models, Animal , Humans , Immunoglobulin E/biosynthesis , Immunoglobulin E/genetics , Interleukin-4/genetics , Interleukin-4/metabolism , Lymph Nodes/microbiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Permeability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, IgE/genetics , Trinitrobenzenesulfonic Acid/toxicity
16.
Mol Cell Biol ; 20(19): 7178-82, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10982834

ABSTRACT

Immune responses are controlled by a combination of positive and negative cellular signals. Effector cells in the immune system express inhibitory receptors that serve to limit effector cell expansion and to protect the host from autoreactivity. gp49B is a receptor of unknown function that is expressed on activated mast cells and natural killer (NK) cells and whose cytoplasmic tail endows it with inhibitory potential. To gain insight into the function of gp49B in mice, we disrupted the gp49B gene by homologous recombination. gp49B(0) mice were born at expected ratios, were healthy and fertile, and displayed normal long-term survival rates. gp49B(0) mice showed no defect in NK or mast cell development. Furthermore, NK and mast cells from the gp49B(0) mice showed activation properties in vitro similar to those of cells isolated from wild-type mice. Therefore, gp49B is not critical for the development, expansion, and maturation of mast cells and NK cells in vivo. The healthy status of gp49B(0) mice makes them suitable for testing the role of gp49B in immune responses to infectious agents.


Subject(s)
Antigens, Surface/immunology , Cytotoxicity, Immunologic/genetics , Killer Cells, Natural/immunology , Mast Cells/immunology , Membrane Glycoproteins/immunology , Receptors, Immunologic , Animals , Antigens, Surface/genetics , Bone Marrow Cells/immunology , Fertility , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Phenotype
17.
J Immunol ; 165(3): 1266-71, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10903725

ABSTRACT

Besides its crucial role in type I hypersensitivity reactions, IgE is involved in anti-parasite immunity. This role has been clearly demonstrated in both human and rat schistosomiasis, but remains controversial in the mouse. Since the cellular distribution of the high affinity IgE receptor, Fc epsilon RI, differs in humans and mice, it might explain the differences in effector function of IgE between the two species. In humans, eosinophils and macrophages induce IgE-dependent cytotoxicity toward Schistosoma mansoni larvae, which involves Fc epsilon RI in the case of eosinophils. In the present study, we have investigated the expression and function of Fc epsilon RI in rat eosinophils and macrophages. We demonstrate, by flow cytometry, fluorescence microscopy, and western blot analysis, that in rats, as in humans, a functional alpha gamma 2 trimeric Fc epsilon RI is expressed on eosinophils and macrophages. We also show that these two cell types can induce IgE-mediated, Fc epsilon RI-dependent cellular cytotoxicity toward schistosomula. These results thus provide a molecular basis for the differences observed between rat and mouse regarding IgE-mediated anti-parasite immunity.


Subject(s)
Eosinophils/metabolism , Macrophages, Peritoneal/metabolism , Receptors, IgE/biosynthesis , Animals , Antibody-Dependent Cell Cytotoxicity , Cytotoxicity Tests, Immunologic , Eosinophils/immunology , Flow Cytometry , Immunoglobulin E/physiology , Larva/immunology , Macrophages, Peritoneal/immunology , Mice , Rats , Rats, Inbred Strains , Receptors, IgE/chemistry , Receptors, IgE/physiology , Schistosoma mansoni/immunology , Tumor Cells, Cultured , Up-Regulation/immunology
20.
J Exp Med ; 190(4): 487-95, 1999 Aug 16.
Article in English | MEDLINE | ID: mdl-10449520

ABSTRACT

Eosinophils are the source of various immunoregulatory cytokines, but the membrane molecules involved in their secretion have not been clearly identified. Here we show that peripheral blood eosinophils from hypereosinophilic patients could express membrane CD86 but not CD80. The T cell costimulatory molecule CD28 is also detected on the eosinophil surface. CD28 ligation but not CD86 ligation resulted in interleukin (IL)-2 and interferon (IFN)-gamma secretion by eosinophils, whereas IL-4, IL-5, and IL-10 were not detected. In contrast to T cells requiring two signals for effective stimulation, CD28 ligation alone was sufficient for optimal eosinophil activation. Eosinophil-derived IL-2 and IFN-gamma were biologically active, as supernatants from anti-CD28-treated cells were able to induce CTLL-2 proliferation and major histocompatibility complex class II expression on the colon carcinoma cell line Colo 205, respectively. Addition of secretory immunoglobulin (Ig)A-anti-IgA complexes, which could induce the release of IL-10, very significantly inhibited both CD28-mediated IL-2 and IFN-gamma release. These results suggest that the release of type 1 (IFN-gamma and IL-2) versus type 2 cytokines by eosinophils is not only differential but also dependent on cross-regulatory signals. They confirm that through activation of costimulatory molecules, eosinophils could function as an immunoregulatory cell involved in the release of both type 1 and type 2 cytokines.


Subject(s)
Antigens, CD/isolation & purification , CD28 Antigens/isolation & purification , Eosinophils/immunology , Immunoglobulin A, Secretory/pharmacology , Interferon-gamma/metabolism , Interleukin-2/metabolism , Membrane Glycoproteins/isolation & purification , Antigen-Antibody Complex/pharmacology , B7-2 Antigen , Colonic Neoplasms/immunology , Eosinophilia , Histocompatibility Antigens Class II/biosynthesis , Humans , Lymphocyte Activation , Signal Transduction , T-Lymphocytes, Cytotoxic
SELECTION OF CITATIONS
SEARCH DETAIL
...