Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
J Immunol ; 197(7): 2583-8, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27566822

ABSTRACT

Exogenous IgE acts as an adjuvant in tumor vaccination in mice, and therefore a direct role of endogenous IgE in tumor immunosurveillance was investigated. By using genetically engineered mice, we found that IgE ablation rendered mice more susceptible to the growth of transplantable tumors. Conversely, a strengthened IgE response provided mice with partial or complete resistance to tumor growth, depending on the tumor type. By genetic crosses, we showed that IgE-mediated tumor protection was mostly lost in mice lacking FcεRI. Tumor protection was also lost after depletion of CD8(+) T cells, highlighting a cross-talk between IgE and T cell-mediated tumor immunosurveillance. Our findings provide the rationale for clinical observations that relate atopy with a lower risk for developing cancer and open new avenues for the design of immunotherapeutics relevant for clinical oncology.


Subject(s)
Immunoglobulin E/immunology , Immunologic Surveillance/immunology , Neoplasms/immunology , Receptors, IgE/immunology , Adjuvants, Immunologic , Animals , Genetic Engineering , Immunotherapy , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Receptors, IgE/deficiency
2.
J Immunol ; 190(2): 539-48, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23241885

ABSTRACT

Histamine H(4) receptor (H(4)R)-deficient mice (H(4)R(-/-)), H(4)R antagonist-treated wild-type (WT) mice, and WT mice depleted of basophils failed to develop early (EPR) or late phase (LPR) nasal responses following allergen sensitization and challenge. Basophil transfer from WT but not H(4)R(-/-) mice restored the EPR and LPR in H(4)R(-/-) mice. Following passive sensitization with OVA-specific IgE, FcεRI(-/-) recipients of WT basophils plus OVA and histamine developed an EPR and LPR. OVA-IgE passively sensitized FcεRI(-/-) recipients of H(4)R(-/-) basophils and OVA and histamine challenge failed to develop an EPR or LPR, and basophils were not detected in nasal tissue. In contrast, recipients of basophils from IL-13(-/-) and IL-4(-/-)/IL-13(-/-) mice developed an EPR but not an LPR. These results demonstrate the development of allergic rhinitis proceeded in two distinct stages: histamine release from FcεRI-activated mast cells, followed by histamine-mediated recruitment of H(4)R-expressing basophils to the nasal cavity and activation through FcεRI.


Subject(s)
Basophils/immunology , Mast Cells/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Histamine/immunology , Receptors, IgE/immunology , Rhinitis, Allergic, Perennial/immunology , Adoptive Transfer , Animals , Basophils/drug effects , Basophils/metabolism , Female , Histamine Antagonists/pharmacology , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Knockout , Models, Immunological , Nasal Mucosa/drug effects , Nasal Mucosa/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, Histamine/genetics , Receptors, Histamine H4 , Receptors, IgE/deficiency , Receptors, IgE/genetics , Rhinitis, Allergic , Rhinitis, Allergic, Perennial/genetics , Rhinitis, Allergic, Perennial/therapy
3.
J Immunol ; 187(1): 164-71, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21622859

ABSTRACT

The IgE-mediated and Th2-dependent late-phase reaction remains a mechanistically enigmatic and daunting element of human allergic inflammation. In this study, we uncover the FcεRI on dendritic cells (DCs) as a key in vivo component of this form of allergy. Because rodent, unlike human, DCs lack FcεRI, this mechanism could be revealed only by using a new transgenic mouse model with human-like FcεRI expression on DCs. In the presence of IgE and allergen, FcεRI(+) DCs instructed naive T cells to differentiate into Th2 cells in vitro and boosted allergen-specific Th2 responses and Th2-dependent eosinophilia at the site of allergen exposure in vivo. Thus, FcεRI on DCs drives the cascade of pathogenic reactions linking the initial allergen capture by IgE with subsequent Th2-dominated T cell responses and the development of late-phase allergic tissue inflammation.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/pathology , Inflammation Mediators/metabolism , Receptors, IgE/metabolism , Th2 Cells/immunology , Th2 Cells/pathology , Allergens/toxicity , Animals , Antigens, Plant/toxicity , Cells, Cultured , Coculture Techniques , Dendritic Cells/metabolism , Female , Humans , Inflammation Mediators/physiology , Inflammation Mediators/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/toxicity , Protein Binding/genetics , Protein Binding/immunology , Receptors, IgE/deficiency , Receptors, IgE/physiology , Th2 Cells/metabolism , Time Factors
4.
J Exp Med ; 203(4): 1105-16, 2006 Apr 17.
Article in English | MEDLINE | ID: mdl-16606668

ABSTRACT

Type 2 immunity, which involves coordinated regulation of innate and adaptive immune responses, can protect against helminth parasite infection, but may lead to allergy and asthma after inappropriate activation. We demonstrate that il25(-/-) mice display inefficient Nippostrongylus brasiliensis expulsion and delayed cytokine production by T helper 2 cells. We further establish a key role for interleukin (IL)-25 in regulating a novel population of IL-4-, IL-5-, IL-13-producing non-B/non-T (NBNT), c-kit+, FcepsilonR1- cells during helminth infection. A deficit in this population in il25(-/-) mice correlates with inefficient N. brasiliensis expulsion. In contrast, administration of recombinant IL-25 in vivo induces the appearance of NBNT, c-kit+, FcepsilonR1- cells and leads to rapid worm expulsion that is T and B cell independent, but type 2 cytokine dependent. We demonstrate that these IL-25-regulated cells appear rapidly in the draining lymph nodes, implicating them as a source of type 2 cytokines during initiation of worm expulsion.


Subject(s)
B-Lymphocytes/cytology , Basophils/metabolism , Interleukins/physiology , Nippostrongylus/immunology , Strongylida Infections/immunology , Strongylida Infections/parasitology , T-Lymphocytes/cytology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Cytokines/biosynthesis , Cytokines/classification , Interleukin-13/biosynthesis , Interleukin-13/deficiency , Interleukin-13/genetics , Interleukin-4/biosynthesis , Interleukin-4/deficiency , Interleukin-4/genetics , Interleukin-5/biosynthesis , Interleukin-5/deficiency , Interleukin-5/genetics , Interleukins/administration & dosage , Interleukins/deficiency , Interleukins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Proto-Oncogene Proteins c-kit/metabolism , Receptors, IgE/deficiency , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
5.
J Immunol ; 182(12): 8110-7, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494337

ABSTRACT

IgE plays a pivotal role in allergic reactions and asthma through its ability to bind to the mast cell FcR for IgE (FcepsilonRI). Current therapies to suppress such reactions include passive treatment with neutralizing Abs to IgE that block its binding to FcepsilonRI. In theory, induction of immune tolerance in the B lymphocytes that carry IgE Ag receptors and give rise to IgE-secreting cells should provide longer term efficacy. However, recent data have suggested that such memory cells may lack cell surface IgE. Using a gene therapy approach, we show that a recombinant single-chain neutralizing anti-IgE could not only neutralize circulating IgE, but also reduce IgE(+) B cell numbers and H chain transcripts. Therapeutic anti-IgE stimulated a calcium response in primary B cells or in a B cell line expressing membrane IgE and suppressed IgE secretion in vitro, suggesting that active signaling through membrane IgE likely promoted tolerance. Interestingly, upon subsequent challenge of anti-IgE-treated mice with an IgE cross-linking reagent capable of inducing activation of IgE-decorated mast cells, an anaphylaxis reaction was induced, apparently via a FcgammaRIII pathway involving recognition of anti-IgE Ab itself. These studies have important implications for the optimal design of safe and effective anti-IgE therapies and suggest that the IgE memory B cells may be targeted by such genetic Ab therapies.


Subject(s)
B-Lymphocytes/immunology , Genetic Therapy , Immunoglobulin E/immunology , Receptors, IgE/immunology , Anaphylaxis/immunology , Animals , B-Lymphocytes/metabolism , Cell Line , Humans , Immunization , Immunoglobulin E/genetics , Immunoglobulin E/metabolism , Mice , Mice, Inbred BALB C , Protein Binding/immunology , Receptors, IgE/deficiency , Receptors, IgE/genetics , Receptors, IgE/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
6.
J Immunol ; 183(7): 4530-6, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19748979

ABSTRACT

Working with C57BL/6 mouse tumor models, we had previously demonstrated that vaccination with IgE-coated tumor cells can protect against tumor challenge, an observation that supports the involvement of IgE in antitumor immunity. The adjuvant effect of IgE was shown to result from eosinophil-dependent priming of the T cell-mediated adaptive immune response. The protective effect is likely to be mediated by the interaction of tumor cell-bound IgE with receptors, which then trigger the release of mediators, recruitment of effector cells, cell killing and tumor Ag cross-priming. It was therefore of utmost importance to demonstrate the strict dependence of the protective effect on IgE receptor activation. First, the protective effect of IgE was confirmed in a BALB/c tumor model, in which IgE-loaded modified VV Ankara-infected tumor cells proved to be an effective cellular vaccine. However, the protective effect was lost in Fc(epsilon)RIalpha(-/-) (but not in CD23(-/-)) knockout mice, showing the IgE-Fc(epsilo)nRI interaction to be essential. Moreover, human IgE (not effective in BALB/c mice) had a protective effect in the humanized knockin mouse (Fc(epsilon)RIalpha(-/-) hFc(epsilon)RIalpha(+)). This finding suggests that the adjuvant effect of IgE could be exploited for human therapeutics.


Subject(s)
Adjuvants, Immunologic/physiology , Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents/therapeutic use , Immunoglobulin E/physiology , Immunoglobulin E/therapeutic use , Receptors, IgE/physiology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Adjuvants, Immunologic/metabolism , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents/administration & dosage , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Cross-Linking Reagents/administration & dosage , Cross-Linking Reagents/metabolism , Cross-Linking Reagents/therapeutic use , Female , Gene Knock-In Techniques , Humans , Immunoglobulin E/metabolism , Leukemia, Basophilic, Acute/immunology , Leukemia, Basophilic, Acute/pathology , Leukemia, Basophilic, Acute/therapy , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell/therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Rats , Receptors, IgE/deficiency , Receptors, IgE/genetics
7.
Cell Death Dis ; 9(5): 510, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29724998

ABSTRACT

Binding of allergen-specific IgE to its primary receptor FcεRI on basophils and mast cells represents a central event in the development of allergic diseases. The high-affinity interaction between IgE and FcεRI results in permanent sensitization of these allergic effector cells and critically regulates their release of pro-inflammatory mediators upon IgE cross-linking by allergens. In addition, binding of monomeric IgE has been reported to actively regulate FcεRI surface levels and promote survival of mast cells in the absence of allergen through the induction of autocrine cytokine secretion including interleukin-3 (IL-3). As basophils and mast cells share many biological commonalities we sought to assess the role of monomeric IgE binding and IL-3 signaling in FcεRI regulation and cell survival of primary human basophils. FcεRI cell surface levels and survival of isolated blood basophils were assessed upon addition of monomeric IgE or physiologic removal of endogenous cell-bound IgE with a disruptive IgE inhibitor by flow cytometry. We further determined basophil cell numbers in both low and high serum IgE blood donors and mice that are either sufficient or deficient for FcεRI. Ultimately, we investigated the effect of IL-3 on basophil surface FcεRI levels by protein and gene expression analysis. Surface levels of FcεRI were passively stabilized but not actively upregulated in the presence of monomeric IgE. In contrast to previous observations with mast cells, monomeric IgE binding did not enhance basophil survival. Interestingly, we found that IL-3 transcriptionally regulates surface levels of FcεRI in human primary basophils. Our data suggest that IL-3 but not monomeric IgE regulates FcεRI expression and cell survival in primary human basophils. Thus, blocking of IL-3 signaling in allergic effector cells might represent an interesting approach to diminish surface FcεRI levels and to prevent prolonged cell survival in allergic inflammation.


Subject(s)
Basophils/immunology , Hypersensitivity/genetics , Immunoglobulin E/genetics , Interleukin-3/genetics , Receptors, IgE/genetics , Animals , Basophils/drug effects , Basophils/pathology , Cell Survival/drug effects , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Hypersensitivity/immunology , Hypersensitivity/physiopathology , Immunoglobulin E/immunology , Interleukin-3/immunology , Interleukin-3/pharmacology , Interleukin-5/genetics , Interleukin-5/immunology , Interleukin-5/pharmacology , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture , Receptors, IgE/deficiency , Receptors, IgE/immunology , Signal Transduction , Transcription, Genetic
8.
Endocrinology ; 156(11): 4047-58, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26295369

ABSTRACT

Prior studies demonstrated increased plasma IgE in diabetic patients, but the direct participation of IgE in diabetes or obesity remains unknown. This study found that plasma IgE levels correlated inversely with body weight, body mass index, and body fat mass among a population of randomly selected obese women. IgE receptor FcϵR1-deficient (Fcer1a(-/-)) mice and diet-induced obesity (DIO) mice demonstrated that FcϵR1 deficiency in DIO mice increased food intake, reduced energy expenditure, and increased body weight gain but improved glucose tolerance and glucose-induced insulin secretion. White adipose tissue from Fcer1a(-/-) mice showed an increased expression of phospho-AKT, CCAAT/enhancer binding protein-α, peroxisome proliferator-activated receptor-γ, glucose transporter-4 (Glut4), and B-cell lymphoma 2 (Bcl2) but reduced uncoupling protein 1 (UCP1) and phosphorylated c-Jun N-terminal kinase (JNK) expression, tissue macrophage accumulation, and apoptosis, suggesting that IgE reduces adipogenesis and glucose uptake but induces energy expenditure, adipocyte apoptosis, and white adipose tissue inflammation. In 3T3-L1 cells, IgE inhibited the expression of CCAAT/enhancer binding protein-α and peroxisome proliferator-activated receptor-γ, and preadipocyte adipogenesis and induced adipocyte apoptosis. IgE reduced the 3T3-L1 cell expression of Glut4, phospho-AKT, and glucose uptake, which concurred with improved glucose tolerance in Fcer1a(-/-) mice. This study established two novel pathways of IgE in reducing body weight gain in DIO mice by suppressing adipogenesis and inducing adipocyte apoptosis while worsening glucose tolerance by reducing Glut4 expression, glucose uptake, and insulin secretion.


Subject(s)
Energy Metabolism/genetics , Obesity/genetics , Receptors, IgE/genetics , Weight Gain/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Diet, High-Fat/adverse effects , Female , Gene Expression , Glucose Tolerance Test , Humans , Immunoblotting , Immunoglobulin E/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/etiology , Obesity/metabolism , Obesity, Morbid/blood , PPAR gamma/genetics , PPAR gamma/metabolism , RNA Interference , Receptors, IgE/deficiency , Reverse Transcriptase Polymerase Chain Reaction
9.
Mol Immunol ; 53(3): 270-3, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22964482

ABSTRACT

The high-affinity IgE receptor (FcɛRI) is formed by the IgE-binding α subunit, ß subunit and γ subunits homodimer. All three subunits are required for proper expression of the receptor on the plasma membrane of mast cells and basophils. However, the exact molecular mechanism of inter-subunit interactions required for correct expression and function of the FcɛRI complex remains to be identified. A recent study suggested that polar aspartate at position 194 within the transmembrane domain of the α subunit could interact by hydrogen bonding with polar threonine at position 22 in the transmembrane domains of the γ subunits. To verify this, we used previously isolated rat basophilic leukemia (RBL)-2H3 variant cells deficient in the expression of the FcɛRI-γ subunit (FcR-γ), and transfected them with DNA vectors coding for FcR-γ of the wild-type or mutants in which T22 was substituted for nonpolar alanine (T22A mutant) or polar serine (T22S mutant). Analysis of the transfectants showed that both T22A and T22S mutants were capable to restore surface expression of the FcɛRI similar to wild-type FcR-γ. Furthermore, cells transfected with wild-type, T22A or T22S FcR-γ showed comparably enhanced FcɛRI-mediated degranulation. Our data indicate that substitution of FcR-γ T22 with non-polar amino acid does not interfere with surface expression of the FcɛRI and its signaling capacity.


Subject(s)
Basophils/immunology , Receptors, IgE/chemistry , Receptors, IgE/metabolism , Alanine/chemistry , Amino Acid Substitution , Animals , Cell Degranulation/immunology , Cell Line, Tumor , Hydrogen Bonding , Leukemia, Basophilic, Acute/genetics , Leukemia, Basophilic, Acute/immunology , Leukemia, Basophilic, Acute/metabolism , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Interaction Domains and Motifs , Protein Subunits , Rats , Receptors, IgE/deficiency , Receptors, IgE/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Signal Transduction , Threonine/chemistry , Transfection
10.
PLoS One ; 6(7): e21760, 2011.
Article in English | MEDLINE | ID: mdl-21765910

ABSTRACT

IgE antibodies, administered to mice together with their specific antigen, enhance antibody and CD4(+) T cell responses to this antigen. The effect is dependent on the low affinity receptor for IgE, CD23, and the receptor must be expressed on B cells. In vitro, IgE-antigen complexes are endocytosed via CD23 on B cells, which subsequently present the antigen to CD4(+) T cells. This mechanism has been suggested to explain also IgE-mediated enhancement of immune responses in vivo. We recently found that CD23(+) B cells capture IgE-antigen complexes in peripheral blood and rapidly transport them to B cell follicles in the spleen. This provides an alternative explanation for the requirement for CD23(+) B cells. The aim of the present study was to determine whether B-cell mediated antigen presentation of IgE-antigen complexes explains the enhancing effect of IgE on immune responses in vivo. The ability of spleen cells, taken from mice 1-4 h after immunization with IgE-antigen, to present antigen to specific CD4(+) T cells was analyzed. Antigen presentation was intact when spleens were depleted of CD19(+) cells (i.e., primarily B cells) but was severely impaired after depletion of CD11c(+) cells (i.e., primarily dendritic cells). In agreement with this, the ability of IgE to enhance proliferation of CD4(+) T cells was abolished in CD11c-DTR mice conditionally depleted of CD11c(+) cells. Finally, the lack of IgE-mediated enhancemen of CD4(+) T cell responses in CD23(-/-) mice could be rescued by transfer of MHC-II-compatible as well as by MHC-II-incompatible CD23(+) B cells. These findings argue against the idea that IgE-mediated enhancement of specific CD4(+) T cell responses in vivo is caused by increased antigen presentation by B cells. A model where CD23(+) B cells act as antigen transporting cells, delivering antigen to CD11c(+) cells for presentation to T cells is consistent with available experimental data.


Subject(s)
Antigen Presentation/immunology , B-Lymphocytes/immunology , CD11c Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , Immunoglobulin E/immunology , Animals , Antigen Presentation/drug effects , Antigens/immunology , B-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Diphtheria Toxin/pharmacology , Histocompatibility Antigens Class II/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, IgE/deficiency , Receptors, IgE/metabolism
11.
J Exp Med ; 205(6): 1343-55, 2008 Jun 09.
Article in English | MEDLINE | ID: mdl-18519649

ABSTRACT

Over 80% of the body's activated B cells are located in mucosal sites, including the intestine. The intestine contains IgM(+) B cells, but these cells have not been characterized phenotypically or in terms of their developmental origins. We describe a previously unidentified and unique subset of immunoglobulin M(+) B cells that present with an AA4.1(-)CD21(-)CD23(-) major histocompatibility complex class II(bright) surface phenotype and are characterized by a low frequency of somatic hypermutation and the potential ability to produce interleukin-12p70. This B cell subset resides within the normal mucosa of the large intestine and expands in response to inflammation. Some of these intestinal B cells originate from the AA4.1(+) immature B2 cell pool in the steady state and are also recruited from the recirculating naive B cell pool in the context of intestinal inflammation. They develop in an antigen-independent and BAFF-dependent manner in the absence of T cell help. Expansion of these cells can be induced in the absence of the spleen and gut-associated lymphoid tissues. These results describe the existence of an alternative pathway of B cell maturation in the periphery that gives rise to a tissue-specific B cell subset.


Subject(s)
Adaptor Proteins, Signal Transducing/analysis , B-Lymphocyte Subsets/immunology , Immunity, Mucosal , Immunoglobulin M/immunology , Intestinal Mucosa/immunology , Intestine, Large/immunology , Membrane Glycoproteins/analysis , Receptors, Complement 3d/deficiency , Receptors, IgE/deficiency , Animals , Antibodies/immunology , Autophagy-Related Proteins , HLA-D Antigens/immunology , Humans , Immunophenotyping , Inflammation/immunology , Mice
12.
Scand J Immunol ; 66(2-3): 261-70, 2007.
Article in English | MEDLINE | ID: mdl-17635803

ABSTRACT

IgE administered with its specific antigen in vivo induces enhanced proliferation of specific T cells as well as enhanced production of specific antibodies. Both effects are dependent on the low-affinity receptor for IgE (CD23) and the underlying mechanism is thought to be increased antigen presentation following uptake of IgE/antigen complexes via CD23(+) B cells. By contrast, CD23 negatively regulates antibody responses to antigens administered with alum, i.e. without IgE. This effect has been observed as low IgG1 and IgE responses in transgenic mice overexpressing CD23 (CD23Tg). The present study was designed to test whether IgE could enhance antibody and T-cell responses in CD23Tg animals or whether CD23's downregulatory effect precludes IgE-mediated enhancement. IgE-anti-TNP administered with OVA-TNP enhances the OVA-specific antibody responses in wild-type (wt) and CD23Tg mice equally well. Interestingly, the total magnitude of antibody responses to IgE + OVA-TNP and to uncomplexed OVA-TNP, as well as to sheep erythrocytes and keyhole limpet haemocyanine, were lower in the CD23Tg mice. IgE induced proliferation of OVA-specific CD4(+) T cells to the same degree in wt and CD23Tg mice. The effect on T cells was dependent on CD23(+) B cells as demonstrated in in vitro proliferation assays. In conclusion, CD23 does indeed have dual immunoregulatory effects in the same animal. The receptor mediates enhancement of antibody and T-cell responses to IgE-complexed antigen, most likely via increased presentation of complexed antigen, while it negatively regulates the total antibody response to a variety of antigens.


Subject(s)
Antibody Specificity , Immunoglobulin E/physiology , Receptors, IgE/biosynthesis , Receptors, IgE/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antibody Specificity/genetics , Antigen Presentation/genetics , Cells, Cultured , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Transgenic , Receptors, IgE/deficiency
13.
Blood ; 109(12): 5308-17, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17341663

ABSTRACT

RabGEF1/Rabex-5, a guanine nucleotide exchange factor (GEF) for the endocytic pathway regulator, Rab5, contains a Vps9 domain, an A20-like zinc finger (ZnF) domain, and a coiled coil domain. To investigate the importance of these domains in regulating receptor internalization and cell activation, we lentivirally delivered RabGEF1 mutants into RabGEF1-deficient (-/-) mast cells and examined Fc epsilon RI-dependent responses. Wild-type RabGEF1 expression corrected phenotypic abnormalities in -/- mast cells, including decreased basal Fc epsilon RI expression, slowed Fc epsilon RI internalization, elevated IgE + Ag-induced degranulation and IL-6 production, and the decreased ability of -/- cytosol to support endosome fusion. We showed that RabGEF1's ZnF domain has ubiquitin ligase activity. Moreover, the coiled coil domain of RabGEF1 is required for Rabaptin-5 binding and for maintaining basal levels of Rabaptin-5 and surface Fc epsilon RI. However, mutants lacking either of these domains normalized phenotypic abnormalities in IgE + antigen-activated -/- mast cells. By contrast, correction of these -/- phenotypes required a functional Vps9 domain. Thus, Fc epsilon RI-mediated mast cell functional activation is dependent on RabGEF1's GEF activity.


Subject(s)
Gene Expression Regulation , Guanine Nucleotide Exchange Factors/physiology , Mast Cells/physiology , Receptors, IgE/genetics , Bone Marrow Cells , Cell Degranulation , Cells, Cultured , Endocytosis , Guanine Nucleotide Exchange Factors/chemistry , Humans , Interleukin-6/biosynthesis , Mast Cells/cytology , Receptors, IgE/deficiency
14.
Eur J Immunol ; 36(7): 1917-25, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16783846

ABSTRACT

Immunoglobulin E (IgE) is the key effector element in allergic diseases ranging from innocuous hay fever to life-threatening anaphylactic shock. Compared to other Ig classes, IgE serum levels are very low. In its membrane-bound form (mIgE), IgE behaves as a classical antigen receptor on B lymphocytes. Expression of mIgE is essential for subsequent recruitment of IgE-secreting cells. We show that in activated, mIgE-bearing B cells, mRNA for the membrane forms of both murine and human epsilon (epsilon) heavy chains (HC) are poorly expressed compared to mRNA for the secreted forms. In contrast, in mIgG-bearing B cells, mRNA for the membrane forms of murine gamma-1 (gamma1) and the corresponding human gamma4 HC are expressed at a much higher level than mRNA for the respective secreted forms. We show that these findings correlate with the presence of deviant polyadenylation signal hexamers in the 3' untranslated region (UTR) of both murine and human epsilon genes, causing inefficient processing of primary transcripts and thus poor expression of the proteins and poor recruitment of IgE-producing cells in the immune response. Thus, we have identified a genetic steering mechanism in the regulation of IgE synthesis that represents a further means to restrain potentially dangerous, high serum IgE levels.


Subject(s)
Antibody-Producing Cells/immunology , Immunoglobulin E/biosynthesis , Immunoglobulin E/genetics , RNA Processing, Post-Transcriptional/immunology , RNA, Messenger/metabolism , Animals , Antibody-Producing Cells/cytology , Antibody-Producing Cells/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Humans , Immunoglobulin E/metabolism , Mice , Mice, Knockout , Receptors, IgE/deficiency , Receptors, IgE/genetics
15.
J Immunol ; 176(5): 3157-64, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16493075

ABSTRACT

Studies in both humans and rodents have suggested that CD8+ T cells contribute to the development of airway hyperresponsiveness (AHR) and that leukotriene B4 (LTB4) is involved in the chemotaxis of effector CD8+ T cells (T(EFF)) to the lung by virtue of their expression of BLT1, the receptor for LTB4. In the present study, we used a mast cell-CD8-dependent model of AHR to further define the role of BLT1 in CD8+ T cell-mediated AHR. C57BL/6+/+ and CD8-deficient (CD8-/-) mice were passively sensitized with anti-OVA IgE and exposed to OVA via the airways. Following passive sensitization and allergen exposure, C57BL/6+/+ mice developed altered airway function, whereas passively sensitized and allergen-exposed CD8-/- mice failed to do so. CD8-/- mice reconstituted with CD8+ T(EFF) developed AHR in response to challenge. In contrast, CD8-/- mice reconstituted with BLT1-deficient effector CD8+ T cells did not develop AHR. The induction of increased airway responsiveness following transfer of CD8+ T(EFF) or in wild-type mice could be blocked by administration of an LTB4 receptor antagonist confirming the role of BLT1 in CD8+ T cell-mediated AHR. Together, these data define the important role for mast cells and the LTB4-BLT1 pathway in the development of CD8+ T cell-mediated allergic responses in the lung.


Subject(s)
Bronchial Hyperreactivity/immunology , CD8-Positive T-Lymphocytes/immunology , Leukotriene B4/metabolism , Mast Cells/immunology , Receptors, Leukotriene B4/physiology , Receptors, Purinergic P2/physiology , Adoptive Transfer , Animals , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/metabolism , CD8-Positive T-Lymphocytes/transplantation , Female , Interleukin-13/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, IgE/biosynthesis , Receptors, IgE/deficiency , Receptors, IgE/genetics , Receptors, Leukotriene B4/deficiency , Receptors, Leukotriene B4/genetics , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/genetics
16.
J Allergy Clin Immunol ; 116(5): 1020-7, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16275370

ABSTRACT

BACKGROUND: There have been few reports using animal models to study the development of allergic rhinitis. Characterization of such a model in mice would be advantageous given the availability of reagents and gene-manipulated strains. OBJECTIVE: We sought to develop a murine model of allergic rhinitis in the absence of lower airway changes. METHODS: After sensitization and challenge, both wild-type and FcepsilonRI-deficient mice were studied for their ability to develop early- and late-phase nasal responses. In the invasive approach, direct measurements of nasal airway resistance (R(NA)) were obtained; in the noninvasive approach using whole-body plethysmography, respiratory frequency and expiratory and inspiratory times were monitored. In both approaches, nasal responses were determined either acutely after challenge (early phase) or 24 hours after challenge (late phase). RESULTS: After challenge of sensitized mice, R(NA) significantly increased. In parallel, respiratory frequency significantly decreased and was highly correlated with the increases in R(NA). Sensitized wild-type mice had an early-phase nasal response and persistent nasal blockage (late-phase response) after allergen challenge. In contrast, sensitized and challenged FcepsilonRI alpha-chain-deficient mice did not have an early-phase nasal reaction and exhibited reduced nasal blockage and lower IL-13 levels in nasal tissue homogenates. CONCLUSIONS: These data indicate that FcepsilonRI is essential to development of an early-phase nasal response and contributes to the development of the late-phase nasal response. These invasive and noninvasive approaches provide new opportunities to evaluate the mechanisms underlying the development of nasal responses to allergen and to assess various therapeutic interventions.


Subject(s)
Receptors, IgE/metabolism , Rhinitis, Allergic, Perennial/physiopathology , Administration, Intranasal , Airway Resistance , Animals , Binding, Competitive , Drug Administration Schedule , Eosinophils/pathology , Immunization , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Injections, Intraperitoneal , Interleukin-13/metabolism , Kinetics , Mice , Mice, Knockout , Nasal Cavity/metabolism , Nasal Cavity/pathology , Nasal Cavity/physiopathology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Receptors, IgE/deficiency , Respiratory Mechanics , Rhinitis, Allergic, Perennial/immunology , Rhinitis, Allergic, Perennial/pathology
17.
J Immunol ; 175(3): 1473-82, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16034084

ABSTRACT

IgE Abs, passively administered together with their specific Ag, can enhance the production of Abs recognizing this Ag by >100-fold. IgE-mediated feedback enhancement requires the low affinity receptor for IgE, CD23. One possible mechanism is that B cells take up IgE-Ag via CD23 and efficiently present Ag to Th cells, resulting in better Ab responses. To test whether IgE Abs have an effect on Th cells in vivo, mice were adoptively transferred with CD4+ T cells expressing a transgenic OVA-specific TCR, before immunization with IgE anti-TNP (2,4,6-trinitrophenyl) plus OVA-TNP or with OVA-TNP alone. IgE induced a 6- to 21-fold increase in the number of OVA-specific T cells. These cells acquired an activated phenotype and were visible in splenic T cell zones. The T cell response peaked 3 days after immunization and preceded the OVA-specific Ab response by a few days. Transfer of CD23+ B cells to CD23-deficient mice rescued their ability to respond to IgE-Ag. Interestingly, in this situation also CD23-negative B cells produce enhanced levels of OVA-specific Abs. The data are compatible with the Ag presentation model and suggest that B cells can take up Ag via "unspecific" receptors and activate naive T cells in vivo.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/physiology , Antibody Formation , B-Lymphocyte Subsets/immunology , Epitopes, T-Lymphocyte/immunology , Immunoglobulin E/administration & dosage , Immunoglobulin E/physiology , Receptors, IgE/biosynthesis , T-Lymphocytes/immunology , Animals , Antibody Formation/genetics , Antigen Presentation/genetics , Antigen-Antibody Complex/administration & dosage , Antigen-Antibody Complex/physiology , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/transplantation , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Epitopes, T-Lymphocyte/metabolism , Haptens/administration & dosage , Haptens/immunology , Immunoglobulin E/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Receptors, IgE/deficiency , Receptors, IgE/genetics , T-Lymphocytes/metabolism , Trinitrobenzenes/administration & dosage , Trinitrobenzenes/immunology
18.
J Immunol ; 175(10): 6885-92, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16272347

ABSTRACT

Aggregation of the high affinity receptor for IgE (FcepsilonRI) induces activation of mast cells. In this study we show that upon low intensity stimulation of FcepsilonRI with monomeric IgE, IgE plus anti-IgE, or IgE plus low Ag, Lyn (a Src family kinase) positively regulates degranulation, cytokine production, and survival, whereas Lyn works as a negative regulator of high intensity stimulation with IgE plus high Ag. Low intensity stimulation suppressed Lyn kinase activity and its association with FcepsilonRI beta subunit, whereas high intensity stimulation enhanced Lyn activity and its association with FcepsilonRI beta. The latter induced much higher levels of FcepsilonRI beta phosphorylation and Syk activity than the former. Downstream positive signaling molecules, such as Akt and p38, were positively and negatively regulated by Lyn upon low and high intensity stimulations, respectively. In contrast, the negative regulators, SHIP and Src homology 2 domain-containing protein tyrosine phosphatase-1, interacted with FcepsilonRI beta, and their phosphorylation was controlled by Lyn. Therefore, we conclude that Lyn-mediated positive vs negative regulation depends on the intensity of the stimuli. Studies of mutant FcepsilonRI beta showed that FcepsilonRI beta subunit-ITAM (ITAM motif) regulates degranulation and cytokine production positively and negatively depending on the intensity of FcepsilonRI stimulation. Furthermore, Lyn-mediated negative regulation was shown to be exerted via the FcepsilonRI beta-ITAM.


Subject(s)
Mast Cells/immunology , Mast Cells/metabolism , Receptors, IgE/metabolism , src-Family Kinases/metabolism , Animals , Antigens/administration & dosage , Cell Degranulation , Cell Survival , Cells, Cultured , Cytokines/biosynthesis , Immunoglobulin E/administration & dosage , Intracellular Signaling Peptides and Proteins/metabolism , Mast Cells/cytology , Mast Cells/physiology , Mice , Mice, Knockout , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Phosphatase 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor Aggregation , Receptors, IgE/deficiency , Receptors, IgE/genetics , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/deficiency , src-Family Kinases/genetics
19.
Int Immunol ; 7(8): 1237-41, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7495730

ABSTRACT

Human intestinal intraepithelial lymphocytes (iIEL) are a unique population of predominantly CD8 alpha beta+, TCR alpha beta+ lymphocytes and, to a lesser extent, TCP gamma delta+ lymphocytes that proliferate poorly to anti-CD3 mitogenic signals but display significant cytolytic activity. Studies in mouse model systems have shown that the gamma chain of the high-affinity receptor for IgE (Fc epsilon RI gamma) may substitute for the zeta chain in the TCR-CD3 complex of iIEL. This has suggested that the functional properties of these cells may be associated with an altered composition of the TCR-CD3 complex. We therefore analyzed the TCR-CD3 complex of normal human iIEL. One- and two-dimensional non-reducing/reducing SDS-PAGE analysis of CD3 gamma, CD3 delta, CD3 epsilon, zeta and Fc epsilon RI gamma chain immunoprecipitates of cell surface radiolabeled proteins with subunit-specific antibodies revealed a TCR-CD3 complex without associated Fc epsilon RI gamma chains. Thus, normal human iIEL contain a TCR-CD3 complex that consists predominantly of zeta homodimers in association with the alpha beta TCR and CD3 gamma, delta and epsilon, similar to the majority of peripheral lymphocytes. This indicates that the distinct properties of human iIEL are not associated with substitutions of the Fc epsilon RI gamma chain in the TCR-CD3 complex.


Subject(s)
Intestinal Mucosa/immunology , Receptor-CD3 Complex, Antigen, T-Cell/chemistry , Receptors, IgE/deficiency , T-Lymphocytes/immunology , Animals , Humans
20.
Proc Natl Acad Sci U S A ; 91(15): 6835-9, 1994 Jul 19.
Article in English | MEDLINE | ID: mdl-8041705

ABSTRACT

The CD23 antigen, a low-affinity receptor for IgE (Fc epsilon RII), is a type II membrane-bound glycoprotein expressed on various cells, particularly mature B cells. A number of functions have been ascribed to CD23, including specific regulation of IgE production, IgE-mediated cytotoxicity and release of mediators, IgE-dependent antigen focusing, promotion of B-cell growth, prevention of germinal center B cells from apoptosis, proliferation of myeloid precursors, and maturation of early thymocytes. It is not clear whether these activities represent in vivo functions. To explore in vivo functions of CD23, we have produced CD23-deficient mice. These mice displayed normal lymphocyte differentiation and could mount normal antibody responses, including IgE responses upon immunization with T-dependent antigens and infection with Nippostrongyrus brasiliensis. Germinal center formation after immunization and in vitro proliferative response of B cells were not affected in mutant mice. However, antigen-specific IgE-mediated enhancement of antibody responses was severely impaired.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin E/immunology , Receptors, IgE/immunology , Animals , Antibody Formation , B-Lymphocytes/cytology , Cell Differentiation , Cell Division , Enzyme-Linked Immunosorbent Assay , Immunophenotyping , Mice , Microscopy, Fluorescence , Phenotype , Receptors, IgE/deficiency , Spleen/cytology , Spleen/immunology
SELECTION OF CITATIONS
SEARCH DETAIL