Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 9(5): 510, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29724998

RESUMO

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.


Assuntos
Basófilos/imunologia , Hipersensibilidade/genética , Imunoglobulina E/genética , Interleucina-3/genética , Receptores de IgE/genética , Animais , Basófilos/efeitos dos fármacos , Basófilos/patologia , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Hipersensibilidade/imunologia , Hipersensibilidade/fisiopatologia , Imunoglobulina E/imunologia , Interleucina-3/imunologia , Interleucina-3/farmacologia , Interleucina-5/genética , Interleucina-5/imunologia , Interleucina-5/farmacologia , Mastócitos/imunologia , Mastócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cultura Primária de Células , Receptores de IgE/deficiência , Receptores de IgE/imunologia , Transdução de Sinais , Transcrição Gênica
2.
J Immunol ; 197(7): 2583-8, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27566822

RESUMO

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.


Assuntos
Imunoglobulina E/imunologia , Vigilância Imunológica/imunologia , Neoplasias/imunologia , Receptores de IgE/imunologia , Adjuvantes Imunológicos , Animais , Engenharia Genética , Imunoterapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neoplasias/genética , Neoplasias/patologia , Neoplasias/terapia , Receptores de IgE/deficiência
3.
Endocrinology ; 156(11): 4047-58, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26295369

RESUMO

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.


Assuntos
Metabolismo Energético/genética , Obesidade/genética , Receptores de IgE/genética , Aumento de Peso/genética , Células 3T3-L1 , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Dieta Hiperlipídica/efeitos adversos , Feminino , Expressão Gênica , Teste de Tolerância a Glucose , Humanos , Immunoblotting , Imunoglobulina E/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Obesidade/etiologia , Obesidade/metabolismo , Obesidade Mórbida/sangue , PPAR gama/genética , PPAR gama/metabolismo , Interferência de RNA , Receptores de IgE/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Mol Immunol ; 53(3): 270-3, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22964482

RESUMO

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.


Assuntos
Basófilos/imunologia , Receptores de IgE/química , Receptores de IgE/metabolismo , Alanina/química , Substituição de Aminoácidos , Animais , Degranulação Celular/imunologia , Linhagem Celular Tumoral , Ligação de Hidrogênio , Leucemia Basofílica Aguda/genética , Leucemia Basofílica Aguda/imunologia , Leucemia Basofílica Aguda/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Domínios e Motivos de Interação entre Proteínas , Subunidades Proteicas , Ratos , Receptores de IgE/deficiência , Receptores de IgE/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina/química , Transdução de Sinais , Treonina/química , Transfecção
5.
J Immunol ; 190(2): 539-48, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23241885

RESUMO

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.


Assuntos
Basófilos/imunologia , Mastócitos/imunologia , Receptores Acoplados a Proteínas G/imunologia , Receptores Histamínicos/imunologia , Receptores de IgE/imunologia , Rinite Alérgica Perene/imunologia , Transferência Adotiva , Animais , Basófilos/efeitos dos fármacos , Basófilos/metabolismo , Feminino , Antagonistas dos Receptores Histamínicos/farmacologia , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Camundongos , Camundongos Knockout , Modelos Imunológicos , Mucosa Nasal/efeitos dos fármacos , Mucosa Nasal/imunologia , Receptores Acoplados a Proteínas G/genética , Receptores Histamínicos/genética , Receptores Histamínicos H4 , Receptores de IgE/deficiência , Receptores de IgE/genética , Rinite Alérgica , Rinite Alérgica Perene/genética , Rinite Alérgica Perene/terapia
6.
PLoS One ; 6(7): e21760, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21765910

RESUMO

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.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos B/imunologia , Antígeno CD11c/imunologia , Linfócitos T CD4-Positivos/imunologia , Imunoglobulina E/imunologia , Animais , Apresentação de Antígeno/efeitos dos fármacos , Antígenos/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Toxina Diftérica/farmacologia , Antígenos de Histocompatibilidade Classe II/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores de IgE/deficiência , Receptores de IgE/metabolismo
7.
J Immunol ; 187(1): 164-71, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21622859

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/patologia , Mediadores da Inflamação/metabolismo , Receptores de IgE/metabolismo , Células Th2/imunologia , Células Th2/patologia , Alérgenos/toxicidade , Animais , Antígenos de Plantas/toxicidade , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/metabolismo , Feminino , Humanos , Mediadores da Inflamação/fisiologia , Mediadores da Inflamação/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/toxicidade , Ligação Proteica/genética , Ligação Proteica/imunologia , Receptores de IgE/deficiência , Receptores de IgE/fisiologia , Células Th2/metabolismo , Fatores de Tempo
8.
J Immunol ; 183(7): 4530-6, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19748979

RESUMO

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.


Assuntos
Adjuvantes Imunológicos/fisiologia , Adjuvantes Imunológicos/uso terapêutico , Antineoplásicos/uso terapêutico , Imunoglobulina E/fisiologia , Imunoglobulina E/uso terapêutico , Receptores de IgE/fisiologia , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Adenocarcinoma/terapia , Adjuvantes Imunológicos/metabolismo , Animais , Antígenos de Neoplasias/imunologia , Antineoplásicos/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Reagentes de Ligações Cruzadas/administração & dosagem , Reagentes de Ligações Cruzadas/metabolismo , Reagentes de Ligações Cruzadas/uso terapêutico , Feminino , Técnicas de Introdução de Genes , Humanos , Imunoglobulina E/metabolismo , Leucemia Basofílica Aguda/imunologia , Leucemia Basofílica Aguda/patologia , Leucemia Basofílica Aguda/terapia , Linfoma de Células T/imunologia , Linfoma de Células T/patologia , Linfoma de Células T/terapia , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ratos , Receptores de IgE/deficiência , Receptores de IgE/genética
9.
J Immunol ; 182(12): 8110-7, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19494337

RESUMO

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.


Assuntos
Linfócitos B/imunologia , Terapia Genética , Imunoglobulina E/imunologia , Receptores de IgE/imunologia , Anafilaxia/imunologia , Animais , Linfócitos B/metabolismo , Linhagem Celular , Humanos , Imunização , Imunoglobulina E/genética , Imunoglobulina E/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Ligação Proteica/imunologia , Receptores de IgE/deficiência , Receptores de IgE/genética , Receptores de IgE/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo
10.
J Exp Med ; 205(6): 1343-55, 2008 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-18519649

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/análise , Subpopulações de Linfócitos B/imunologia , Imunidade nas Mucosas , Imunoglobulina M/imunologia , Mucosa Intestinal/imunologia , Intestino Grosso/imunologia , Glicoproteínas de Membrana/análise , Receptores de Complemento 3d/deficiência , Receptores de IgE/deficiência , Animais , Anticorpos/imunologia , Proteínas Relacionadas à Autofagia , Antígenos HLA-D/imunologia , Humanos , Imunofenotipagem , Inflamação/imunologia , Camundongos
11.
Scand J Immunol ; 66(2-3): 261-70, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17635803

RESUMO

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.


Assuntos
Especificidade de Anticorpos , Imunoglobulina E/fisiologia , Receptores de IgE/biossíntese , Receptores de IgE/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Especificidade de Anticorpos/genética , Apresentação de Antígeno/genética , Células Cultivadas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Receptores de IgE/deficiência
12.
Blood ; 109(12): 5308-17, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17341663

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Mastócitos/fisiologia , Receptores de IgE/genética , Células da Medula Óssea , Degranulação Celular , Células Cultivadas , Endocitose , Fatores de Troca do Nucleotídeo Guanina/química , Humanos , Interleucina-6/biossíntese , Mastócitos/citologia , Receptores de IgE/deficiência
13.
Eur J Immunol ; 36(7): 1917-25, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16783846

RESUMO

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.


Assuntos
Células Produtoras de Anticorpos/imunologia , Imunoglobulina E/biossíntese , Imunoglobulina E/genética , Processamento Pós-Transcricional do RNA/imunologia , RNA Mensageiro/metabolismo , Animais , Células Produtoras de Anticorpos/citologia , Células Produtoras de Anticorpos/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Humanos , Imunoglobulina E/metabolismo , Camundongos , Camundongos Knockout , Receptores de IgE/deficiência , Receptores de IgE/genética
14.
J Exp Med ; 203(4): 1105-16, 2006 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-16606668

RESUMO

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.


Assuntos
Linfócitos B/citologia , Basófilos/metabolismo , Interleucinas/fisiologia , Nippostrongylus/imunologia , Infecções por Strongylida/imunologia , Infecções por Strongylida/parasitologia , Linfócitos T/citologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Células Cultivadas , Citocinas/biossíntese , Citocinas/classificação , Interleucina-13/biossíntese , Interleucina-13/deficiência , Interleucina-13/genética , Interleucina-4/biossíntese , Interleucina-4/deficiência , Interleucina-4/genética , Interleucina-5/biossíntese , Interleucina-5/deficiência , Interleucina-5/genética , Interleucinas/administração & dosagem , Interleucinas/deficiência , Interleucinas/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores de IgE/deficiência , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
15.
J Immunol ; 176(5): 3157-64, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16493075

RESUMO

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.


Assuntos
Hiper-Reatividade Brônquica/imunologia , Linfócitos T CD8-Positivos/imunologia , Leucotrieno B4/metabolismo , Mastócitos/imunologia , Receptores do Leucotrieno B4/fisiologia , Receptores Purinérgicos P2/fisiologia , Transferência Adotiva , Animais , Hiper-Reatividade Brônquica/genética , Hiper-Reatividade Brônquica/metabolismo , Linfócitos T CD8-Positivos/transplante , Feminino , Interleucina-13/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/genética , Receptores de IgE/biossíntese , Receptores de IgE/deficiência , Receptores de IgE/genética , Receptores do Leucotrieno B4/deficiência , Receptores do Leucotrieno B4/genética , Receptores Purinérgicos P2/deficiência , Receptores Purinérgicos P2/genética
16.
J Immunol ; 175(10): 6885-92, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16272347

RESUMO

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.


Assuntos
Mastócitos/imunologia , Mastócitos/metabolismo , Receptores de IgE/metabolismo , Quinases da Família src/metabolismo , Animais , Antígenos/administração & dosagem , Degranulação Celular , Sobrevivência Celular , Células Cultivadas , Citocinas/biossíntese , Imunoglobulina E/administração & dosagem , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mastócitos/citologia , Mastócitos/fisiologia , Camundongos , Camundongos Knockout , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Proteína Fosfatase 1 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Agregação de Receptores , Receptores de IgE/deficiência , Receptores de IgE/genética , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Quinases da Família src/deficiência , Quinases da Família src/genética
17.
J Allergy Clin Immunol ; 116(5): 1020-7, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16275370

RESUMO

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.


Assuntos
Receptores de IgE/metabolismo , Rinite Alérgica Perene/fisiopatologia , Administração Intranasal , Resistência das Vias Respiratórias , Animais , Ligação Competitiva , Esquema de Medicação , Eosinófilos/patologia , Imunização , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Imunoglobulina G/sangue , Injeções Intraperitoneais , Interleucina-13/metabolismo , Cinética , Camundongos , Camundongos Knockout , Cavidade Nasal/metabolismo , Cavidade Nasal/patologia , Cavidade Nasal/fisiopatologia , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Receptores de IgE/deficiência , Mecânica Respiratória , Rinite Alérgica Perene/imunologia , Rinite Alérgica Perene/patologia
18.
J Immunol ; 175(3): 1473-82, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16034084

RESUMO

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.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/fisiologia , Formação de Anticorpos , Subpopulações de Linfócitos B/imunologia , Epitopos de Linfócito T/imunologia , Imunoglobulina E/administração & dosagem , Imunoglobulina E/fisiologia , Receptores de IgE/biossíntese , Linfócitos T/imunologia , Animais , Formação de Anticorpos/genética , Apresentação de Antígeno/genética , Complexo Antígeno-Anticorpo/administração & dosagem , Complexo Antígeno-Anticorpo/fisiologia , Subpopulações de Linfócitos B/metabolismo , Subpopulações de Linfócitos B/transplante , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/transplante , Epitopos de Linfócito T/metabolismo , Haptenos/administração & dosagem , Haptenos/imunologia , Imunoglobulina E/metabolismo , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Receptores de IgE/deficiência , Receptores de IgE/genética , Linfócitos T/metabolismo , Trinitrobenzenos/administração & dosagem , Trinitrobenzenos/imunologia
19.
J Immunol ; 172(10): 6398-406, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15128831

RESUMO

In certain models of allergic airway disease, mast cells facilitate the development of inflammation and airway hyper-responsiveness (AHR). To define the role of the high affinity IgE receptor (FcepsilonRI) in the development of AHR, mice with a disruption of the alpha subunit of the high affinity IgE receptor (FcepsilonRI(-/-)) were exposed on 10 consecutive days to nebulized OVA. Forty-eight hours after the last nebulization, airway responsiveness was monitored by the contractile response of tracheal smooth muscle to electrical field stimulation (EFS). After the 10-day OVA challenge protocol, wild-type mice demonstrated increased responsiveness to EFS, whereas similarly challenged FcepsilonRI(-/-) mice showed a low response to EFS, similar to nonexposed animals. Further, allergen-challenged FcepsilonRI(-/-) mice showed less airway inflammation, goblet cell hyperplasia, and lower levels of IL-13 in lung homogenates compared with the controls. IL-13-deficient mice failed to develop an increased response to EFS or goblet cell hyperplasia after the 10-day OVA challenge. We transferred bone marrow-derived mast cells from wild-type mice to FcepsilonRI(-/-) mice 1 day before initiating the challenge protocol. After the 10-day OVA challenge, recipient FcepsilonRI(-/-) mice demonstrated EFS-induced responses similar to those of challenged wild-type mice. Transferred mast cells could be detected in tracheal preparations. These results show that FcepsilonRI is important for the development of AHR after an aerosolized allergen sensitization protocol and that this effect is mediated through FcepsilonRI on mast cells and production of IL-13 in the lung.


Assuntos
Adjuvantes Imunológicos , Alérgenos/administração & dosagem , Hiper-Reatividade Brônquica/imunologia , Interleucina-13/fisiologia , Mastócitos/imunologia , Receptores de IgE/fisiologia , Transferência Adotiva , Aerossóis , Alérgenos/imunologia , Animais , Transplante de Medula Óssea/imunologia , Hiper-Reatividade Brônquica/genética , Estimulação Elétrica , Feminino , Células Caliciformes/imunologia , Células Caliciformes/patologia , Hiperplasia , Imunoglobulina E/biossíntese , Imunoglobulina G/biossíntese , Inflamação/genética , Inflamação/patologia , Interleucina-13/deficiência , Interleucina-13/genética , Interleucina-13/metabolismo , Leucopenia/genética , Leucopenia/imunologia , Pulmão/imunologia , Pulmão/patologia , Mastócitos/patologia , Mastócitos/transplante , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Nebulizadores e Vaporizadores , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Receptores de IgE/deficiência , Receptores de IgE/genética , Traqueia/citologia , Traqueia/imunologia , Traqueia/metabolismo
20.
J Immunol ; 172(9): 5329-37, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15100272

RESUMO

The latent membrane protein 2A (LMP2A) of EBV plays a key role in regulating viral latency and EBV pathogenesis by functionally mimicking a constitutively active B cell Ag receptor. When expressed as a B cell-specific transgene in mice, LMP2A drives B cell development, resulting in the bypass of normal developmental checkpoints. In this study, we have demonstrated that expression of LMP2A in transgenic mice results in B cell development that exclusively favors B-1 cells. This switch to B-1 cell development occurs at the pre-B-cell stage of normal B cell development in the bone marrow, a B cell stage much earlier than appreciated for B-1 commitment. This finding indicates that all pre-B cells have the capacity to assume a B-1 cell phenotype if they encounter the appropriate signal during normal development. Furthermore, these studies offer insight into EBV latency and pathogenesis in the human host.


Assuntos
Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/imunologia , Antígenos CD5/biossíntese , Herpesvirus Humano 4/imunologia , Receptores de Antígenos de Linfócitos B/química , Proteínas da Matriz Viral/fisiologia , Animais , Antígenos CD/biossíntese , Subpopulações de Linfócitos B/enzimologia , Subpopulações de Linfócitos B/metabolismo , Biomarcadores/análise , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Divisão Celular/imunologia , Linhagem Celular , Precursores Enzimáticos/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Imunoglobulina M/biossíntese , Imunoglobulina M/sangue , Peptídeos e Proteínas de Sinalização Intracelular , Leucossialina , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Tirosina Quinases/fisiologia , Receptores de Antígenos de Linfócitos B/deficiência , Receptores de IgE/deficiência , Sialoglicoproteínas/biossíntese , Homologia Estrutural de Proteína , Quinase Syk , Acetato de Tetradecanoilforbol/farmacologia , Proteínas da Matriz Viral/biossíntese , Proteínas da Matriz Viral/química , Latência Viral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...