Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 57(2): 319-332.e6, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38295798

RESUMO

Tuft cells in mucosal tissues are key regulators of type 2 immunity. Here, we examined the impact of the microbiota on tuft cell biology in the intestine. Succinate induction of tuft cells and type 2 innate lymphoid cells was elevated with loss of gut microbiota. Colonization with butyrate-producing bacteria or treatment with butyrate suppressed this effect and reduced intestinal histone deacetylase activity. Epithelial-intrinsic deletion of the epigenetic-modifying enzyme histone deacetylase 3 (HDAC3) inhibited tuft cell expansion in vivo and impaired type 2 immune responses during helminth infection. Butyrate restricted stem cell differentiation into tuft cells, and inhibition of HDAC3 in adult mice and human intestinal organoids blocked tuft cell expansion. Collectively, these data define a HDAC3 mechanism in stem cells for tuft cell differentiation that is dampened by a commensal metabolite, revealing a pathway whereby the microbiota calibrate intestinal type 2 immunity.


Assuntos
Mucosa Intestinal , Microbiota , Adulto , Camundongos , Humanos , Animais , Células em Tufo , Butiratos/farmacologia , Butiratos/metabolismo , Imunidade Inata , Linfócitos/metabolismo , Intestinos , Histona Desacetilases/metabolismo , Diferenciação Celular
2.
J Allergy Clin Immunol ; 149(2): 671-684.e9, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34186142

RESUMO

BACKGROUND: Administering allergens in increasing doses can temporarily suppress IgE-mediated allergy and anaphylaxis by desensitizing mast cells and basophils; however, allergen administration during desensitization therapy can itself induce allergic responses. Several small molecule drugs and nutraceuticals have been used clinically and experimentally to suppress these allergic responses. OBJECTIVES: This study sought to optimize drug inhibition of IgE-mediated anaphylaxis. METHODS: Several agents were tested individually and in combination for ability to suppress IgE-mediated anaphylaxis in conventional mice, FcεRIα-humanized mice, and reconstituted immunodeficient mice that have human mast cells and basophils. Hypothermia was the readout for anaphylaxis; therapeutic efficacy was measured by degree of inhibition of hypothermia. Serum mouse mast cell protease 1 level was used to measure extent of mast cell degranulation. RESULTS: Histamine receptor 1 (HR1) antagonists, ß-adrenergic agonists, and a spleen tyrosine kinase (Syk) inhibitor were best at individually inhibiting IgE-mediated anaphylaxis. A Bruton's tyrosine kinase (BTK) inhibitor, administered alone, only inhibited hypothermia when FcεRI signaling was suboptimal. Combinations of these agents could completely or nearly completely inhibit IgE-mediated hypothermia in these models. Both Syk and BTK inhibition decreased mast cell degranulation, but only Syk inhibition also blocked desensitization. Many other agents that are used clinically and experimentally had little or no beneficial effect. CONCLUSIONS: Combinations of an HR1 antagonist, a ß-adrenergic agonist, and a Syk or a BTK inhibitor protect best against IgE-mediated anaphylaxis, while an HR1 antagonist plus a ß-adrenergic agonist ± a BTK antagonist is optimal for inhibiting IgE-mediated anaphylaxis without suppressing desensitization.


Assuntos
Anafilaxia/prevenção & controle , Imunoglobulina E/imunologia , Agonistas Adrenérgicos beta/uso terapêutico , Animais , Quimioterapia Combinada , Antagonistas dos Receptores Histamínicos/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Tirosina Quinases/antagonistas & inibidores
3.
J Allergy Clin Immunol ; 147(5): 1838-1854.e4, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33326804

RESUMO

BACKGROUND: Mast cell and basophil activation by antigen cross-linking of FcεRI-bound IgE is central to allergy pathogenesis. We previously demonstrated global suppression of this process by rapid desensitization with anti-FcεRIα mAbs. OBJECTIVES: We sought to determine whether use of monovalent (mv) anti-FcεRIα mAbs increases desensitization safety without loss of efficacy. METHODS: mv anti-human (hu) FcεRIα mAbs were produced with mouse-derived immunoglobulin variable regions and huIgG1 or huIgG4 C regions and were used to suppress murine IgE-mediated anaphylaxis and food allergy. mAbs were administered as a single dose or as serially increasing doses to mice that express hu instead of mouse FcεRIα; mice that additionally have an allergy-promoting IL-4Rα mutation; and hu cord blood-reconstituted immunodeficient, hu cytokine-secreting, mice that have large numbers of activated hu mast cells. Anaphylaxis susceptibility was sometimes increased by treatment with IL-4 or a ß-adrenergic receptor antagonist. RESULTS: mv anti-hu FcεRIα mAbs are considerably less able than divalent mAbs are to induce anaphylaxis and deplete mast cell and basophil IgE, but mv mAbs still strongly suppress IgE-mediated disease. The mv mAbs can be safely administered as a single large dose to mice with typical susceptibility to anaphylaxis, while a rapid desensitization approach safely suppresses disease in mice with increased susceptibility. Our huIgG4 variant of mv anti-huFcεRIα mAb is safer than our huIgG1 variant is, apparently because reduced interactions with FcεRs decrease ability to indirectly cross-link FcεRI. CONCLUSIONS: mv anti-FcεRIα mAbs more safely suppress IgE-mediated anaphylaxis and food allergy than divalent variants of the same mAbs do. These mv mAbs may be useful for suppression of huIgE-mediated disease.


Assuntos
Anafilaxia/tratamento farmacológico , Antialérgicos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Hipersensibilidade Alimentar/tratamento farmacológico , Imunoglobulina E/imunologia , Receptores de IgE/imunologia , Anafilaxia/imunologia , Animais , Antialérgicos/farmacologia , Anticorpos Monoclonais/farmacologia , Feminino , Hipersensibilidade Alimentar/imunologia , Imunoglobulina G/imunologia , Masculino , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/imunologia , Receptores de IgE/genética , Quinase Syk/imunologia
4.
J Allergy Clin Immunol ; 145(3): 907-921.e3, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31836406

RESUMO

BACKGROUND: Anaphylaxis is classically mediated by allergen cross-linking of IgE bound to the α chain of FcεRI, the mast cell/basophil high affinity IgE receptor. Allergen cross-linking of the IgE/FcεRI complex activates these cells, inducing release of disease-causing mediators, cytokines, and enzymes. We previously demonstrated that IgE-mediated anaphylaxis could be safely prevented in wild-type BALB/c mice by rapid desensitization with anti-mouse FcεRIα mAb. OBJECTIVE: This study sought to use humanized mice to extend these results to humans. METHODS: We actively immunized huFcεRIα/F709 mice, which express human (hu) instead of mouse FcεRIα and a mutant IL-4 receptor that lacks inhibitory function. We passively immunized huFcεRIα mice, as well as human cord blood-reconstituted reNSGS mice, which are immune-deficient, produce mast cell-stimulating human cytokines, and develop numerous human mast cells. For desensitization, we used anti-huFcεRIα mAbs that bind FcεRIα regardless of its association with IgE (noncompeting mAbs), and/or mAbs that compete with IgE for huFcεRIα binding (competing mAbs). Anaphylaxis was induced by intravenous injection of antigen or anti-huIgE mAb. RESULTS: Anti-huFcεRIα mAb rapid desensitization was safer and more effective than allergen rapid desensitization and suppressed anaphylaxis more rapidly than omalizumab or ligelizumab. Rapid desensitization of naïve, IgE-sensitized huFcεRIα mice and huFcεRIα/F709 mice that were egg-allergic with anti-FcεRIα mAbs safely removed >98% of IgE from peritoneal mast cells and completely suppressed IgE-mediated anaphylaxis. Rapid desensitization of reNSGS mice with anti-FcεRIα mAbs also safely removed ∼98% of mast cell IgE and prevented IgE-mediated anaphylaxis. CONCLUSIONS: Rapid desensitization with anti-FcεRIα mAbs may be a safe, effective, and practical way to prevent IgE-mediated anaphylaxis.


Assuntos
Anafilaxia/imunologia , Anticorpos Monoclonais/farmacologia , Dessensibilização Imunológica/métodos , Receptores de IgE/antagonistas & inibidores , Anafilaxia/prevenção & controle , Animais , Humanos , Camundongos , Camundongos Endogâmicos BALB C
5.
J Exp Med ; 200(7): 857-70, 2004 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-15466620

RESUMO

Experiments were performed to characterize and identify the cellular sources of the secondary interleukin (IL)-4 response to a T cell-dependent antigen. Mice were primed by immunization with goat anti-mouse immunoglobulin (Ig)D antibody (GaMD), which stimulates naive CD4+ T cells to secrete IL-4 in 3-4 d. When challenged with goat serum 14 d after immunization, GaMD-primed mice generated an IL-4 response that exceeded the primary response by approximately 100-fold, started in <2 h, and lasted for 4 d. Studies with 4get mice, in which cells with an accessible Il4 gene express a green fluorescent protein (GFP), revealed CD4+ memory T cells, natural killer T cells, basophils, mast cells, and eosinophils as possible rapid producers of IL-4. GFP(+)CD4+ T cells and basophils expanded more in the spleen than the other cell types during the primary response to GaMD. Quantitation of in vivo IL-4 production by the in vivo cytokine capture assay after individual cell types were selectively stimulated or deleted demonstrated that basophils and memory CD4+ T cells account for most of the secondary IL-4 response, with basophils initiating that response through IgE/FcepsilonRI-mediated signaling but secreting IL-4 for <4 h and memory T cells secreting IL-4 within 4 h and continuing to secrete this cytokine for 4 d.


Assuntos
Basófilos/metabolismo , Imunização , Memória Imunológica , Interleucina-4/biossíntese , Linfócitos T/metabolismo , Animais , Basófilos/imunologia , Quimases , Citocinas/imunologia , Citometria de Fluxo , Imunofluorescência , Proteínas de Fluorescência Verde/metabolismo , Interleucina-4/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Serina Endopeptidases/sangue , Baço/imunologia , Linfócitos T/imunologia , Fatores de Tempo
6.
Mucosal Immunol ; 13(2): 283-292, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31745261

RESUMO

Airway hyperresponsiveness (AHR) often defines asthma. Murine allergic airway disease (AAD), like human eosinophilic asthma, is characterized by AHR, eosinophilia, goblet cell metaplasia (GCM), smooth muscle hypercontractility, and increased production of IL-4 and IL-13-cytokines that induce these characteristics by binding to the IL-4Rα chain. We evaluated the epithelial and smooth muscle IL-4Rα-dependent contributions to AHR of BALB/c mice that possessed 0-2 functional IL-4Rα alleles and had airway disease induced by house dust mite extract (HDM) or exogenous IL-13. Two functional IL-4Rα alleles were required for maximal AHR, while only one functional allele was required for maximal GCM and systemic IL-4/IL-13 levels. Deletion of IL-4Rα from both smooth muscle and epithelial cells inhibited AHR >83% in mice with two functional IL-4Rα alleles. In mice with one functional IL-4Rα allele, selective epithelial cell IL-4Rα deletion maximally inhibited AHR, while selective smooth muscle IL-4Rα deletion decreased IL-13-induced, but not HDM-induced, AHR. Less IL-4Rα signaling is required to maximize the epithelial cell contribution to AHR compared to the smooth muscle contribution to AHR. In addition, epithelial cell responses to IL-4/IL-13 can increase the IL-4Rα-dependent smooth muscle contribution to AHR. These findings carry increasing relevance as IL-4Rα-targeted therapy is administered to human asthmatics.


Assuntos
Células Caliciformes/patologia , Hipersensibilidade/imunologia , Músculo Liso/metabolismo , Receptores de Superfície Celular/metabolismo , Hipersensibilidade Respiratória/imunologia , Mucosa Respiratória/metabolismo , Animais , Antígenos de Dermatophagoides/imunologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Pyroglyphidae , Receptores de Superfície Celular/genética , Transdução de Sinais
7.
J Exp Med ; 208(4): 853-67, 2011 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-21464224

RESUMO

Production of the cytokines IL-4 and IL-13 is increased in both human asthma and mouse asthma models, and Stat6 activation by the common IL-4/IL-13R drives most mouse model pathophysiology, including airway hyperresponsiveness (AHR). However, the precise cellular mechanisms through which IL-4Rα induces AHR remain unclear. Overzealous bronchial smooth muscle constriction is thought to underlie AHR in human asthma, but the smooth muscle contribution to AHR has never been directly assessed. Furthermore, differences in mouse versus human airway anatomy and observations that selective IL-13 stimulation of Stat6 in airway epithelium induces murine AHR raise questions about the importance of direct IL-4R effects on smooth muscle in murine asthma models and the relevance of these models to human asthma. Using transgenic mice in which smooth muscle is the only cell type that expresses or fails to express IL-4Rα, we demonstrate that direct smooth muscle activation by IL-4, IL-13, or allergen is sufficient but not necessary to induce AHR. Five genes known to promote smooth muscle migration, proliferation, and contractility are activated by IL-13 in smooth muscle in vivo. These observations demonstrate that IL-4Rα promotes AHR through multiple mechanisms and provide a model for testing smooth muscle-directed asthma therapeutics.


Assuntos
Hiper-Reatividade Brônquica/etiologia , Músculo Liso/fisiologia , Receptores de Interleucina-4/fisiologia , Alérgenos/imunologia , Animais , Feminino , Regulação da Expressão Gênica , Interleucina-13/farmacologia , Interleucina-4/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Músculo Liso/efeitos dos fármacos , Receptores de Interleucina-4/genética
8.
J Immunol ; 179(10): 6429-38, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17982031

RESUMO

IL-4 and IL-13 are each bound by soluble receptors (sRs) that block their activity. Both of these sRs (sIL-4Ralpha and sIL-13Ralpha2) are present in low nanogram per milliliter concentrations in the serum from unstimulated mice, but differences in affinity and half-life suggest differences in function. Serum IL-4/sIL-4Ralpha complexes rapidly dissociate, releasing active IL-4, whereas sIL-13Ralpha2 and IL-13 form a stable complex that has a considerably longer half-life than uncomplexed IL-13, sIL-13Ralpha2, IL-4, or sIL-4Ralpha. Approximately 25% of sIL-13Ralpha2 in serum is complexed to IL-13; this percentage and the absolute quantity of sIL-13Ralpha2 in serum increase considerably during a Th2 response. sIL-13Ralpha2 gene expression is up-regulated by both IL-4 and IL-13; the effect of IL-4 is totally IL-4Ralpha-dependent while the effect of IL-13 is partially IL-4Ralpha-independent. Inhalation of an IL-13/sIL-13Ralpha2 complex does not affect the expression of IL-13-inducible genes but increases the expression of two genes, Vnn1 and Pira-1, whose products activate APCs and promote neutrophilic inflammation. These observations suggest that sIL-4Ralpha predominantly sustains, increases, and diffuses the effects of IL-4, whereas sIL-13Ralpha2 limits the direct effects of IL-13 to the site of IL-13 production and forms a stable complex with IL-13 that may modify the quality and intensity of an allergic inflammatory response.


Assuntos
Regulação da Expressão Gênica/imunologia , Hipersensibilidade/imunologia , Subunidade alfa2 de Receptor de Interleucina-13/imunologia , Interleucina-13/imunologia , Interleucina-4/imunologia , Complexos Multiproteicos/imunologia , Receptores de Superfície Celular/imunologia , Amidoidrolases , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/imunologia , Proteínas Ligadas por GPI , Regulação da Expressão Gênica/efeitos dos fármacos , Meia-Vida , Hipersensibilidade/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-13/biossíntese , Interleucina-13/farmacologia , Subunidade alfa2 de Receptor de Interleucina-13/metabolismo , Interleucina-4/biossíntese , Interleucina-4/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Complexos Multiproteicos/biossíntese , Complexos Multiproteicos/farmacologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos/biossíntese , Receptores Imunológicos/imunologia , Solubilidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA