Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunity ; 57(2): 319-332.e6, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38295798

RESUMEN

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.


Asunto(s)
Mucosa Intestinal , Microbiota , Adulto , Ratones , Humanos , Animales , Células en Penacho , Butiratos/farmacología , Butiratos/metabolismo , Inmunidad Innata , Linfocitos/metabolismo , Intestinos , Histona Desacetilasas/metabolismo , Diferenciación Celular
2.
J Allergy Clin Immunol ; 152(1): 195-204.e3, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36804390

RESUMEN

BACKGROUND: Histamine is a critical mediator of anaphylaxis, a neurotransmitter, and a regulator of gastric acid secretion. Histidine decarboxylase is a rate-limiting enzyme for histamine synthesis. However, in vivo regulation of Hdc, the gene that encodes histidine decarboxylase, is poorly understood. OBJECTIVE: We sought to investigate how enhancers regulate Hdc gene transcription and histamine synthesis in resting conditions and in a mouse model of anaphylaxis. METHODS: H3K27 acetylation histone modification and chromatin accessibility were used to identify candidate enhancers. The enhancer activity of candidate enhancers was measured in a reporter gene assay, and the function enhancers were validated by CRISPR deletion. RESULTS: Deletion of the GC box, which binds to zinc finger transcription factors, in the proximal Hdc enhancer reduced Hdc gene transcription and histamine synthesis in mouse and human mast cell lines. Mast cells, basophils, brain cells, and stomach cells from GC box-deficient mice transcribed the Hdc gene much less than similar cells from wild-type mice, and Hdc GC box-deficient mice failed to develop anaphylaxis. CONCLUSION: The HDC GC box within the proximal enhancer in the mouse and human HDC gene is essential for Hdc gene transcription, histamine synthesis, and histamine-mediated anaphylaxis in vitro and in vivo.


Asunto(s)
Anafilaxia , Histidina Descarboxilasa , Humanos , Ratones , Animales , Histidina Descarboxilasa/genética , Histamina/metabolismo , Anafilaxia/genética , Línea Celular , Transcripción Genética
3.
Mucosal Immunol ; 15(6): 1375-1388, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-36114245

RESUMEN

Mutated and unmutated IgE and IgG play different and partly opposing roles in allergy development, but the mechanisms controlling their relative production are incompletely understood. Here, we analyzed the IgE-response in murine food allergy. Deep sequencing of the complementary-determining region (CDR) repertoires indicated that an ongoing unmutated extrafollicular IgE response coexists with a germinal center response, even after long-lasting allergen challenges. Despite overall IgG1-dominance, a significant proportion of clonotypes contained several-fold more IgE than IgG1. Clonotypes with differential bias to either IgE or IgG1 showed distinct hypermutation and clonal expansion. Hypermutation rates were associated with different physiochemical binding properties of individual B-cell receptors (BCR). Increasing BCR signaling strength inhibited class switching from IgG1 to IgE in vitro, preferentially constraining IgE formation. These data indicate that antigen-binding properties of individual BCRs determine differential IgE hypermutation and IgE versus IgG1 production on the level of single B-cell clones.


Asunto(s)
Hipersensibilidad al Huevo , Ratones , Animales , Hipersensibilidad al Huevo/metabolismo , Inmunoglobulina G/metabolismo , Inmunoglobulina E/metabolismo , Receptores de Antígenos de Linfocitos B/genética , Linfocitos B , Alérgenos
4.
J Allergy Clin Immunol ; 149(2): 671-684.e9, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34186142

RESUMEN

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.


Asunto(s)
Anafilaxia/prevención & control , Inmunoglobulina E/inmunología , Agonistas Adrenérgicos beta/uso terapéutico , Animales , Quimioterapia Combinada , Antagonistas de los Receptores Histamínicos/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores
5.
J Leukoc Biol ; 111(3): 563-571, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34218441

RESUMEN

Glomerulonephritis (GN), an important pathologic feature of many renal diseases, is frequently characterized by mesangial cell proliferation. We and others have previously shown that the TAM family receptor tyrosine kinases Axl, Mer, and Tyro-3 contribute to cell survival, proliferation, migration, and clearance of apoptotic cells (ACs); that Axl contributes to GN by promoting mesangial cell proliferation; and that small molecule inhibition of Axl ameliorates nephrotoxic serum-induced GN in mice. We now show that stimulation of renal mesangial cell Axl causes a modest increase in intracellular Ca2+ and activates NF-κB, mTOR, and the mTOR-containing mTORC1 complex, which phosphorylates the ribosomal protein S6. Axl-induction of Akt activation is upstream of NF-κB and mTOR activation, which are mutually codependent. Axl-induced NF-κB activation leads to Bcl-xl up-regulation. Axl is more important than Mer at mediating AC phagocytosis by mesangial cells, but less important than Mer at mediating phagocytosis of ACs by peritoneal macrophages. Taken together, our data suggest the possibility that Axl mediates mesangial cell phagocytosis of ACs and promotes mesangial cell proliferation by activating NF-κB and mTORC1.


Asunto(s)
Glomerulonefritis , Proteínas Proto-Oncogénicas c-akt , Animales , Proliferación Celular , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , FN-kappa B , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras , Serina-Treonina Quinasas TOR , Tirosina Quinasa c-Mer , Tirosina Quinasa del Receptor Axl
6.
Front Immunol ; 12: 599296, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717078

RESUMEN

We have demonstrated that intranasal immunotherapy with allergens formulated in a nanoemulsion (NE) mucosal adjuvant suppresses Th2/IgE-mediated allergic responses and protects from allergen challenge in murine food allergy models. Protection conferred by this therapy is associated with strong suppression of allergen specific Th2 cellular immunity and increased Th1 cytokines. Here we extend these studies to examine the effect of NE-allergen immunization in mice sensitized to multiple foods. Mice were sensitized to both egg and peanut and then received NE vaccine formulated with either one or both of these allergens. The animals were then subjected to oral challenges with either egg or peanut to assess reactivity. Immunization with NE formulations containing both egg and peanut markedly reduced reactivity after oral allergen challenge with either allergen. Interestingly, mice that received the vaccine containing only peanut also had reduced reactivity to challenge with egg. Protection from oral allergen challenge was achieved despite the persistence of allergen-specific IgE and was associated with strong suppression of both Th2-polarized immune responses, alarmins and type 2 innate lymphoid cells (ILC2). NE-induced bystander suppression of reactivity required IFN-γ and the presence of an allergen in the NE vaccine. These results demonstrate that anaphylactic reactions to food allergens can be suppressed using allergen-specific immunotherapy without having to eliminate allergen-specific IgE and suggests that modulation of Th2 immunity towards one allergen may induce bystander effects that suppress reactivity to other allergens through the induction of IFN-γ and suppression of alarmins in the intestine. In addition, these data suggest that a NE vaccine for a single food allergen may lead to a global suppression of allergic responses to multiple foods.


Asunto(s)
Alarminas/genética , Alérgenos/inmunología , Hipersensibilidad a los Alimentos/etiología , Hipersensibilidad a los Alimentos/terapia , Regulación de la Expresión Génica , Vacunas/inmunología , Adyuvantes Inmunológicos , Administración Intranasal , Animales , Efecto Espectador , Citocinas/metabolismo , Desensibilización Inmunológica , Modelos Animales de Enfermedad , Inmunoglobulina E/inmunología , Inmunomodulación , Ratones , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Vacunas/administración & dosificación
7.
J Allergy Clin Immunol ; 147(5): 1838-1854.e4, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33326804

RESUMEN

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.


Asunto(s)
Anafilaxia/tratamiento farmacológico , Antialérgicos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Hipersensibilidad a los Alimentos/tratamiento farmacológico , Inmunoglobulina E/inmunología , Receptores de IgE/inmunología , Anafilaxia/inmunología , Animales , Antialérgicos/farmacología , Anticuerpos Monoclonales/farmacología , Femenino , Hipersensibilidad a los Alimentos/inmunología , Inmunoglobulina G/inmunología , Masculino , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Ratones Endogámicos BALB C , Ratones Transgénicos , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/inmunología , Receptores de IgE/genética , Quinasa Syk/inmunología
8.
Front Immunol ; 11: 596772, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33362780

RESUMEN

Food allergies are common, costly and potentially life-threatening disorders. They are driven by Th2, but inhibited by Th1 reactions. There is also evidence indicating that IL-2 agonist treatment inhibits allergic sensitization through expansion of regulatory T cells. Here, we tested the impact of an IL-2 agonist in a novel model for food allergy to hen´s egg in mice sensitized without artificial adjuvants. Prophylactic IL-2 agonist treatment expanded Treg populations and inhibited allergen-specific sensitization. However, IL-2 agonist treatment of already sensitized mice increased mast cell responses and allergic anaphylaxis upon allergen re-challenge. These effects depended on allergen-specific IgE and were mediated through IFN-γ, as shown by IgE transfer and blockade of IFN-γ with monoclonal antibodies. These results suggest that although shifting the allergic reaction toward a Treg/Th1 response inhibits allergic sensitization, the prototypic Th1 cytokine IFN-γ promotes mast cell activation and allergen-induced anaphylaxis in individuals that are already IgE-sensitized. Hence, while a Th1 response can prevent the development of food allergy, IFN-γ has the ability to exacerbate already established food allergy.


Asunto(s)
Alérgenos/inmunología , Anafilaxia/etiología , Anafilaxia/metabolismo , Alimentos/efectos adversos , Interferón gamma/metabolismo , Interleucina-2/agonistas , Animales , Pollos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Clara de Huevo/efectos adversos , Femenino , Hipersensibilidad a los Alimentos/inmunología , Inmunización , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Ratones
9.
Blood Adv ; 4(18): 4406-4416, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32931581

RESUMEN

The family of nuclear factor of activated T cells (NFAT) transcription factors plays a critical role in mediating immune responses. Our previous clinical pharmacogenetic studies suggested that NFATC2 is associated with the risk of hypersensitivity reactions to the chemotherapeutic agent L-asparaginase (ASNase) that worsen outcomes during the treatment of pediatric acute lymphoblastic leukemia. We therefore hypothesized that the genetic inhibition of NFATC2 would protect against the development of anti-ASNase antibodies and ASNase hypersensitivity. Our study demonstrates that ASNase-immunized NFATC2-deficient mice are protected against ASNase hypersensitivity and develop lower antigen-specific and total immunoglobulin E (IgE) levels compared with wild-type (WT) controls. Furthermore, ASNase-immunized NFATC2-deficient mice develop more CD4+ regulatory T cells, fewer CD4+ interleukin-4-positive (IL-4+) cells, higher IL-10/TGF-ß1 levels, and lower IL-4/IL-13 levels relative to WT mice. Basophils and peritoneal mast cells from ASNase-immunized, but not naïve, NFATC2-deficient mice had lower FcεRI expression and decreased IgE-mediated mast cell activation than WT mice. Furthermore, ASNase-immunized, but not naïve, NFATC2-deficient mice developed less severe shock than WT mice after induction of passive anaphylaxis or direct histamine administration. Thus, inhibition of NFATC2 protects against ASNase hypersensitivity by impairing T helper 2 responses, which may provide a novel strategy for attenuating hypersensitivity and the development of antidrug antibodies, including to ASNase.


Asunto(s)
Antineoplásicos , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Anticuerpos/uso terapéutico , Antineoplásicos/uso terapéutico , Asparaginasa/uso terapéutico , Ratones , Factores de Transcripción NFATC , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Factores de Transcripción
10.
Cell Immunol ; 356: 104134, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32862025

RESUMEN

Mast cells, well established effectors in allergic disease, can be activated by numerous stimuli. We previously found that the Fyn-Stat5B pathway is critical for FcεRI-stimulated mast cell function. Because IgG receptors employ similar signaling pathways, we investigated Fyn-Stat5B function downstream of FcγR. We report that FcγR elicits Fyn-dependent Stat5B tyrosine phosphorylation in mast cells. As we previously found for Fyn kinase, Stat5B is indispensable for IgG-mediated mast cell cytokine expression and secretion. However, Stat5B KO macrophages responded normally to FcγR signaling, indicating a lineage-restricted role for Stat5B. This was consistent in vivo, since passive FcγR activation induced anaphylaxis in a macrophage-dominated response even when Stat5B was deleted. We further investigated this lineage restriction using the K/BxN model of inflammatory arthritis. This model exhibits a rapid and transient mast cell-dependent joint inflammation followed days later by a macrophage- and neutrophil-dependent response. Consistent with our hypothesis, Fyn or Stat5B deficiency did not protect mice from late joint swelling, but greatly reduced the early mast cell-dependent response. This was associated with decreased joint and plasma histamine. We conclude that Fyn-Stat5B is a linage-restricted pathway critical for IgG-mediated mast cell responses.


Asunto(s)
Mastocitos/fisiología , Receptores de IgG/metabolismo , Factor de Transcripción STAT5/metabolismo , Anafilaxia/inmunología , Animales , Degranulación de la Célula/fisiología , Femenino , Humanos , Masculino , Mastocitos/citología , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores de IgE/metabolismo , Receptores de IgG/inmunología , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/inmunología , Transducción de Señal , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Familia-src Quinasas/metabolismo
11.
Arthritis Res Ther ; 22(1): 133, 2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32503684

RESUMEN

BACKGROUND: The mechanisms involved in the pathogenesis of autoimmune disorders, including systemic lupus erythematosus (SLE), have not been fully elucidated. Some of these mechanisms involve epigenetic regulation of gene expression. The histone methyltransferase Ezh2 contributes to epigenetic regulation of gene expression, is highly expressed in germinal center (GC) B cells and follicular T helper (TFH) cells, and may be involved in lupus pathogenesis. METHODS: The murine bm12 model of lupus-like chronic graft versus host disease (cGVHD) was induced by intra-peritoneal injection of negatively isolated allogeneic CD4+ T cells. Lupus-like disease development was monitored by ELISA determination of serum anti-dsDNA and anti-chromatin antibody titers. Immune cell activation and Ezh2 expression were evaluated by flow cytometry and Western blotting. RESULTS: Decreased autoantibody production and GC formation are observed when Ezh2-deficient CD4+ T cells are used instead of wild-type (WT) to induce cGVHD and when mice that receive allogeneic WT donor T cells to induce cGVHD are treated with GSK503, an Ezh2-specific inhibitor. In the bm12 cGVHD model, WT donor T cells are normally fully activated 1 week after infusion into an allogeneic host, exhibit a TFH cell (PD-1hi/CXCR5hi) phenotype with upregulated Ezh2, and activate B cells to form germinal centers (GCs). In contrast, Ezh2-deficient donor T cells generate fewer TFH cells that fail to activate B cells or promote GC formation. Despite similar T-independent, LPS-induced B cell responses, OVA-immunized CD4.Ezh2-KO mice had a skewed low-affinity IgM phenotype in comparison to similarly treated WT mice. In addition, early after OVA immunization, more CD4+ T cells from B6.CD4.Ezh2-KO mice had a CD44lo/CD62Llo phenotype, which suggests arrested or delayed activation, than CD4+ T cells from ovalbumin-immunized B6.WT mice. CONCLUSION: Ezh2 gene deletion or pharmacological Ezh2 inhibition suppresses autoantibody production and GC formation in bm12 lupus-like cGVHD and decreases affinity maturation and isotype switching in response to immunization with a T cell-dependent antigen. Ezh2 inhibition may be useful for the treatment of lupus and other autoimmune disorders.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Lupus Eritematoso Sistémico , Animales , Epigénesis Genética , Centro Germinal , Enfermedad Injerto contra Huésped/genética , Lupus Eritematoso Sistémico/genética , Ratones , Linfocitos T , Linfocitos T Colaboradores-Inductores
12.
J Allergy Clin Immunol ; 145(3): 907-921.e3, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31836406

RESUMEN

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.


Asunto(s)
Anafilaxia/inmunología , Anticuerpos Monoclonales/farmacología , Desensibilización Inmunológica/métodos , Receptores de IgE/antagonistas & inhibidores , Anafilaxia/prevención & control , Animales , Humanos , Ratones , Ratones Endogámicos BALB C
13.
Mucosal Immunol ; 13(2): 283-292, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31745261

RESUMEN

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.


Asunto(s)
Células Caliciformes/patología , Hipersensibilidad/inmunología , Músculo Liso/metabolismo , Receptores de Superficie Celular/metabolismo , Hipersensibilidad Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Animales , Antígenos Dermatofagoides/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Pyroglyphidae , Receptores de Superficie Celular/genética , Transducción de Señal
14.
FASEB J ; 33(10): 10996-11005, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31284767

RESUMEN

Asparaginase (ASNase) is an important drug for the treatment of leukemias. However, hypersensitivity to ASNase can increase the risk of leukemia relapse. Two mechanisms of ASNase hypersensitivity have been identified in mice. The existence of a pathway involving anti-ASNase IgG and Fc-γ receptor III (Fc-γRIII) implies that IgG and ASNase immune complexes (ICs) could directly induce hypersensitivity. The aim of this study was to detect ASNase ICs in mice after hypersensitivity reactions and determine their role in hypersensitivity. Protein G beads were used to detect plasma ASNase ICs by flow cytometry. Anti-ASNase IgG was purified from the plasma of sensitized mice, and ASNase ICs were prepared ex vivo at various ratios of ASNase to anti-ASNase IgG. The levels of ASNase ICs detected after hypersensitivity reactions correlated with reaction severity (R2 = 0.796; P = 0.0005). ASNase ICs prepared ex vivo required high levels of anti-ASNase IgG for formation, and binding to naive and sensitized immune cells depended on soluble anti-ASNase IgG, antigen:antibody ratio, and Fc-γRIII. Similarly, basophil activation by ASNase ICs depended on the antigen:antibody ratio and Fc-γRIII. Consistent with the ex vivo results, naive mice receiving ASNase ICs developed hypersensitivity reactions. Our data demonstrate that ASNase ICs can directly contribute to the onset and severity of ASNase hypersensitivity.-Rathod, S., Ramsey, M., DiGiorgio, D., Berrios, R., Finkelman, F. D., Fernandez, C. A. Asparaginase immune complexes induce Fc-γRIII-dependent hypersensitivity in naive mice.


Asunto(s)
Antineoplásicos/inmunología , Asparaginasa/inmunología , Hipersensibilidad a las Drogas/inmunología , Receptores de IgG/inmunología , Animales , Antineoplásicos/efectos adversos , Asparaginasa/efectos adversos , Células Cultivadas , Hipersensibilidad a las Drogas/etiología , Femenino , Ratones , Ratones Endogámicos C57BL
15.
Immunity ; 50(5): 1262-1275.e4, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31027995

RESUMEN

Mast cell (MC) mediator release after crosslinking of surface-bound IgE antibody by ingested antigen underlies food allergy. However, IgE antibodies are not uniformly associated with food allergy, and intestinal MC load is an important determinant. Atopic dermatitis (AD), characterized by pruritis and cutaneous sensitization to allergens, including foods, is strongly associated with food allergy. Tape stripping mouse skin, a surrogate for scratching, caused expansion and activation of small intestinal MCs, increased intestinal permeability, and promoted food anaphylaxis in sensitized mice. Tape stripping caused keratinocytes to systemically release interleukin-33 (IL-33), which synergized with intestinal tuft-cell-derived IL-25 to drive the expansion and activation of intestinal type-2 innate lymphoid cells (ILC2s). These provided IL-4, which targeted MCs to expand in the intestine. Duodenal MCs were expanded in AD. In addition to promoting cutaneous sensitization to foods, scratching may promote food anaphylaxis in AD by expanding and activating intestinal MCs.


Asunto(s)
Dermatitis Atópica/inmunología , Hipersensibilidad a los Alimentos/inmunología , Mucosa Intestinal/inmunología , Linfocitos/inmunología , Mastocitos/inmunología , Adolescente , Anafilaxia/inmunología , Animales , Proliferación Celular , Niño , Preescolar , Femenino , Humanos , Inmunoglobulina E/inmunología , Interleucina-13/metabolismo , Interleucina-33/metabolismo , Interleucina-4/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Piel/inmunología , Piel/lesiones
16.
EMBO J ; 38(7)2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30842097

RESUMEN

BRD4 is a BET family protein that binds acetylated histones and regulates transcription. BET/BRD4 inhibitors block blood cancer growth and inflammation and serve as a new therapeutic strategy. However, the biological role of BRD4 in normal hematopoiesis and inflammation is not fully understood. Analysis of Brd4 conditional knockout (KO) mice showed that BRD4 is required for hematopoietic stem cell expansion and progenitor development. Nevertheless, BRD4 played limited roles in macrophage development and inflammatory response to LPS ChIP-seq analysis showed that despite its limited importance, BRD4 broadly occupied the macrophage genome and participated in super-enhancer (SE) formation. Although BRD4 is critical for SE formation in cancer, BRD4 was not required for macrophage SEs, as KO macrophages created alternate, BRD4-less SEs that compensated BRD4 loss. This and additional mechanisms led to the retention of inflammatory responses in macrophages. Our results illustrate a context-dependent role of BRD4 and plasticity of epigenetic regulation.


Asunto(s)
Biomarcadores/análisis , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Inflamación/inmunología , Macrófagos Peritoneales/inmunología , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Animales , Células Cultivadas , Perfilación de la Expresión Génica , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/patología , Lipopolisacáridos/toxicidad , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Haematologica ; 104(2): 319-329, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30237274

RESUMEN

Asparaginase is an important drug for the treatment of leukemias. However, anti-asparaginase antibodies often develop, which can decrease asparaginase drug levels and increase the risk of relapse. The aim of this study is to identify the immunoglobulin isotypes and receptors responsible for asparaginase hypersensitivities. Mice immunized with asparaginase developed anti-asparaginase IgG1 and IgE antibodies, and challenging the sensitized mice with asparaginase induced severe hypersensitivity reactions. Flow cytometry analysis indicated that macrophages/monocytes, neutrophils, and basophils bind asparaginase ex vivo through FcγRIII. In contrast, asparaginase binding to basophils was dependent on FcγRIII and IgE. Consistent with the asparaginase binding data, basophil activation by asparaginase occurred via both IgG/FcγRIII and IgE/FcεRI. Depleting >95% of B cells suppressed IgG but not IgE-dependent hypersensitivity, while depleting CD4+ T cells provided complete protection. Combined treatment with either anti-IgE mAb plus a platelet-activating factor receptor antagonist or anti-FcγRIII mAb plus a H1 receptor antagonist suppressed asparaginase hypersensitivity. The observations indicate that asparaginase hypersensitivity is mediated by antigen-specific IgG and/or IgE through the immunoglobulin receptors FcγRIII and FcεRI, respectively. Provided that these results apply to humans, they emphasize the importance of monitoring both IgE- and IgG-mediated asparaginase hypersensitivities in patients receiving this agent.


Asunto(s)
Antineoplásicos/efectos adversos , Asparaginasa/efectos adversos , Hipersensibilidad a las Drogas/inmunología , Hipersensibilidad a las Drogas/metabolismo , Inmunoglobulina E/inmunología , Inmunoglobulina G/inmunología , Receptores de IgE/metabolismo , Receptores de IgG/metabolismo , Animales , Modelos Animales de Enfermedad , Hipersensibilidad a las Drogas/diagnóstico , Femenino , Inmunización , Recuento de Leucocitos , Leucocitos/inmunología , Leucocitos/metabolismo , Ratones , Unión Proteica/inmunología
18.
Front Immunol ; 9: 1392, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29973935

RESUMEN

Histamine is a critical mediator of IgE/mast cell-mediated anaphylaxis, a neurotransmitter and a regulator of gastric acid secretion. Histamine is a monoamine synthesized from the amino acid histidine through a reaction catalyzed by the enzyme histidine decarboxylase (HDC), which removes carboxyl group from histidine. Despite the importance of histamine, transcriptional regulation of HDC gene expression in mammals is still poorly understood. In this review, we focus on discussing advances in the understanding of molecular regulation of mammalian histamine synthesis.

19.
Vaccine ; 36(31): 4734-4741, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-29941326

RESUMEN

BACKGROUND: Influenza immunization is universally recommended during pregnancy to protect mothers and their offspring. However, pregnancy-induced shifts in vaccine responsiveness remain poorly defined. METHODS: Quantitative and qualitative shifts in the serological response to influenza vaccination were evaluated in healthy women throughout the course of pregnancy. Serum was obtained before and after vaccination among 71 pregnant and 67 non-pregnant women during the 2011-12 and 2012-13 influenza seasons. Serum hemagglutination inhibition (HAI) assay was used to investigate anti-influenza antibody responses by comparing pre-vaccine and post-vaccine geometric mean titers (GMTs) between groups for each antigen. IgG1, IgG2, IgG3, and IgG4 anti-influenza titers were also evaluated by enzyme-linked immunosorbent assay (ELISA). Pregnancy induced shifts in HAI titers and levels of each anti-influenza antibody isotype were evaluated using linear regression models. RESULTS: Post-vaccine GMTs at day 28 were significantly reduced for women vaccinated during pregnancy for A/California (H1N1) in 2011 (p = 0.027), A/Perth (H3N2) in 2011 (p = 0.037), and B/Wisconsin in 2012 (p = 0.039). Vaccine responses progressively declined with the initiation of vaccination later in pregnancy. Anti-H1N1 IgG1, IgG2, and IgG3 titers were reduced in pregnant women compared to non-pregnant controls, and these titers declined with pregnancy progression. The most striking differences were found for anti-H1N1 IgG1, where titers decreased by approximately 7% each week throughout pregnancy. CONCLUSIONS: HAI responses elicited by immunization were significantly reduced during pregnancy for three different influenza vaccine antigens. Anti-H1N1 IgG1 was significantly lower in pregnant women and decreased throughout the course of pregnancy. Waning serological responsiveness to influenza vaccination with the progression of human pregnancy has important translational implications for when immunization should be optimally administered during pregnancy.


Asunto(s)
Formación de Anticuerpos , Esquemas de Inmunización , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Virus de la Influenza B/inmunología , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/prevención & control , Adolescente , Adulto , Anticuerpos Antivirales/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Voluntarios Sanos , Pruebas de Inhibición de Hemaglutinación , Humanos , Inmunoglobulina G/sangre , Vacunas contra la Influenza/inmunología , Embarazo , Estudios Prospectivos , Factores de Tiempo , Adulto Joven
20.
J Autoimmun ; 93: 37-44, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29895432

RESUMEN

Glomerulonephritis (GN) is a typical lesion in autoantibody and immune complex disorders, including SLE. Because the Gas6/Axl pathway has been implicated in the pathogenesis of many types of GN, targeting this pathway might ameliorate GN. Consequently, we have studied the efficacy and mechanism of R428, a potent selective Axl inhibitor, in the prevention of experimental anti-GBM nephritis. Axl upregulation was investigated with Sp1/3 siRNA in the SV40-transformed mesangial cells. For Axl inhibition, a daily dose of R428 (125 mg/kg) or vehicle was administered orally. GN was induced with anti-GBM sera. Renal disease development was followed by serial blood urine nitrogen (BUN) determinations and by evaluation of kidney histology at the time of sacrifice. Axl-associated signaling proteins were analyzed by Western blotting and inflammatory cytokine secretion was analyzed by Proteome array. SiRNA data revealed the transcription factor Sp1 to be an important regulator of mesangial Axl expression. Anti-GBM serum induced severe nephritis with azotemia, protein casts and necrotic cell death. R428 treatment diminished renal Axl expression and improved kidney function, with significantly decreased BUN and glomerular proliferation. R428 treatment inhibited Axl and significantly decreased Akt phosphorylation and renal inflammatory cytokine and chemokine expression; similar effects were observed in anti-GBM antiserum-treated Axl-KO mice. These studies support a role for Axl inhibition in glomerulonephritis.


Asunto(s)
Benzocicloheptenos/farmacología , Factores Inmunológicos/farmacología , Nefritis Lúpica/tratamiento farmacológico , Células Mesangiales/efectos de los fármacos , Terapia Molecular Dirigida/métodos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Triazoles/farmacología , Administración Oral , Animales , Anticuerpos/administración & dosificación , Línea Celular Transformada , Esquema de Medicación , Regulación de la Expresión Génica , Membrana Basal Glomerular/efectos de los fármacos , Membrana Basal Glomerular/inmunología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/inmunología , Nefritis Lúpica/inducido químicamente , Nefritis Lúpica/genética , Nefritis Lúpica/inmunología , Células Mesangiales/inmunología , Células Mesangiales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/inmunología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/inmunología , Transducción de Señal , Factor de Transcripción Sp1/antagonistas & inhibidores , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/inmunología , Factor de Transcripción Sp3/antagonistas & inhibidores , Factor de Transcripción Sp3/genética , Factor de Transcripción Sp3/inmunología , Tirosina Quinasa del Receptor Axl
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...