Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.227
Filtrar
1.
Eur J Immunol ; 52(1): 44-53, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34606636

RESUMO

Crosslinking of FcεRI-bound IgE triggers the release of a large number of biologically active, potentially anaphylactic compounds by mast cells. FcεRI activation ought to be well-controlled to restrict adverse activation. As mast cells are embedded in tissues, adhesion molecules may contribute to limiting premature activation. Here, we report that E-Cadherin serves that purpose. Having confirmed that cultured mast cells express E-Cadherin, a mast-cell-specific E-Cadherin deficiency, Mcpt5-Cre E-Cdhfl/fl mice, was used to analyze mast cell degranulation in vitro and in vivo. Cultured peritoneal mast cells from Mcpt5-Cre E-Cdhfl/fl mice were normal with respect to many parameters but showed much-enhanced degranulation in three independent assays. Soluble E-Cadherin reduced the degranulation of control cells. The release of some newly synthesized inflammatory cytokines was decreased by E-Cadherin deficiency. Compared to controls, Mcpt5-Cre E-Cdhfl/fl mice reacted much stronger to IgE-dependent stimuli, developing anaphylactic shock. We suggest E-Cadherin-mediated tissue interactions restrict mast cell degranulation to prevent their precocious activation.


Assuntos
Caderinas/imunologia , Degranulação Celular/imunologia , Mastócitos/imunologia , Animais , Caderinas/genética , Degranulação Celular/genética , Citocinas/genética , Citocinas/imunologia , Imunoglobulina E/genética , Imunoglobulina E/imunologia , Inflamação/genética , Inflamação/imunologia , Camundongos , Camundongos Transgênicos , Receptores de IgE/genética , Receptores de IgE/imunologia
2.
J Allergy Clin Immunol ; 149(1): 156-167.e7, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34051221

RESUMO

BACKGROUND: Binding IgE to a cognate allergen causes aggregation of Fcε receptor I (FcεRI) in mast cells, resulting in activation of receptor-associated Src family tyrosine kinases, including Lyn and Syk. Protein tyrosine phosphatase, receptor type C (PTPRC), also known as CD45, has emerged as a positive regulator of FcεRI signaling by dephosphorylation of the inhibitory tyrosine of Lyn. OBJECTIVE: Sirtuin 6 (Sirt6), a NAD+-dependent deacetylase, exhibits an anti-inflammatory property. It remains to be determined, however, whether Sirt6 attenuates mast cell-associated diseases, including anaphylaxis. METHODS: FcεRI signaling and mast cell degranulation were measured after IgE cross-linking in murine bone marrow-derived mast cells (BMMCs) and human cord blood-derived mast cells. To investigate the function of Sirt6 in mast cell activation in vivo, we used mast cell-dependent animal models of passive systemic anaphylaxis (PSA) and passive cutaneous anaphylaxis (PCA). RESULTS: Sirt6-deficient BMMCs augmented IgE-FcεRI-mediated signaling and degranulation compared to wild-type BMMCs. Reconstitution of mast cell-deficient KitW-sh/W-sh mice with BMMCs received from Sirt6 knockout mice developed more severe PSA and PCA compared to mice engrafted with wild-type BMMCs. Similarly, genetic overexpression or pharmacologic activation of Sirt6 suppressed mast cell degranulation and blunted responses to PCA. Mechanistically, Sirt6 deficiency increased PTPRC transcription via acetylating histone H3, leading to enhanced aggregation of FcεRI in BMMCs. Finally, we recapitulated the Sirt6 regulation of PTPRC and FcεRI signaling in human mast cells. CONCLUSIONS: Sirt6 acts as a negative regulator of FcεRI signaling cascade in mast cells by suppressing PTPRC transcription. Activation of Sirt6 may therefore represent a promising and novel therapeutic strategy for anaphylaxis.


Assuntos
Anafilaxia/imunologia , Mastócitos/imunologia , Receptores de IgE/imunologia , Sirtuínas/imunologia , Animais , Células da Medula Óssea/citologia , Sangue Fetal/citologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Sirtuínas/genética
3.
Allergol Int ; 71(1): 94-102, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34465532

RESUMO

BACKGROUND: Autoantibodies (AAbs) against immunoglobulin E (IgE) antibodies (Abs) and their high-affinity receptor alpha subunits (FcεRIα) are key factors in the elicitation of type IIb autoimmune chronic spontaneous urticaria (type IIb aiCSU). In this study, we aimed to develop a new method to detect functional anti-FcεRIα and anti-IgE AAbs, which can crosslink the plural FcεRІα molecules and IgE Abs on the surface of mast cells and basophils, in sera from aiCSU patients using the amplified luminescence proximity homogeneous assay (Alpha). METHODS: Sera were obtained from 14 aiCSU patients, as diagnosed by recurrent chronic spontaneous urticaria episodes and positive results for the autologous serum skin test and/or histamine release test (HRT). The AAbs to FcεRIα and IgE Abs were determined in sera from aiCSU patients using enzyme-linked immunosorbent assay (ELISA) and Alpha by cross-linking (AlphaCL) of IgE Abs and/or FcεRІα. RESULTS: Serum anti-FcεRIα and anti-IgE AAb levels were not significantly different between aiCSU patients and healthy subjects in ELISA. Anti-FcεRIα AAbs were detected in 10 of 14 aiCSU patients who displayed positive (5/5) and negative (5/9) results in the HRT for anti-FcεRIα AAbs by AlphaCL, whereas no signals were observed in healthy subjects. Additionally, anti-IgE AAbs were detected in two of four aiCSU patients who displayed positive results in the HRT for anti-IgE AAbs. CONCLUSIONS: A new assay method using AlphaCL can detect anti-FcεRIα and anti-IgE AAbs with FcεRIα- and IgE-crosslinking abilities in sera from aiCSU patients. This simple and practical assay method may be available as a diagnostic tool for urticaria patients.


Assuntos
Autoanticorpos/imunologia , Urticária Crônica/sangue , Receptores de IgE/imunologia , Adulto , Idoso , Estudos de Casos e Controles , Ensaio de Imunoadsorção Enzimática , Feminino , Liberação de Histamina , Humanos , Masculino , Pessoa de Meia-Idade , Receptores de IgE/antagonistas & inibidores , Receptores de IgE/sangue , Pele/química , Testes Cutâneos
4.
Yakugaku Zasshi ; 141(9): 1057-1061, 2021.
Artigo em Japonês | MEDLINE | ID: mdl-34471006

RESUMO

Mast cells (MCs) are immune cells that are distributed in all tissues throughout the body, and their cytoplasm is rich in granules containing histamine and tryptase. When MCs recognize antigens through IgE bound to FcεRI, they release these mediators by degranulation. Because degranulation induces various type I allergic reactions, such as anaphylactic shock and hay fever, elucidation of the control mechanism of degranulation is important to the development of a therapeutic strategy for allergic diseases. It is known that the antigen-induced degranulation response is fine-tuned by various humoral factors via the activation of G protein-coupled receptors. We found that extracellular ATP enhanced antigen-dependent and -independent MC degranulation via activation of ionotropic P2X4 receptors. P2X4 receptor activation itself had no effect on MC degranulation, but significantly enhanced antigen-triggered degranulation. Stimulation of the P2X4 receptor potentiated the FcεRI-mediated tyrosine kinase signaling cascade. In addition to antigen-induced responses, P2X4 receptor signaling also affected antigen-independent MC responses. Thus, co-stimulation of ATP and Gi-coupled receptor agonists, such as prostaglandin E2 (PGE2) and adenosine, resulted in synergistic degranulation. The significance of P2X4 receptor signaling in allergic and inflammatory responses in vivo was confirmed by impaired responses of antigen-induced passive anaphylaxis and PGE2-induced increases in vascular permeability in P2rx4 knockout mice compared to that of wild-type mice. These results suggest that the P2X4 receptor is a potential therapeutic target for both antigen-dependent and -independent allergic reactions.


Assuntos
Mastócitos/imunologia , Receptores Purinérgicos P2X4/imunologia , Receptores Purinérgicos P2X4/metabolismo , Transdução de Sinais/imunologia , Animais , Degranulação Celular/imunologia , Grânulos Citoplasmáticos/metabolismo , Histamina/metabolismo , Humanos , Hipersensibilidade/imunologia , Hipersensibilidade/terapia , Imunoglobulina E/imunologia , Mastócitos/citologia , Mastócitos/metabolismo , Camundongos Knockout , Terapia de Alvo Molecular , Receptores de IgE/imunologia , Triptases/metabolismo
7.
J Allergy Clin Immunol ; 148(4): 1049-1060, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33991582

RESUMO

BACKGROUND: Anaphylaxis represents one of the most severe and fatal forms of allergic reactions. Like most other allergies, it is caused by activation of basophils and mast cells by allergen-mediated cross-linking of IgE bound to its high-affinity receptor, FcεRI, on the cell surface. The systemic release of soluble mediators induces an inflammatory cascade, rapidly causing symptoms with peak severity in minutes to hours after allergen exposure. Primary treatment for anaphylaxis consists of immediate intramuscular administration of adrenaline. OBJECTIVE: While adrenaline alleviates life-threatening symptoms of an anaphylactic reaction, there are currently no disease-modifying interventions available. We sought to develop potent and fast-acting IgE inhibitors with the potential to rapidly terminate acute allergic reactions. METHODS: Using affinity maturation by yeast display and structure-guided molecular engineering, we generated 3 optimized disruptive IgE inhibitors based on designed ankyrin repeat proteins and assessed their ability to actively remove IgE from allergic effector cells in vitro as well as in vivo in mice. RESULTS: The engineered IgE inhibitors rapidly dissociate preformed IgE:FcεRI complexes, terminate IgE-mediated signaling in preactivated human blood basophils in vitro, and shut down preinitiated allergic reactions and anaphylaxis in mice in vivo. CONCLUSIONS: Fast-acting disruptive IgE inhibitors demonstrate the feasibility of developing kinetically optimized inhibitors for the treatment of anaphylaxis and the rapid desensitization of allergic individuals.


Assuntos
Anafilaxia/tratamento farmacológico , Imunoglobulina E/imunologia , Proteínas Recombinantes de Fusão , Alérgenos/imunologia , Anafilaxia/imunologia , Animais , Basófilos/efeitos dos fármacos , Basófilos/imunologia , Desenho de Fármacos , Humanos , Imunoglobulina E/química , Imunoglobulina E/genética , Camundongos Transgênicos , Estrutura Molecular , Ovalbumina/imunologia , Receptores de IgE/química , Receptores de IgE/genética , Receptores de IgE/imunologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico
8.
J Exp Med ; 218(7)2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-33944900

RESUMO

Food allergies are a leading cause of anaphylaxis, and cellular mechanisms involving antigen presentation likely play key roles in their pathogenesis. However, little is known about the response of specific antigen-presenting cell (APC) subsets to food allergens in the setting of food allergies. Here, we show that in peanut-allergic humans, peanut allergen drives the differentiation of CD209+ monocyte-derived dendritic cells (DCs) and CD23+ (FcєRII) myeloid dendritic cells through the action of allergen-specific CD4+ T cells. CD209+ DCs act reciprocally on the same peanut-specific CD4+ T cell population to reinforce Th2 cytokine expression in a positive feedback loop, which may explain the persistence of established food allergy. In support of this novel model, we show clinically that the initiation of oral immunotherapy (OIT) in peanut-allergic patients is associated with a decrease in CD209+ DCs, suggesting that breaking the cycle of positive feedback is associated with therapeutic effect.


Assuntos
Alérgenos/imunologia , Arachis/imunologia , Imunidade/imunologia , Hipersensibilidade a Amendoim/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Moléculas de Adesão Celular/imunologia , Citocinas/imunologia , Células Dendríticas/imunologia , Retroalimentação , Humanos , Imunoterapia/métodos , Lectinas Tipo C/imunologia , Camundongos , Monócitos/imunologia , Receptores de Superfície Celular/imunologia , Receptores de IgE/imunologia , Linfócitos T Reguladores/imunologia , Células Th2/imunologia
9.
Int J Mol Sci ; 22(7)2021 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-33808264

RESUMO

The IL-1 family cytokine IL-33 activates and re-shapes mast cells (MCs), but whether and by what mechanisms it elicits cytokines in MCs from human skin remains poorly understood. The current study found that IL-33 activates CCL1, CCL2, IL-5, IL-8, IL-13, and TNF-α, while IL-1ß, IL-6, IL-31, and VEGFA remain unaffected in cutaneous MCs, highlighting that each MC subset responds to IL-33 with a unique cytokine profile. Mechanistically, IL-33 induced the rapid (1-2 min) and durable (2 h) phosphorylation of p38, whereas the phosphorylation of JNK was weaker and more transient. Moreover, the NF-κB pathway was potently activated, as revealed by IκB degradation, increased nuclear abundance of p50/p65, and vigorous phosphorylation of p65. The activation of NF-κB occurred independently of p38 or JNK. The induced transcription of the cytokines selected for further study (CCL1, CCL2, IL-8, TNF-α) was abolished by interference with NF-κB, while p38/JNK had only some cytokine-selective effects. Surprisingly, at the level of the secreted protein products, p38 was nearly as effective as NF-κB for all entities, suggesting post-transcriptional involvement. IL-33 did not only instruct skin MCs to produce selected cytokines, but it also efficiently co-operated with the allergic and pseudo-allergic/neurogenic activation networks in the production of IL-8, TNF-α, CCL1, and CCL2. Synergism was more pronounced at the protein than at the mRNA level and appeared stronger for MRGPRX2 ligands than for FcεRI. Our results underscore the pro-inflammatory nature of an acute IL-33 stimulus and imply that especially in combination with allergens or MRGPRX2 agonists, IL-33 will efficiently amplify skin inflammation and thereby aggravate inflammatory dermatoses.


Assuntos
Citocinas/imunologia , Interleucina-33/farmacologia , Mastócitos/efeitos dos fármacos , Proteínas do Tecido Nervoso/imunologia , Receptores Acoplados a Proteínas G/imunologia , Receptores de IgE/imunologia , Receptores de Neuropeptídeos/imunologia , Pele/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Prepúcio do Pênis/citologia , Humanos , Interleucina-33/imunologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Mastócitos/citologia , Mastócitos/imunologia , Fosforilação , Cultura Primária de Células , Transdução de Sinais/efeitos dos fármacos , Pele/imunologia
10.
J Allergy Clin Immunol ; 147(4): 1123-1131, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33832694

RESUMO

Anaphylaxis is a rapidly evolving, acute, life-threatening reaction that occurs rapidly on contact with a trigger. Anaphylaxis is classically defined as an allergen-driven process that induces specific IgE and the activation of mast cells and basophils through the cross-linking of IgE receptors. However, it is clear that non-IgE-mediated pathways can induce symptoms indistinguishable from those of classic anaphylaxis, and their activation could explain the severity of IgE-mediated anaphylaxis. Indeed, mast cells and basophils can be activated by antibodies against IgE or their receptors, by molecules such as anaphylatoxins, or through G-coupled receptors. Some other allergens can induce antibodies of class IgG that can activate neutrophils to produce a molecule similar to histamine to induce anaphylaxis. Finally, some inflammatory mediators such as bradykinin or prostaglandin can also modulate mast cell and basophil activation as well as directly cause vasodilation and bronchoconstriction, resulting in anaphylaxis-like reactions.


Assuntos
Anafilatoxinas/metabolismo , Anafilaxia/imunologia , Basófilos/imunologia , Hipersensibilidade/imunologia , Mastócitos/imunologia , Neutrófilos/imunologia , Alérgenos/imunologia , Animais , Broncoconstrição , Degranulação Celular , Histamina/metabolismo , Humanos , Imunoglobulina E/imunologia , Imunoglobulina E/metabolismo , Receptores de IgE/imunologia , Receptores de IgE/metabolismo , Vasodilatação
11.
Cell Rep ; 35(2): 108974, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33852838

RESUMO

Clinical definitions of asthma fail to capture the heterogeneity of immune dysfunction in severe, treatment-refractory disease. Applying mass cytometry and machine learning to bronchoalveolar lavage (BAL) cells, we find that corticosteroid-resistant asthma patients cluster largely into two groups: one enriched in interleukin (IL)-4+ innate immune cells and another dominated by interferon (IFN)-γ+ T cells, including tissue-resident memory cells. In contrast, BAL cells of a healthier population are enriched in IL-10+ macrophages. To better understand cellular mediators of severe asthma, we developed the Immune Cell Linkage through Exploratory Matrices (ICLite) algorithm to perform deconvolution of bulk RNA sequencing of mixed-cell populations. Signatures of mitosis and IL-7 signaling in CD206-FcεRI+CD127+IL-4+ innate cells in one patient group, contrasting with adaptive immune response in T cells in the other, are preserved across technologies. Transcriptional signatures uncovered by ICLite identify T-cell-high and T-cell-poor severe asthma patients in an independent cohort, suggesting broad applicability of our findings.


Assuntos
Asma/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Aprendizado de Máquina , Macrófagos/imunologia , Imunidade Adaptativa , Corticosteroides/uso terapêutico , Antiasmáticos/uso terapêutico , Asma/tratamento farmacológico , Asma/genética , Asma/patologia , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/patologia , Estudos de Casos e Controles , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Imunidade Inata , Memória Imunológica , Interferon gama/genética , Interferon gama/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-7/genética , Interleucina-7/imunologia , Macrófagos/patologia , Proteômica/métodos , Receptores de IgE/genética , Receptores de IgE/imunologia , Índice de Gravidade de Doença , Transdução de Sinais
12.
Allergol Int ; 70(3): 327-334, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33674191

RESUMO

BACKGROUND: Although basophils are considered to play an important role for maintenance of type 2 inflammation in atopic dermatitis (AD), studies on basophils in AD patients are limited. Some studies have reported the activation status, including CD203c and CD63, of peripheral blood basophils in AD patients. METHODS: We examined the features of circulating basophils in AD patients, assessed cell surface marker expressions and total serum IgE, and compared basophil responsiveness to stimulation between AD patients and healthy controls (HCs). In addition, the correlations among AD severity, laboratory factors, and features of basophils were examined. Blood samples from 38 AD patients and 21 HCs were analyzed. Basophil response markers CD203c and CD63, and expression of surface-bound IgE and FcεRI on basophils were measured. CD203c and CD63 expressions induced by stimulation with anti-IgE and anti-FcεRI antibodies were measured. Clinical/laboratory factors including total serum IgE were examined for correlations with these basophil parameters. RESULTS: Baseline CD203c and CD63 expression on basophils were significantly higher in AD patients compared with HCs. The CD203c/CD63 response ratio to anti-FcεRI stimulation was higher than that to anti-IgE stimulation in AD patients, but not HCs. FcεRI expression on basophils was higher in AD patients than in HCs, although surface-bound IgE on basophils was equivalent. Total serum IgE had negative correlations with surface-bound IgE and CD63 responsiveness to anti-IgE stimulation. CONCLUSIONS: Basophils were spontaneously activated under steady-state conditions in AD patients and responsiveness to anti-IgE stimulation was lower than in HCs. Despite high serum IgE and high basophil FcεRI expression, surface-bound IgE on basophils remained relatively low. Basophils might be suppressed or exhausted regarding FcεRI signaling via IgE in severe AD.


Assuntos
Basófilos/imunologia , Dermatite Atópica/imunologia , Imunoglobulina E/imunologia , Receptores de IgE/imunologia , Adulto , Basófilos/metabolismo , Estudos de Casos e Controles , Feminino , Humanos , Imunoglobulina E/metabolismo , Masculino , Pessoa de Meia-Idade , Diester Fosfórico Hidrolases/metabolismo , Pirofosfatases/metabolismo , Receptores de IgE/metabolismo , Tetraspanina 30/metabolismo , Adulto Jovem
13.
Immunobiology ; 226(2): 152056, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33535092

RESUMO

Mast cells, which express the high-affinity IgE receptor (FcεRI) on their surface, play a crucial role in inducing allergic inflammation. Since mast cells are activated by crosslinking of FcεRI with IgE and allergens, the cell surface expression level of FcεRI is an important factor in determining the sensitivity to allergens. Recently, the involvement of gut microbiota in the prevalence and regulation of allergy has attracted attention but the precise underlying mechanisms are not fully understood. In this study, the effect of intestinal bacteria on cell surface expression of FcεRI was examined. Bacteroides acidifaciens type A 43 specifically suppressed cell surface expression of FcεRI on mouse bone marrow-derived mast cells (BMMCs) without reduction in FcεRI α and ß-chain mRNA and total protein expression. The suppressive effect required sustained exposure to this bacterium, with a corresponding reduction in Erk activation. Inhibition of Erk decreased cell surface distribution of FcεRI in BMMCs, at least in part, through facilitated endocytosis of FcεRI. These results indicate that B. acidifaciens type A 43 suppresses cell surface expression of FcεRI on mast cells in a post-translational manner via inhibition of Erk. The suppression of FcεRI expression on mast cells by specific bacteria might be the underlying mechanism involved in the regulation of allergy by gut microbiota.


Assuntos
Bacteroides , Mastócitos/imunologia , Receptores de IgE/imunologia , Animais , Células Cultivadas , Feminino , Microbioma Gastrointestinal , Intestinos/microbiologia , Camundongos Endogâmicos C57BL , Processamento de Proteína Pós-Traducional , Receptores de IgE/genética
14.
Eur J Immunol ; 51(3): 531-543, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33527384

RESUMO

Food allergy is becoming a major public health issue, with no regulatory approved therapy to date. Food allergy symptoms range from skin rash and gastrointestinal symptoms to anaphylaxis, a potentially fatal systemic allergic shock reaction. IgE antibodies are thought to contribute importantly to key features of food allergy and anaphylaxis, and measurement of allergen-specific IgE is fundamental in diagnosing food allergy. This review will discuss recent advances in the regulation of IgE production and IgE repertoires in food allergy. We will describe the current understanding of the role of IgE and its high-affinity receptor FcεRI in food allergy and anaphylaxis, by reviewing insights gained from analyses of mouse models. Finally, we will review data derived from clinical studies of the effect of anti-IgE therapeutic monoclonal antibodies (mAbs) in food allergy, and recent insight on the efficiency and mechanisms through which these mAbs block IgE effector functions.


Assuntos
Hipersensibilidade Alimentar/imunologia , Imunoglobulina E/imunologia , Anafilaxia/imunologia , Animais , Anticorpos Monoclonais/imunologia , Humanos , Receptores de IgE/imunologia
15.
J Allergy Clin Immunol ; 147(5): 1878-1891, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33465368

RESUMO

BACKGROUND: Mast cells (MCs) are key regulators of IgE-mediated allergic inflammation. Cell-derived extracellular vesicles (EVs) contain bioactive compounds such as microRNAs. EVs can transfer signals to recipient cells, thus using a novel mechanism of cell-to-cell communication. However, whether MC-derived EVs are involved in FcεRI-mediated allergic inflammation is unclear. OBJECTIVE: We sought to investigate the effect of EVs derived from FcεRI-aggregated human MCs on the function of human group 2 innate lymphoid cells (ILC2s). METHODS: Human cultured MCs were sensitized with and without IgE for 1 hour and then incubated with anti-IgE antibody, IL-33, or medium alone for 24 hours. EVs in the MC supernatant were isolated by using ExoQuick-TC. RESULTS: Coculture of ILC2s with EVs derived from the FcεRI-aggregated MCs significantly enhanced IL-5 production and sustained upregulation of IL-5 mRNA expression in IL-33-stimulated ILC2s, but IL-13 production and IL-13 mRNA expression were unchanged. miR103a-3p expression was upregulated in IL-33-stimulated ILC2s that had been cocultured with EVs derived from anti-IgE antibody-stimulated MCs. Transduction of an miR103a-3p mimic to ILC2s significantly enhanced IL-5 production by IL-33-stimulated ILC2s. miR103a-3p promoted demethylation of an arginine residue of GATA3 by downregulating protein arginine methyltransferase 5 (PRMT5) mRNA. Reduction of protein arginine methyltransferase 5 expression in ILC2s by using a small interfering RNA technique resulted in upregulation of IL-5 production by IL-33-stimulated ILC2s. Furthermore, the level of miR103a-3p expression was significantly higher in EVs from sera of patients with atopic dermatitis than in EVs from nonatopic healthy control subjects. CONCLUSION: Eosinophilic allergic inflammation may be exacerbated owing to ILC2 activation by MC-derived miR103a-3p.


Assuntos
Citocinas/imunologia , Vesículas Extracelulares/imunologia , Linfócitos/imunologia , Mastócitos/imunologia , MicroRNAs/imunologia , Receptores de IgE/imunologia , Adulto , Idoso , Células Cultivadas , Dermatite Atópica/imunologia , Eosinófilos/imunologia , Feminino , Humanos , Imunidade Inata , Masculino , Pessoa de Meia-Idade , Adulto Jovem
16.
Int Arch Allergy Immunol ; 182(3): 190-194, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33032294

RESUMO

BACKGROUND: Subcutaneous immunotherapy (SCIT) is used to treat Japanese cedar (JC) pollinosis. The formation of IgE-allergen-CD23 complex after SCIT for JC pollinosis has not yet been fully elucidated. OBJECTIVE: The objective of this study was to investigate the formation of IgE-allergen-CD23 complex after SCIT for JC pollinosis. METHODS: Eleven patients were treated with 3-year SCIT for JC pollinosis at Sa-gamihara National Hospital from 2013 to 2014. Nasal and ocular symptoms (in terms of symptom scores) during the scattering of JC pollen and immunological changes were investigated. Levels of JC pollen-specific antibodies (IgE and IgG4) were measured by ImmunoCAP assays. To detect the changes in allergen-presenting ability of B cells, the levels of IgE-allergen-CD23 complexes in serum were measured by a cell-free, enzyme-linked immunosorbent-facilitated antigen-binding assay. RESULTS: The median (interquartile range) age of the subjects was 8 (6-10) years. Three patients (27%) had comorbid atopic dermatitis, and 5 patients (45%) had comorbid bronchial asthma. Before starting SCIT, the total IgE level was 373 (75-2,870) kU/L, and the level of JC pollen-specific IgE was 77.2 (15.4-528) kUA/L. Symptom scores improved significantly from the year after treatment. JC pollen-specific IgE levels did not change after 3 years of treatment. JC pollen-specific IgG4 levels increased significantly throughout the treatment period. The levels of IgE-allergen-CD23 complexes decreased significantly after 3 years of treatment. CONCLUSION: The ability of IgE-allergen complexes to bind to CD23 decreased after SCIT, suggesting that increasing levels of IgE-blocking antibodies, including IgG4, may play an important role in the mechanism of SCIT.


Assuntos
Alérgenos/imunologia , Complexo Antígeno-Anticorpo/imunologia , Dessensibilização Imunológica , Imunoglobulina E/imunologia , Receptores de IgE/imunologia , Rinite Alérgica Sazonal/imunologia , Rinite Alérgica Sazonal/terapia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Criança , Pré-Escolar , Cryptomeria/imunologia , Dessensibilização Imunológica/métodos , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoglobulina E/sangue , Pólen/imunologia , Rinite Alérgica Sazonal/diagnóstico , Resultado do Tratamento
17.
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
18.
Allergol Int ; 70(3): 360-367, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33279402

RESUMO

BACKGROUND: Propolis is a resinous mixture produced by honey bees that contains cinnamic acid derivatives and flavonoids. Although propolis has been reported to inhibit mast cell functions and mast cell-dependent allergic responses, the effect of propolis on basophil biology remains unknown. This study aimed to investigate the inhibitory effect of propolis on FcεRI-mediated basophil activation. METHODS: To determine the inhibitory effect of propolis on basophil activation in vitro, cytokine production and FcεRI signal transduction were analyzed by ELISA and western blotting, respectively. To investigate the inhibitory effect of propolis in vivo, IgE-CAI and a food allergy mouse model were employed. RESULTS: Propolis treatment resulted in the suppression of IgE/antigen-induced production of IL-4, IL-6 and IL-13 in basophils. Phosphorylation of FcεRI signaling molecules Lyn, Akt and ERK was inhibited in basophils treated with propolis. While propolis did not affect the basophil population in the treated mice, propolis did inhibit IgE-CAI. Finally, ovalbumin-induced intestinal anaphylaxis, which involves basophils and basophil-derived IL-4, was attenuated in mice prophylactically treated with propolis. CONCLUSIONS: Taken together, these results demonstrate the ability of propolis to suppress IgE-dependent basophil activation and basophil-dependent allergic inflammation. Therefore, prophylactic treatment with propolis may be useful for protection against food allergic reactions in sensitive individuals.


Assuntos
Basófilos/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Hipersensibilidade Alimentar/imunologia , Imunoglobulina E/efeitos dos fármacos , Inflamação/imunologia , Intestinos/efeitos dos fármacos , Própole/farmacologia , Pele/efeitos dos fármacos , Anafilaxia/imunologia , Animais , Basófilos/imunologia , Citocinas/imunologia , Imunoglobulina E/imunologia , Técnicas In Vitro , Interleucina-13/imunologia , Interleucina-4/imunologia , Interleucina-6/imunologia , Intestinos/imunologia , Camundongos , Receptores de IgE/imunologia , Pele/imunologia
20.
Blood ; 137(9): 1166-1180, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33150420

RESUMO

The terminal differentiation of B cells into antibody-secreting cells (ASCs) is a critical component of adaptive immune responses. However, it is a very sensitive process, and dysfunctions lead to a variety of lymphoproliferative neoplasias including germinal center-derived lymphomas. To better characterize the late genomic events that drive the ASC differentiation of human primary naive B cells, we used our in vitro differentiation system and a combination of RNA sequencing and Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC sequencing). We discovered 2 mechanisms that drive human terminal B-cell differentiation. First, after an initial response to interleukin-4 (IL-4), cells that were committed to an ASC fate downregulated the CD23 marker and IL-4 signaling, whereas cells that maintained IL-4 signaling did not differentiate. Second, human CD23- cells also increased IRF4 protein to levels required for ASC differentiation, but they did that independently of the ubiquitin-mediated degradation process previously described in mice. Finally, we showed that CD23- cells carried the imprint of their previous activated B-cell status, were precursors of plasmablasts, and had a phenotype similar to that of in vivo preplasmablasts. Altogether, our results provide an unprecedented genomic characterization of the fate decision between activated B cells and plasmablasts, which provides new insights into the pathological mechanisms that drive lymphoma biology.


Assuntos
Linfócitos B/imunologia , Fatores Reguladores de Interferon/imunologia , Interleucina-4/imunologia , Plasmócitos/imunologia , Receptores de IgE/imunologia , Fator de Transcrição STAT6/imunologia , Células Cultivadas , Humanos , Ativação Linfocitária , Linfoma/imunologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...