Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pediatr Allergy Immunol ; 32(7): 1508-1518, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34057765

RESUMO

BACKGROUND: Immediate food-allergic reactions are IgE-mediated, but many individuals with detectable allergen-specific IgE do not react to the food. Allergen-specific IgG may interfere with allergen-IgE interaction and/or through intracellular inhibitory signalling to suppress mast cell and basophil response to food allergens. We aimed to understand the role of allergen-specific IgG in food allergy and natural tolerance. METHODS: IgG and IgG isotypes specific to peanut, cow's milk and egg were measured using ImmunoCAP and ELISA respectively in samples of children with suspected food allergies. Expression of IgE and IgG and their receptors and expression of activation markers following allergen stimulation were measured on basophils and mast cells by flow cytometry, with and without blockade of FcγRIIα or FcγRIIß receptors. RESULTS: The levels of peanut-specific IgG, IgG1, IgG2, IgG3 and IgG4 in ELISA were higher in peanut-allergic than in non-peanut-allergic children. No difference in allergen-specific IgG isotypes was observed between allergic and non-allergic children to milk or egg, except for milk-specific IgG4 that was higher in non-cow's milk-allergic than in cow's milk-allergic children. Basophils and LAD2 cells expressed IgG receptors, but IgG and IgA were not detected on the surface of either cell type and blocking FcγRIIα or FcγRIIß did not modify basophil or mast cell activation in response to allergen in allergic or tolerant children. CONCLUSION: Allergen-specific IgG patterns were distinct in persistent (peanut) versus transient (milk and egg) food allergies. We found no evidence that FcγRIIα or FcγRIIß receptors affect allergen-induced activation of mast cells and basophils in food allergy or natural tolerance.


Assuntos
Hipersensibilidade Alimentar , Hipersensibilidade a Leite , Alérgenos , Animais , Basófilos , Bovinos , Feminino , Humanos , Imunoglobulina E , Imunoglobulina G
3.
Am J Respir Crit Care Med ; 193(6): 642-51, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26517304

RESUMO

RATIONALE: Patients with chronic obstructive pulmonary disease (COPD) are susceptible to respiratory viral infections that cause exacerbations. The mechanisms underlying this susceptibility are not understood. Effectors of the adaptive immune response-CD8(+) T cells that clear viral infections-are present in increased numbers in the lungs of patients with COPD, but they fail to protect against infection and may contribute to the immunopathology of the disease. OBJECTIVES: CD8(+) function and signaling through the programmed cell death protein (PD)-1 exhaustion pathway were investigated as a potential key mechanism of viral exacerbation of the COPD lung. METHODS: Tissue from control subjects and patients with COPD undergoing lung resection was infected with live influenza virus ex vivo. Viral infection and expression of lung cell markers were analyzed using flow cytometry. MEASUREMENTS AND MAIN RESULTS: The proportion of lung CD8(+) T cells expressing PD-1 was greater in COPD (mean, 16.2%) than in controls (4.4%, P = 0.029). Only epithelial cells and macrophages were infected with influenza, and there was no difference in the proportion of infected cells between controls and COPD. Infection up-regulated T-cell PD-1 expression in control and COPD samples. Concurrently, influenza significantly up-regulated the marker of cytotoxic degranulation (CD107a) on CD8(+) T cells (P = 0.03) from control subjects but not on those from patients with COPD. Virus-induced expression of the ligand PD-L1 was decreased on COPD macrophages (P = 0.04) with a corresponding increase in IFN-γ release from infected COPD explants compared with controls (P = 0.04). CONCLUSIONS: This study has established a signal of cytotoxic immune dysfunction and aberrant immune regulation in the COPD lung that may explain both the susceptibility to viral infection and the excessive inflammation associated with exacerbations.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Influenza Humana/imunologia , Pulmão/imunologia , Receptor de Morte Celular Programada 1/imunologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Idoso , Feminino , Citometria de Fluxo , Humanos , Influenza Humana/complicações , Masculino , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/complicações , Transdução de Sinais/imunologia , Regulação para Cima/imunologia
4.
PLoS One ; 10(3): e0121527, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25775126

RESUMO

Lung macrophages are an important defence against respiratory viral infection and recent work has demonstrated that influenza-induced macrophage PDL1 expression in the murine lung leads to rapid modulation of CD8+ T cell responses via the PD1 receptor. This PD1/PDL1 pathway may downregulate acute inflammatory responses to prevent tissue damage. The aim of this study was to investigate the mechanisms of PDL1 regulation by human macrophages in response to viral infection. Ex-vivo viral infection models using influenza and RSV were established in human lung explants, isolated lung macrophages and monocyte-derived macrophages (MDM) and analysed by flow cytometry and RT-PCR. Incubation of lung explants, lung macrophages and MDM with X31 resulted in mean cellular infection rates of 18%, 18% and 29% respectively. Viral infection significantly increased cell surface expression of PDL1 on explant macrophages, lung macrophages and MDM but not explant epithelial cells. Infected MDM induced IFNγ release from autologous CD8+ T cells, an effect enhanced by PDL1 blockade. We observed increases in PDL1 mRNA and IFNß mRNA and protein release by MDM in response to influenza infection. Knockdown of IFNß by siRNA, resulted in a 37.5% reduction in IFNß gene expression in response to infection, and a significant decrease in PDL1 mRNA. Furthermore, when MDM were incubated with IFNß, this cytokine caused increased expression of PDL1 mRNA. These data indicate that human macrophage PDL1 expression modulates CD8+ cell IFNγ release in response to virus and that this expression is regulated by autologous IFNß production.


Assuntos
Antígeno B7-H1/genética , Expressão Gênica , Interferon beta/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virologia , Antígeno B7-H1/metabolismo , Citocinas/genética , Citocinas/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Vírus da Influenza A , Macrófagos/metabolismo , Macrófagos/virologia , Monócitos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...