Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 314(3): L514-L527, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29192094

RESUMO

Human rhinovirus (HRV) is the most common virus contributing to acute exacerbations of chronic obstructive pulmonary disease (COPD) nearly year round, but the mechanisms have not been well elucidated. Recent clinical studies suggest that high levels of growth differentiation factor 15 (GDF15) protein in the blood are associated with an increased yearly rate of all-cause COPD exacerbations. Therefore, in the current study, we investigated whether GDF15 promotes HRV infection and virus-induced lung inflammation. We first examined the role of GDF15 in regulating host defense and HRV-induced inflammation using human GDF15 transgenic mice and cultured human GDF15 transgenic mouse tracheal epithelial cells. Next, we determined the effect of GDF15 on viral replication, antiviral responses, and inflammation in human airway epithelial cells with GDF15 knockdown and HRV infection. Finally, we explored the signaling pathways involved in airway epithelial responses to HRV infection in the context of GDF15. Human GDF15 protein overexpression in mice led to exaggerated inflammatory responses to HRV, increased infectious particle release, and decreased IFN-λ2/3 (IL-28A/B) mRNA expression in the lung. Moreover, GDF15 facilitated HRV replication and inflammation via inhibiting IFN-λ1/IL-29 protein production in human airway epithelial cells. Lastly, Smad1 cooperated with interferon regulatory factor 7 (IRF7) to regulate airway epithelial responses to HRV infection partly via GDF15 signaling. Our results reveal a novel function of GDF15 in promoting lung HRV infection and virus-induced inflammation, which may be a new mechanism for the increased susceptibility and severity of respiratory viral (i.e., HRV) infection in cigarette smoke-exposed airways with GDF15 overproduction.


Assuntos
Brônquios/virologia , Células Epiteliais/virologia , Fator 15 de Diferenciação de Crescimento/metabolismo , Infecções por Picornaviridae/complicações , Pneumonia/etiologia , Rhinovirus/patogenicidade , Traqueia/virologia , Animais , Brônquios/metabolismo , Brônquios/patologia , Células Cultivadas , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Fator 15 de Diferenciação de Crescimento/genética , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infecções por Picornaviridae/virologia , Pneumonia/metabolismo , Pneumonia/patologia , Transdução de Sinais , Traqueia/metabolismo , Traqueia/patologia , Replicação Viral
2.
PLoS One ; 11(10): e0163927, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27701461

RESUMO

BACKGROUND: MUC18 is upregulated in the lungs of asthma and COPD patients. It has been shown to have pro-inflammatory functions in cultured human airway epithelial cells during viral infections and in mice during lung bacterial infections. However, the in vivo role of MUC18 in the context of viral infections remains poorly understood. The goal of this study is to define the in vivo function of MUC18 during respiratory rhinovirus infection. METHODS: Muc18 wild-type (WT) and knockout (KO) mice were infected with human rhinovirus 1B (HRV-1B) and sacrificed after 1 day to determine the inflammatory and antiviral responses. To examine the direct effects of Muc18 on viral infection, tracheal epithelial cells isolated from WT and KO mice were grown under air-liquid interface and infected with HRV-1B. Finally, siRNA mediated knockdown of MUC18 was performed in human airway epithelial cells (AECs) to define the impact of MUC18 on human airway response to HRV-1B. RESULTS: Both viral load and neutrophilic inflammation were significantly decreased in Muc18 KO mice compared to WT mice. In the in vitro setting, viral load was significantly lower and antiviral gene expression was higher in airway epithelial cells of Muc18 KO mice than the WT mice. Furthermore, in MUC18 knockdown human AECs, viral load was decreased and antiviral gene expression was increased compared to controls. CONCLUSIONS: Our study is the first to demonstrate MUC18's pro-inflammatory and pro-viral function in an in vivo mouse model of rhinovirus infection.


Assuntos
Infecções por Picornaviridae/metabolismo , Rhinovirus/fisiologia , Animais , Antígeno CD146/genética , Antígeno CD146/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/virologia , Traqueia/citologia , Traqueia/metabolismo , Traqueia/virologia , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA