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












Base de datos
Intervalo de año de publicación
1.
PLoS One ; 11(12): e0167169, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27935962

RESUMEN

Chronic obstructive pulmonary disease (COPD) is characterized by persistent airflow limitation and lung inflammation resulting in a progressive decline in lung function whose principle cause is cigarette smoke. MAP3K19 is a novel kinase expressed predominantly by alveolar and interstitial macrophages and bronchial epithelial cells in the lung. We found that MAP3K19 mRNA was overexpressed in a limited sampling of lung tissue from COPD patients, and a closer examination found it to be overexpressed in bronchoalveolar macrophages from COPD patients, as well as the bronchial epithelium and inflammatory cells in the lamina propria. We further found MAP3K19 to be induced in various cell lines upon environmental stress, such as cigarette smoke, oxidative and osmotic stress. Exogenous expression of MAP3K19 in cells caused an upregulation of transcriptionally active NF-κB, and secretion of the chemokines CXCL-8, CCL-20 and CCL-7. Inhibition of MAP3K19 activity by siRNA or small molecular weight inhibitors caused a decrease in cigarette smoke-induced inflammation in various murine models, which included a decrease in pulmonary neutrophilia and KC levels. In a chronic cigarette smoke model, inhibition of MAP3K19 significantly attenuated emphysematous changes in airway parenchyma. Finally, in a viral exacerbation model, mice exposed to cigarette smoke and influenza A virus showed a decrease in pulmonary neutrophilia, pro-inflammatory cytokines and viral load upon inhibition of MAP3K19. Collectively, these results suggest that inhibition of MAP3K19 may represent a novel strategy to target COPD that promises to have a potential therapeutic benefit for patients.


Asunto(s)
Mediadores de Inflamación/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Neumonía/enzimología , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Sistema Respiratorio/enzimología , Fumar/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Western Blotting , Línea Celular Tumoral , Citocinas/metabolismo , Células Epiteliales/enzimología , Células Epiteliales/metabolismo , Femenino , Expresión Génica , Células HEK293 , Humanos , Quinasas Quinasa Quinasa PAM/genética , Macrófagos/enzimología , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , FN-kappa B/metabolismo , Neumonía/etiología , Neumonía/genética , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Interferencia de ARN , Sistema Respiratorio/metabolismo , Sistema Respiratorio/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
2.
PLoS One ; 11(5): e0154874, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27144281

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive, debilitating disease for which two medications, pirfenidone and nintedanib, have only recently been approved for treatment. The cytokine TGF-ß has been shown to be a central mediator in the disease process. We investigated the role of a novel kinase, MAP3K19, upregulated in IPF tissue, in TGF-ß-induced signal transduction and in bleomycin-induced pulmonary fibrosis. MAP3K19 has a very limited tissue expression, restricted primarily to the lungs and trachea. In pulmonary tissue, expression was predominantly localized to alveolar and interstitial macrophages, bronchial epithelial cells and type II pneumocytes of the epithelium. MAP3K19 was also found to be overexpressed in bronchoalveolar lavage macrophages from IPF patients compared to normal patients. Treatment of A549 or THP-1 cells with either MAP3K19 siRNA or a highly potent and specific inhibitor reduced phospho-Smad2 & 3 nuclear translocation following TGF-ß stimulation. TGF-ß-induced gene transcription was also strongly inhibited by both the MAP3K19 inhibitor and nintedanib, whereas pirfenidone had a much less pronounced effect. In combination, the MAP3K19 inhibitor appeared to act synergistically with either pirfenidone or nintedanib, at the level of target gene transcription or protein production. Finally, in an animal model of IPF, inhibition of MAP3K19 strongly attenuated bleomycin-induced pulmonary fibrosis when administered either prophylactically ortherapeutically. In summary, these results strongly suggest that inhibition of MAP3K19 may have a beneficial therapeutic effect in the treatment of IPF and represents a novel strategy to target this disease.


Asunto(s)
Bleomicina/farmacología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células A549 , Animales , Lavado Broncoalveolar/métodos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Células HeLa , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Indoles/farmacología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Piridonas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...