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











Base de datos
Intervalo de año de publicación
1.
Respir Res ; 16: 60, 2015 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-26006045

RESUMEN

BACKGROUND: Mechanical ventilation plays a central role in the injury of premature lungs. However, the mechanisms by which mechanical signals trigger an inflammatory cascade to promote lung injury are not well-characterized. Transient receptor potential vanilloid 4 (TRPV4), a calcium-permeable mechanoreceptor channel has been shown to be a major determinant of ventilator-induced acute lung injury in adult models. However, the role of these channels as modulators of inflammation in immature lungs is unknown. In this study, we tested the hypothesis that TRPV4 channels are important mechanotransducers in fetal lung injury. METHODS: Expression of TRPV4 in the mouse fetal lung was investigated by immunohistochemistry, Western blot and qRT-PCR. Isolated fetal epithelial cells were exposed to mechanical stimulation using the Flexcell Strain Unit and inflammation and differentiation were analyzed by ELISA and SP-C mRNA, respectively. RESULTS: TRPV4 is developmentally regulated in the fetal mouse lung; it is expressed in the lung epithelium and increases with advanced gestation. In contrast, in isolated epithelial cells, TRPV4 expression is maximal at E17-E18 of gestation. Mechanical stretch increases TRPV4 in isolated fetal epithelial cells only during the canalicular stage of lung development. Using the TRPV4 agonist GSK1016790A, the antagonist HC-067047, and the cytokine IL-6 as a marker of inflammation, we observed that TRPV4 regulates release of IL-6 via p38 and ERK pathways. Interestingly, stretch-induced differentiation of fetal epithelial cells was also modulated by TRPV4. CONCLUSION: These studies demonstrate that TRPV4 may play an important role in the transduction of mechanical signals in the fetal lung epithelium by modulating not only inflammation but also the differentiation of fetal epithelial cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Epiteliales/fisiología , Mecanotransducción Celular/fisiología , Canales Catiónicos TRPV/biosíntesis , Animales , Células Cultivadas , Células Epiteliales/patología , Femenino , Feto , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Embarazo , Mucosa Respiratoria/embriología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología
2.
J Biol Chem ; 288(35): 25646-25657, 2013 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-23888051

RESUMEN

Mechanical forces are critical for normal fetal lung development. However, the mechanisms regulating this process are not well-characterized. We hypothesized that strain-induced release of HB-EGF and TGF-α is mediated via integrin-ADAM17/TACE interactions. Employing an in vitro system to simulate mechanical forces in fetal lung development, we showed that mechanical strain of fetal epithelial cells actives TACE, releases HB-EGF and TGF-α, and promotes differentiation. In contrast, in samples incubated with the TACE inhibitor IC-3 or in cells isolated from TACE knock-out mice, mechanical strain did not release ligands or promote cell differentiation, which were both rescued after transfection of ADAM17. Cell adhesion assay and co-immunoprecipitation experiments in wild-type and TACE knock-out cells using several TACE constructs demonstrated not only that integrins α6 and ß1 bind to TACE via the disintegrin domain but also that mechanical strain enhances these interactions. Furthermore, force applied to these integrin receptors by magnetic beads activated TACE and shed HB-EGF and TGF-α. The contribution of integrins α6 and ß1 to differentiation of fetal epithelial cells by strain was demonstrated by blocking their binding site with specific antibodies and by culturing the cells on membranes coated with anti-integrin α6 and ß1 antibodies. In conclusion, mechanical strain releases HB-EGF and TGF-α and promotes fetal type II cell differentiation via α6ß1 integrin-ADAM17/TACE signaling pathway. These investigations provide novel mechanistic information on how mechanical forces promote fetal lung development and specifically differentiation of epithelial cells. This information could be also relevant to other tissues exposed to mechanical forces.


Asunto(s)
Proteínas ADAM/metabolismo , Diferenciación Celular/fisiología , Células Epiteliales/metabolismo , Integrina alfa6beta1/metabolismo , Pulmón/embriología , Mucosa Respiratoria/embriología , Transducción de Señal/fisiología , Proteínas ADAM/genética , Proteína ADAM17 , Animales , Células Epiteliales/citología , Factor de Crecimiento Similar a EGF de Unión a Heparina , Integrina alfa6beta1/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Pulmón/citología , Ratones , Ratones Noqueados , Unión Proteica , Mucosa Respiratoria/citología , Estrés Fisiológico/fisiología , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador alfa/metabolismo
3.
J Biol Chem ; 287(22): 18091-102, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22493501

RESUMEN

Stretch-induced differentiation of lung fetal type II epithelial cells is mediated through EGFR (ErbB1) via release of HB-EGF and TGF-α ligands. Employing an EGFR knock-out mice model, we further investigated the role of the ErbB family of receptors in mechanotranduction during lung development. Deletion of EGFR prevented endogenous and mechanical stretch-induced type II cell differentiation via the ERK pathway, which was rescued by overexpression of a constitutively active MEK. Interestingly, the expression of ErbB4, the only ErbB receptor that EGFR co-precipitates in wild-type cells, was decreased in EGFR-deficient type II cells. Similar to EGFR, ErbB4 was activated by stretch and participated in ERK phosphorylation and type II cell differentiation. However, neuregulin (NRG) or stretch-induced ErbB4 activation were blunted in EGFR-deficient cells and not rescued after ErbB4 overexpression, suggesting that induction of ErbB4 phosphorylation is EGFR-dependent. Finally, we addressed how shedding of ligands is regulated by EGFR. In knock-out cells, TGF-α, a ligand for EGFR, was not released by stretch, while HB-EGF, a ligand for EGFR and ErbB4, was shed by stretch although to a lower magnitude than in normal cells. Release of these ligands was inhibited by blocking EGFR and ERK pathway. In conclusion, our studies show that EGFR and ErbB4 regulate stretch-induced type II cell differentiation via ERK pathway. Interactions between these two receptors are important for mechanical signals in lung fetal type II cells. These studies provide novel insights into the cell signaling mechanisms regulating ErbB family receptors in lung cell differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Receptores ErbB/fisiología , Feto/citología , Mecanotransducción Celular/fisiología , Animales , Secuencia de Bases , Cartilla de ADN , Receptores ErbB/genética , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Ratones , Ratones Noqueados , Embarazo , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor ErbB-4
4.
J Vis Exp ; (60)2012 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-22371001

RESUMEN

Mechanical forces generated in utero by repetitive breathing-like movements and by fluid distension are critical for normal lung development. A key component of lung development is the differentiation of alveolar type II epithelial cells, the major source of pulmonary surfactant. These cells also participate in fluid homeostasis in the alveolar lumen, host defense, and injury repair. In addition, distal lung parenchyma cells can be directly exposed to exaggerated stretch during mechanical ventilation after birth. However, the precise molecular and cellular mechanisms by which lung cells sense mechanical stimuli to influence lung development and to promote lung injury are not completely understood. Here, we provide a simple and high purity method to isolate type II cells and fibroblasts from rodent fetal lungs. Then, we describe an in vitro system, The Flexcell Strain Unit, to provide mechanical stimulation to fetal cells, simulating mechanical forces in fetal lung development or lung injury. This experimental system provides an excellent tool to investigate molecular and cellular mechanisms in fetal lung cells exposed to stretch. Using this approach, our laboratory has identified several receptors and signaling proteins that participate in mechanotransduction in fetal lung development and lung injury.


Asunto(s)
Técnicas Citológicas/métodos , Pulmón/citología , Células Epiteliales Alveolares/citología , Animales , Femenino , Feto/citología , Fibroblastos/citología , Inmunohistoquímica/métodos , Pulmón/química , Pulmón/embriología , Pulmón/metabolismo , Mecanotransducción Celular , Ratones , Microscopía Fluorescente/métodos , Embarazo , Proteína C Asociada a Surfactante Pulmonar/química , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA