Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cell Sci ; 120(Pt 6): 929-42, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17327280

RESUMEN

Asymmetric dimethylarginine (ADMA) is an inhibitor of nitric oxide production associated with abnormal blood vessel growth and repair, however, the mechanism of action of ADMA is not well understood. We studied the role of exogenous and endogenous ADMA in the regulation of cell motility and actin cytoskeleton in porcine pulmonary endothelial cells (PAECs) and pulmonary microvascular endothelial cells (PMECs) from knockout mice that lack one of the enzyme metabolising ADMA, dimethylarginine dimethylaminohydrolase I (DDAHI) as well as endothelial cells overexpressing DDAH in vitro. We show that ADMA induced stress fibre and focal adhesion formation and inhibited cell motility in primary pulmonary endothelial cells. The effects of ADMA depended on the activity of RhoA and Rho kinase and were reversed by overexpression of DDAH, nitric oxide donors and protein kinase G activator, 8-bromo-cGMP. ADMA also inhibited the activities of Rac1 and Cdc42 in cells but these changes had a minor effect on cell motility. Endogenous ADMA increased RhoA activity and inhibited cell motility in PMECs from DDAHI knockout mice and inhibited angiogenesis in vitro. These results are the first demonstration that metabolism of cardiovascular risk factor ADMA regulates endothelial cell motility, an important factor in angiogenesis and vascular repair.


Asunto(s)
Amidohidrolasas/metabolismo , Arginina/análogos & derivados , Movimiento Celular/fisiología , Células Endoteliales/fisiología , Neovascularización Fisiológica/efectos de los fármacos , Transducción de Señal , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/fisiología , Amidohidrolasas/genética , Animales , Arginina/farmacología , Arginina/fisiología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/fisiología , Células Endoteliales/efectos de los fármacos , Activación Enzimática , Adhesiones Focales/fisiología , Pulmón/irrigación sanguínea , Pulmón/citología , Ratones , Ratones Noqueados , Fibras de Estrés/fisiología , Porcinos , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoB/metabolismo
2.
Am J Physiol Lung Cell Mol Physiol ; 290(6): L1173-82, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16428270

RESUMEN

Hypoxia is a common cause of persistent pulmonary hypertension in the newborn (PPHN), a condition associated with endothelial dysfunction and abnormal pulmonary vascular remodeling. The GTPase RhoA has been implicated in the pathogenesis of PPHN, but its contribution to endothelial remodeling and function is not known. We studied pulmonary artery endothelial cells (PAECs) taken from piglets with chronic hypoxia-induced pulmonary hypertension and from healthy animals and analyzed the roles of Rho GTPases in the regulation of the endothelial phenotype and function under basal normoxic conditions, acute hypoxia, and reoxygenation. The activities of RhoA, Rac1, and Cdc42 were correlated with changes in the endothelial cytoskeleton, adherens junctions, permeability, ROS production, VEGF levels, and activities of transcription factors hypoxia-inducible factor (HIF)-1alpha and NF-kappaB. Adenoviral gene transfer was used to express dominant-negative GTPases, kinase-dead p21-activated kinase (PAK)-1, and constitutively activated Rac1 in cells. PAECs from pulmonary hypertensive piglets had a stable abnormal phenotype with a sustained reduction in Rac1 activity and an increase in RhoA activity, which correlated with an increase in actin stress fiber formation, increased permeability, and a decrease in VEGF and ROS production. Cells from pulmonary hypertensive animals were still able to respond to acute hypoxia. They also showed high activities of HIF-1alpha and NF-kappaB, likely to result from changes in the activities of Rho GTPases. Activation of Rac1 and its effector PAK-1 as well as inhibition of RhoA restored the abnormal phenotype and permeability of hypertensive PAECs to normal.


Asunto(s)
Endotelio Vascular/fisiopatología , Hipertensión Pulmonar/fisiopatología , Proteína de Unión al GTP rac1/fisiología , Proteína de Unión al GTP rhoA/fisiología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Hipoxia/fisiopatología , Circulación Pulmonar/fisiología , Porcinos , Proteína de Unión al GTP rhoA/antagonistas & inhibidores
3.
Am J Physiol Lung Cell Mol Physiol ; 288(4): L749-60, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15591411

RESUMEN

Hypoxia/reoxygenation-induced changes in endothelial permeability are accompanied by endothelial actin cytoskeletal and adherens junction remodeling, but the mechanisms involved are uncertain. We therefore measured the activities of the Rho GTPases Rac1, RhoA, and Cdc42 during hypoxia/reoxygenation and correlated them with changes in endothelial permeability, remodeling of the actin cytoskeleton and adherens junctions, and production of ROS. Dominant negative forms of Rho GTPases were introduced into cells by adenoviral gene transfer and transfection, and inhibitors of NADPH oxidase, PI3 kinase, and Rho kinase were used to characterize the signaling pathways involved. In some experiments constitutively activated forms of RhoA and Rac1 were also used. We show for the first time that hypoxia/reoxygenation-induced changes in endothelial permeability result from coordinated actions of the Rho GTPases Rac1 and RhoA. Rac1 and RhoA rapidly respond to changes in oxygen tension, and their activity depends on NADPH oxidase- and PI3 kinase-dependent production of ROS. Rac1 acts upstream of RhoA, and its transient inhibition by acute hypoxia leads to activation of RhoA followed by stress fiber formation, dispersion of adherens junctions, and increased endothelial permeability. Reoxygenation strongly activates Rac1 and restores cortical localization of F-actin and VE-cadherin. This effect is a result of Rac1-mediated inhibition of RhoA and can be prevented by activators of RhoA, L63RhoA, and lysophosphatidic acid. Cdc42 activation follows the RhoA pattern of activation but has no effect on actin remodeling, junctional integrity, or endothelial permeability. Our results show that Rho GTPases act as mediators coupling cellular redox state to endothelial function.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Oxígeno/metabolismo , Arteria Pulmonar/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Actinas/metabolismo , Adenoviridae/genética , Uniones Adherentes/efectos de los fármacos , Animales , Antígenos CD , Cadherinas/metabolismo , Citoesqueleto/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Genes Dominantes , Hipoxia , Lisofosfolípidos/farmacología , NADPH Oxidasas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Porcinos , Uniones Estrechas/efectos de los fármacos , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rhoA/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA