Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Circ Res ; 98(6): 818-27, 2006 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-16497988

RESUMEN

Heterozygous germline mutations in the gene encoding the bone morphogenetic protein type II (BMPR-II) receptor underlie the majority (>70%) of cases of familial pulmonary arterial hypertension (FPAH), and dysfunction of BMP signaling has been implicated in other forms of PAH. The reduced disease gene penetrance in FPAH indicates that other genetic and/or environmental factors may also be required for the clinical manifestation of disease. Of these, the serotonin pathway has been implicated as a major factor in PAH pathogenesis. We investigated the pulmonary circulation of mice deficient in BMPR-II (BMPR2(+/-) mice) and show that pulmonary hemodynamics and vascular morphometry of BMPR2(+/-) mice were similar to wild-type littermate controls under normoxic or chronic hypoxic (2- to 3-week) conditions. However, chronic infusion of serotonin caused increased pulmonary artery systolic pressure, right ventricular hypertrophy, and pulmonary artery remodeling in BMPR2(+/-) mice compared with wild-type littermates, an effect that was exaggerated under hypoxic conditions. In addition, pulmonary, but not systemic, resistance arteries from BMPR2(+/-) mice exhibited increased contractile responses to serotonin mediated by both 5-HT2 and 5-HT1 receptors. Furthermore, pulmonary artery smooth muscle cells from BMPR2(+/-) mice exhibited a heightened DNA synthesis and activation of extracellular signal-regulated kinase 1/2 in response to serotonin compared with wild-type cells. In vitro and in vivo experiments suggested that serotonin inhibits BMP signaling via Smad proteins and the expression of BMP responsive genes. These findings provide the first evidence for an interaction between BMPR-II-mediated signaling and the serotonin pathway, perturbation of which may be critical to the pathogenesis of PAH.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/fisiología , Hipertensión Pulmonar/etiología , Serotonina/farmacología , Animales , Proteína Morfogenética Ósea 2 , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Proteínas Morfogenéticas Óseas/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Susceptibilidad a Enfermedades , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipoxia/fisiopatología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiología , ARN Mensajero/análisis , Receptores de Serotonina/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Transducción de Señal/efectos de los fármacos , Proteínas Smad/genética , Factor de Crecimiento Transformador beta/farmacología , Vasoconstricción/efectos de los fármacos
3.
Circ Res ; 96(10): 1053-63, 2005 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-15845886

RESUMEN

Mutations in the bone morphogenetic protein type II receptor gene (BMPR2) are the major genetic cause of familial pulmonary arterial hypertension (FPAH). Although smooth muscle cell proliferation contributes to the vascular remodeling observed in PAH, the role of BMPs in this process and the impact of BMPR2 mutation remains unclear. Studies involving normal human pulmonary artery smooth muscle cells (PASMCs) suggest site-specific responses to BMPs. Thus, BMP-4 inhibited proliferation of PASMCs isolated from proximal pulmonary arteries, but stimulated proliferation of PASMCs from peripheral arteries, and conferred protection from apoptosis. These differences were not caused by differential activation of BMP signaling pathways because exogenous BMP-4 led to phosphorylation of Smad1, p38(MAPK), and ERK1/2 in both cell types. However, the proproliferative effect of BMP-4 on peripheral PASMCs was found to be p38MAPK/ERK-dependent. Conversely, overexpression of dominant-negative Smad1 converted the response to BMP-4 in proximal PASMCs from inhibitory to proliferative. Furthermore, we confirmed that proximal PASMCs harboring kinase domain mutations in BMPR2 are deficient in Smad signaling and are unresponsive to the growth suppressive effect of BMP-4. Moreover, we show that the pulmonary vasculature of patients with familial and idiopathic PAH are deficient in the activated form of Smad1. We conclude that defective Smad signaling and unopposed p38(MAPK)/ERK signaling, as a consequence of mutation in BMPR2, underlie the abnormal vascular cell proliferation observed in familial PAH.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Músculo Liso Vascular/patología , Arteria Pulmonar/patología , Transducción de Señal/fisiología , Transactivadores/fisiología , Apoptosis/efectos de los fármacos , Proteína Morfogenética Ósea 4 , Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Proteínas Morfogenéticas Óseas/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Mutación , Proteínas Serina-Treonina Quinasas/genética , Arteria Pulmonar/efectos de los fármacos , Proteínas Smad , Proteína Smad1 , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
4.
Am J Physiol Lung Cell Mol Physiol ; 288(2): L370-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15516492

RESUMEN

Fibroblast proliferation, differentiation, and migration contribute to the characteristic pulmonary vascular remodeling seen in primary pulmonary hypertension (PPH). The identification of mutations in the bone morphogenetic protein type II receptor (BMPRII) in PPH have led us to question what role BMPRII and its ligands play in pulmonary vascular remodeling. Thus, to further understand the functional significance of BMPRII in the pulmonary vasculature, we examined the expression of TGF-beta superfamily receptors in human fetal lung fibroblasts (HFL) and investigated the role of BMP4 on cell cycle regulation, fibroblast proliferation, and differentiation. Furthermore, signaling pathways involved in these processes were examined. HFL expressed BMPRI and BMPRII mRNA and demonstrated specific I(125)-BMP4 binding sites. BMP4 inhibited [(3)H]thymidine incorporation and proliferation of HFL; protein expression was increased for the cell cycle inhibitor p21 and reduced for the positive regulators cyclin D and cdk2 by BMP4. BMP4 induced differentiation of HFL into a smooth muscle cell phenotype since protein expression of alpha-smooth muscle actin and smooth muscle myosin was increased. Furthermore, p38(MAPK), ERK1/2, JNK, and Smad1 were phosphorylated by BMP4. Using specific MAPK inhibitors, a dominant negative Smad1 construct, and Smad1 siRNA, we found that the antiproliferative and prodifferentiation effects of BMP4 were Smad1 dependent with JNK also contributing to differentiation. Because failure of Smad phosphorylation is a major feature of BMPRII mutations, these results imply that BMPRII mutations may promote the expansion of fibroblasts resistant to the antiproliferative, prodifferentiation effects of BMPs and suggest a mechanism for the vascular obliteration seen in familial PPH.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Fibroblastos/citología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Pulmón/citología , Miocitos del Músculo Liso/citología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas/farmacología , Transactivadores/metabolismo , Receptores de Activinas Tipo I , Receptores de Proteínas Morfogenéticas Óseas , Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular , ADN/biosíntesis , Humanos , Membranas Intracelulares/metabolismo , Ligandos , Receptores de Factores de Crecimiento/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteínas Smad , Proteína Smad1
5.
Am J Physiol Lung Cell Mol Physiol ; 287(5): L919-27, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15220111

RESUMEN

Chronic hypoxia-induced pulmonary hypertension results partly from proliferation of smooth muscle cells in small peripheral pulmonary arteries. Previously, we demonstrated that hypoxia modulates the proliferation of human peripheral pulmonary artery smooth muscle cells (PASMCs) by induction of cyclooxygenase-2 (COX-2) and production of antiproliferative prostaglandins. The transforming growth factor (TGF)-beta superfamily plays a critical role in the regulation of pulmonary vascular remodeling, although to date an interaction with hypoxia has not been examined. We therefore investigated the pathways involved in the hypoxic induction of COX-2 in peripheral PASMCs and the contribution of TGF-beta1 and bone morphogenetic protein (BMP)-4 in this response. In the present study, we demonstrate that hypoxia induces activation of p38MAPK, ERK1/2, and Akt in PASMCs and that these pathways are involved in the hypoxic regulation of COX-2. Whereas inhibition of p38(MAPK) or ERK1/2 activity suppressed hypoxic induction of COX-2, inhibition of the phosphoinositide 3-kinase pathway enhanced hypoxic induction of COX-2. Furthermore, exogenous TGF-beta1 induced COX-2 mRNA and protein expression, and our findings demonstrate that release of TGF-beta1 by PASMCs during hypoxia contributes to the hypoxic induction of COX-2 via the p38MAPK pathway. In contrast, BMP-4 inhibited the hypoxic induction of COX-2 by an MAPK-independent pathway. Together, these findings suggest that the TGF-beta superfamily is part of an autocrine/paracrine system involved in the regulation of COX-2 expression in the distal pulmonary circulation, and this modulates hypoxia-induced pulmonary vascular cell proliferation.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Hipoxia/metabolismo , Isoenzimas/genética , Músculo Liso Vascular/efectos de los fármacos , Prostaglandina-Endoperóxido Sintasas/genética , Arteria Pulmonar/citología , Factor de Crecimiento Transformador beta/farmacología , Anticuerpos/farmacología , Proteína Morfogenética Ósea 4 , Células Cultivadas , Ciclooxigenasa 2 , Proteínas de Unión al ADN/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , Isoenzimas/metabolismo , Proteínas de la Membrana , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Proteínas Smad , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1 , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Cardiovasc Res ; 55(2): 361-8, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12123775

RESUMEN

OBJECTIVE: To test the hypothesis that Vegf-B contributes to the pulmonary vascular remodelling, and the associated pulmonary hypertension, induced by exposure of mice to chronic hypoxia. METHODS: Right ventricular systolic pressure, the ratio of right ventricle/[left ventricle+septum] (RV/[LV+S]) and the thickness of the media (relative to vessel diameter) of intralobar pulmonary arteries (o.d. 50-150 and 151-420 microm) were determined in Vegfb knockout mice (Vegfb(-/-); n=17) and corresponding wild-type mice (Vegfb(+/+); n=17) exposed to chronic hypoxia (10% oxygen) or housed in room air (normoxia) for 4 weeks. RESULTS: In Vegfb(+/+) mice hypoxia caused (i) pulmonary hypertension (a 70% increase in right ventricular systolic pressure compared with normoxic Vegfb(+/+) mice; P<0.001), (ii) right ventricular hypertrophy (a 66% increase in RV/[LV+S]; P<0.001) and (iii) pulmonary vascular remodelling (a 27-36% increase in pulmonary arterial medial thickness; P<0.05). In contrast, in Vegfb(-/-) mice hypoxia did not cause any increase in either right ventricular systolic pressure or pulmonary arterial medial thickness; also right ventricular hypertrophy (41% increase in RV/[LV+S]; P<0.001) was less pronounced (P<0.05) than in Vegfb(+/+) mice. CONCLUSION: Vegf-B may have a role in the development of chronic hypoxic pulmonary hypertension in mice by contributing to pulmonary vascular remodelling. If so, the effect of Vegf-B appears to be different from that of Vegf-A which is reported to protect against, rather than contribute to, hypoxia-induced pulmonary vascular remodelling.


Asunto(s)
Factores de Crecimiento Endotelial/fisiología , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , Animales , Enfermedad Crónica , Femenino , Hematócrito , Hipertensión Pulmonar/patología , Hipertrofia Ventricular Derecha/fisiopatología , Ratones , Ratones Noqueados , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Circulación Pulmonar , Túnica Media/patología , Factor B de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA