Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Am J Physiol Lung Cell Mol Physiol ; 314(4): L593-L605, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29212800

RESUMEN

Pulmonary hypertension (PH) is a lethal condition, and current vasodilator therapy has limited effect. Antiproliferative strategies targeting platelet-derived growth factor (PDGF) receptors, such as imatinib, have generated promising results in animal studies. Imatinib is, however, a nonspecific tyrosine kinase inhibitor and has in clinical studies caused unacceptable adverse events. Further studies are needed on the role of PDGF signaling in PH. Here, mice expressing a variant of PDGF-B with no retention motif ( Pdgfbret/ret), resulting in defective binding to extracellular matrix, were studied. Following 4 wk of hypoxia, right ventricular systolic pressure, right ventricular hypertrophy, and vascular remodeling were examined. Pdgfbret/ret mice did not develop PH, as assessed by hemodynamic parameters. Hypoxia did, however, induce vascular remodeling in Pdgfbret/ret mice; but unlike the situation in controls where the remodeling led to an increased concentric muscularization of arteries, the vascular remodeling in Pdgfbret/ret mice was characterized by a diffuse muscularization, in which cells expressing smooth muscle cell markers were found in the interalveolar septa detached from the normally muscularized intra-acinar vessels. Additionally, fewer NG2-positive perivascular cells were found in Pdgfbret/ret lungs, and mRNA analyses showed significantly increased levels of Il6 following hypoxia, a known promigratory factor for pericytes. No differences in proliferation were detected at 4 wk. This study emphasizes the importance of extracellular matrix-growth factor interactions and adds to previous knowledge of PDGF-B in PH pathobiology. In summary, Pdgfbret/ret mice have unaltered hemodynamic parameters following chronic hypoxia, possibly secondary to a disorganized vascular muscularization.


Asunto(s)
Modelos Animales de Enfermedad , Matriz Extracelular/patología , Hipertensión Pulmonar/patología , Hipoxia/fisiopatología , Linfocinas/fisiología , Músculo Liso Vascular/patología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Remodelación Vascular , Animales , Proliferación Celular , Células Cultivadas , Matriz Extracelular/metabolismo , Femenino , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Pericitos/metabolismo , Pericitos/patología , Transducción de Señal
3.
J Vasc Res ; 54(4): 246-256, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28796998

RESUMEN

BACKGROUND: Serotonin (5-HT) is considered to play a role in pulmonary arterial hypertension by regulating vascular remodeling and smooth muscle contractility. Here, arteries from mice with inducible and smooth muscle-specific deletion of Dicer were used to address mechanisms by which microRNAs control 5-HT-induced contraction. METHODS: Mice were used 5 weeks after Dicer deletion, and pulmonary artery contractility was analyzed by wire myography. RESULTS: No change was seen in right ventricular systolic pressure following dicer deletion, but systemic blood pressure was reduced. Enhanced 5-HT-induced contraction in Dicer KO pulmonary arteries was associated with increased 5-HT2A receptor mRNA expression whereas 5-HT1B and 5-HT2B receptor mRNAs were unchanged. Contraction by the 5-HT2A agonist TCB-2 was increased in Dicer KO as was the response to the 5-HT2B agonist BW723C86. Effects of Src and protein kinase C inhibition were similar in control and KO arteries, but the effect of inhibition of Rho kinase was reduced. We identified miR-30c as a potential candidate for 5-HT2A receptor regulation as it repressed 5-HT2A mRNA and protein. CONCLUSION: Our findings show that 5-HT receptor signaling in the arterial wall is subject to regulation by microRNAs and that this entails altered 5-HT2A receptor expression and signaling.


Asunto(s)
MicroARNs/metabolismo , Arteria Pulmonar/efectos de los fármacos , Serotonina/farmacología , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Animales , Células Cultivadas , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Genotipo , Masculino , Ratones Noqueados , MicroARNs/genética , Miografía , Fenotipo , Proteína Quinasa C/metabolismo , Arteria Pulmonar/metabolismo , Receptor de Serotonina 5-HT2A/efectos de los fármacos , Receptor de Serotonina 5-HT2A/genética , Receptor de Serotonina 5-HT2A/metabolismo , Ribonucleasa III/deficiencia , Ribonucleasa III/genética , Transducción de Señal/efectos de los fármacos , Transfección , Quinasas Asociadas a rho/metabolismo , Familia-src Quinasas/metabolismo
4.
PLoS One ; 11(3): e0152276, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27032083

RESUMEN

Platelet-derived growth factor D (PDGF-D) is the most recently discovered member of the PDGF family. PDGF-D signals through PDGF receptor ß, but its biological role remains largely unknown. In contrast to other members of the PDGF family of growth factors, which have been extensively investigated using different knockout approaches in mice, PDGF-D has until now not been characterized by gene inactivation in mice. Here, we present the phenotype of a constitutive Pdgfd knockout mouse model (Pdgfd-/-), carrying a LacZ reporter used to visualize Pdgfd promoter activity. Inactivation of the Pdgfd gene resulted in a mild phenotype in C57BL/6 mice, and the offspring was viable, fertile and generally in good health. We show that Pdgfd reporter gene activity was consistently localized to vascular structures in both postnatal and adult tissues. The expression was predominantly arterial, often localizing to vascular bifurcations. Endothelial cells appeared to be the dominating source for Pdgfd, but reporter gene activity was occasionally also found in subpopulations of mural cells. Tissue-specific analyses of vascular structures revealed that NG2-expressing pericytes of the cardiac vasculature were disorganized in Pdgfd-/- mice. Furthermore, Pdgfd-/- mice also had a slightly elevated blood pressure. In summary, the vascular expression pattern together with morphological changes in NG2-expressing cells, and the increase in blood pressure, support a function for PDGF-D in regulating systemic arterial blood pressure, and suggests a role in maintaining vascular homeostasis.


Asunto(s)
Linfocinas/genética , Ratones Endogámicos C57BL/genética , Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Arterias/metabolismo , Arterias/ultraestructura , Presión Sanguínea , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Fertilidad , Expresión Génica , Técnicas de Inactivación de Genes , Glucosa/metabolismo , Corazón , Masculino , Ratones Endogámicos C57BL/fisiología , Ratones Noqueados , Fenotipo , Regiones Promotoras Genéticas
5.
PLoS One ; 10(9): e0137949, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26394398

RESUMEN

Despite its known expression in both the vascular endothelium and the lung epithelium, until recently the physiological role of the adhesion receptor Gpr116/ADGRF5 has remained elusive. We generated a new mouse model of constitutive Gpr116 inactivation, with a large genetic deletion encompassing exon 4 to exon 21 of the Gpr116 gene. This model allowed us to confirm recent results defining Gpr116 as necessary regulator of surfactant homeostasis. The loss of Gpr116 provokes an early accumulation of surfactant in the lungs, followed by a massive infiltration of macrophages, and eventually progresses into an emphysema-like pathology. Further analysis of this knockout model revealed cerebral vascular leakage, beginning at around 1.5 months of age. Additionally, endothelial-specific deletion of Gpr116 resulted in a significant increase of the brain vascular leakage. Mice devoid of Gpr116 developed an anatomically normal and largely functional vascular network, surprisingly exhibited an attenuated pathological retinal vascular response in a model of oxygen-induced retinopathy. These data suggest that Gpr116 modulates endothelial properties, a previously unappreciated function despite the pan-vascular expression of this receptor. Our results support the key pulmonary function of Gpr116 and describe a new role in the central nervous system vasculature.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Endotelio Vascular/metabolismo , Surfactantes Pulmonares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Western Blotting , Líquido del Lavado Bronquioalveolar/química , Permeabilidad Capilar/genética , Femenino , Expresión Génica , Homeostasis/genética , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Modelos Biológicos , Miocardio/metabolismo , Miocardio/patología , Receptores Acoplados a Proteínas G/genética , Neovascularización Retiniana/genética , Neovascularización Retiniana/metabolismo , Bazo/metabolismo , Bazo/patología
6.
Cardiovasc Res ; 107(1): 20-31, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25952902

RESUMEN

AIMS: Excessive vascular cell proliferation is an important component of pulmonary hypertension (PH). Perlecan is the major heparan sulfate (HS) proteoglycan in the vascular extracellular matrix. It binds growth factors, including FGF2, and either restricts or promotes cell proliferation. In this study, we have explored the effects of perlecan HS deficiency on pulmonary vascular development and in hypoxia-induced PH. METHODS AND RESULTS: In normoxia, Hspg2(Δ3/Δ3) mice, deficient in perlecan HS, had reduced pericytes and muscularization of intra-acinar vessels. Pulmonary angiography revealed a peripheral perfusion defect. Despite these abnormalities, right ventricular systolic pressure (RVSP) and myocardial mass remained normal. After 4 weeks of hypoxia, increases in the proportion of muscularized vessels, RVSP, and right ventricular hypertrophy were significantly less in Hspg2(Δ3/Δ3) compared with wild type. The early phase of hypoxia induced a significantly lower increase in fibroblast growth factor receptor-1 (FGFR1) protein level and receptor phosphorylation, and reduced pulmonary artery smooth muscle cell (PASMC) proliferation in Hspg2(Δ3/Δ3). At 4 weeks, FGF2 mRNA and protein were also significantly reduced in Hspg2(Δ3/Δ3) lungs. Ligand and carbohydrate engagement assay showed that perlecan HS is required for HS-FGF2-FGFR1 ternary complex formation. In vitro, proliferation assays showed that PASMC proliferation is reduced by selective FGFR1 inhibition. PASMC adhesion to fibronectin was higher in Hspg2(Δ3/Δ3) compared with wild type. CONCLUSIONS: Perlecan HS chains are important for normal vascular arborization and recruitment of pericytes to pulmonary vessels. Perlecan HS deficiency also attenuates hypoxia-induced PH, where the underlying mechanisms involve impaired FGF2/FGFR1 interaction, inhibition of PASMC growth, and altered cell-matrix interactions.


Asunto(s)
Proteoglicanos de Heparán Sulfato/metabolismo , Hipertensión Pulmonar/etiología , Hipoxia/complicaciones , Arteria Pulmonar/fisiología , Animales , Adhesión Celular , Proliferación Celular , Células Cultivadas , Femenino , Factor 2 de Crecimiento de Fibroblastos/análisis , Proteoglicanos de Heparán Sulfato/análisis , Proteoglicanos de Heparán Sulfato/deficiencia , Hipertensión Pulmonar/prevención & control , Ratones , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Fosforilación , Arteria Pulmonar/diagnóstico por imagen , Radiografía , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
7.
J Cell Physiol ; 230(2): 482-7, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25078760

RESUMEN

Smooth muscle cell (SMC) proliferation is a key process in stabilization of atherosclerotic plaques, and during restenosis after interventions. A clearer understanding of SMC growth regulation is therefore needed to design specific anti-proliferative therapies. Retinoic acid has been shown to inhibit proliferation of SMCs both in vitro and in vivo and to affect the expression of extracellular matrix molecules. To explore the mechanisms behind the growth inhibitory activity of retinoic acid, we hypothesized that retinoids may induce the expression of perlecan, a large heparan sulfate proteoglycan with anti-proliferative properties. Perlecan expression and accumulation was induced in murine SMC cultures by all-trans-retinoic acid (AtRA). Moreover, the growth inhibitory effect of AtRA on wild-type cells was greatly diminished in SMCs from transgenic mice expressing heparan sulfate-deficient perlecan, indicating that the inhibition is perlecan heparan sulfate-dependent. In addition, AtRA influenced activation and phosphorylation of PTEN and Akt differently in wild-type and mutant SMCs, consistent with previous studies of perlecan-dependent SMC growth inhibition. We demonstrate that AtRA regulates perlecan expression in SMCs and that the inhibition of SMC proliferation by AtRA is, at least in part, secondary to an increased expression of perlecan and dependent upon its heparan sulfate-chains.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Proteoglicanos de Heparán Sulfato/farmacología , Heparitina Sulfato/farmacología , Músculo Liso Vascular/efectos de los fármacos , Tretinoina/farmacología , Animales , Células Cultivadas , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Ratones , Ratones Transgénicos , Músculo Liso Vascular/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...