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

Banco de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Microvasc Res ; 84(3): 270-7, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22835519

RESUMEN

BACKGROUND: Endothelial progenitor cells (EPCs) participate in post-natal vasculogenesis. Maintaining the preliminary progenitor phenotype and good proliferation capacity of EPCs is key to their use in treating cardiovascular ischemic diseases. However, transcriptional regulation in EPCs remains largely unknown. We investigated the effect of leukemia inhibitory factor (LIF) combined with vascular endothelial growth factor (VEGF) on EPCs and the potential roles of Krüppel-like transcription factors (KLFs). METHODS AND RESULTS: Co-treatment with LIF and VEGF (100 ng/ml each) (V+L) could increase EPC colony-forming units and CD34 expression, which reflects the EPC progenitor phenotype and alleviated differentiation of EPCs. The effect was associated with Akt activation and increased expression of KLF4. Upregulation of KLF4 induced by V+L could be inhibited by transfection with dominant-negative Akt adenovirus. Furthermore, overexpression of KLF4 in EPCs enhanced the expression of CD34 and alleviated cell differentiation but did not increase the phosphorylation of Akt. CONCLUSIONS: LIF combined with VEGF can maintain the preliminary, progenitor phenotype of EPCs and alleviate cell differentiation by upregulating KLF4, which may provide new insights into transcriptional regulation in EPCs.


Asunto(s)
Células Endoteliales/citología , Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Células Madre/citología , Antígenos CD34/biosíntesis , Diferenciación Celular , Proliferación Celular , Sangre Fetal/citología , Citometría de Flujo/métodos , Humanos , Factor 4 Similar a Kruppel , Microcirculación , Fenotipo , Fosforilación , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Cardiovasc Res ; 99(3): 514-24, 2013 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-23667185

RESUMEN

AIMS: Endothelial colony forming cells (ECFCs) participate in post-natal vasculogenesis. We previously reported that vascular endothelial growth factor (VEGF) promotes human ECFC differentiation through AMP-activated protein kinase (AMPK) activation. However, the mechanisms underlying transcriptional regulation of ECFC differentiation still remain largely elusive. Here, we investigated the role of transcription factor Krüppel-like factor 2 (KLF2) in the regulation of ECFC function. METHODS AND RESULTS: Human ECFCs were isolated from cord blood and cultured. Treatment with VEGF significantly increased endothelial markers in ECFCs and their capacity for migration and tube formation. The mRNA and protein levels of KLF2 were also significantly up-regulated. This up-regulation was abrogated by AMPK inhibition or by knockdown of KLF2 with siRNA. Furthermore, adenovirus-mediated overexpression of KLF2 promoted ECFC differentiation by enhancing expression of endothelial cell markers, reducing expression of progenitor cell markers, and increasing the capacity for tube formation in vitro, indicating the important role of KLF2 in ECFC-mediated angiogenesis. Histone deacetylase 5 (HDAC5) was phosphorylated by AMPK activity induced by VEGF and the AMPK agonist AICAR (5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide). In vivo angiogenesis assay revealed that overexpression of KLF2 in bone-marrow-derived pro-angiogenic progenitor cells promoted vessel formation when the cells were implanted in C57BL/6 mice. CONCLUSION: Up-regulation of KLF2 by AMPK activation constitutes a novel mechanism of ECFC differentiation, and may have therapeutic value in the treatment of ischaemic heart disease.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Células Endoteliales/trasplante , Sangre Fetal/citología , Sangre Fetal/metabolismo , Técnicas de Silenciamiento del Gen , Xenoinjertos , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Fosforilación , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
PLoS One ; 6(8): e23554, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21876756

RESUMEN

Prostaglandin E2 (PGE2) has been reported to modulate angiogenesis, the process of new blood vessel formation, by promoting proliferation, migration and tube formation of endothelial cells. Endothelial progenitor cells are known as a subset of circulating bone marrow mononuclear cells that have the capacity to differentiate into endothelial cells. However, the mechanism underlying the stimulatory effects of PGE2 and its specific receptors on bone marrow-derived cells (BMCs) in angiogenesis has not been fully characterized. Treatment with PGE2 significantly increased the differentiation and migration of BMCs. Also, the markers of differentiation to endothelial cells, CD31 and von Willebrand factor, and the genes associated with migration, matrix metalloproteinases 2 and 9, were significantly upregulated. This upregulation was abolished by dominant-negative AMP-activated protein kinase (AMPK) and AMPK inhibitor but not protein kinase, a inhibitor. As a functional consequence of differentiation and migration, the tube formation of BMCs was reinforced. Along with altered BMCs functions, phosphorylation and activation of AMPK and endothelial nitric oxide synthase, the target of activated AMPK, were both increased which could be blocked by EP4 blocking peptide and simulated by the agonist of EP4 but not EP1, EP2 or EP3. The pro-angiogenic role of PGE2 could be repressed by EP4 blocking peptide and retarded in EP4(+/-) mice. Therefore, by promoting the differentiation and migration of BMCs, PGE2 reinforced their neovascularization by binding to the receptor of EP4 in an AMPK-dependent manner. PGE2 may have clinical value in ischemic heart disease.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Células de la Médula Ósea/citología , Diferenciación Celular/efectos de los fármacos , Dinoprostona/farmacología , Células Endoteliales/citología , Células Endoteliales/enzimología , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/enzimología , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Quinasas/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA