Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS One ; 8(1): e54687, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23349951

RESUMEN

Accurate regulation of dermal fibroblast function plays a crucial role in wound healing. Many fibrotic diseases are characterized by a failure to conclude normal tissue repair and the persistence of fibroblasts inside lesions. In the present study we demonstrate that endoglin haploinsufficiency promotes fibroblast accumulation during wound healing. Moreover, scars from endoglin-heterozygous (Eng(+/-)) mice show persisting fibroblasts 12 days after wounding, which could lead to a fibrotic scar. Endoglin haploinsufficiency results in increased proliferation and migration of primary cultured murine dermal fibroblasts (MDFs). Moreover, Eng(+/-) MDF have diminished responses to apoptotic signals compared with control cells. Altogether, these modifications could explain the augmented presence of fibroblasts in Eng(+/-) mice wounds. We demonstrate that endoglin expression regulates Akt phosphorylation and that PI3K inhibition abolishes the differences in proliferation between endoglin haploinsufficient and control cells. Finally, persistent fibroblasts in Eng(+/-) mice wound co-localize with a greater degree of Akt phosphorylation. Thus, endoglin haploinsufficiency seems to promote fibroblast accumulation during wound healing through the activation of the PI3K/Akt pathway. These studies open new non-Smad signaling pathway for endoglin regulating fibroblast cell function during wound healing, as new therapeutic opportunities for the treatment of fibrotic wounds.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Proteínas Proto-Oncogénicas c-akt/metabolismo , Cicatrización de Heridas/fisiología , Animales , Cicatriz/metabolismo , Cicatriz/patología , Endoglina , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/fisiología , Haploinsuficiencia , Heterocigoto , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Transducción de Señal , Piel/lesiones , Piel/metabolismo , Piel/patología , Activación Transcripcional
2.
PLoS One ; 8(1): e53624, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23341958

RESUMEN

The functional mechanisms involved in angiogenesis and the potential role of endoglin (ENG), recently described as a new marker for this process, have not been explored in Myelodysplastic Syndromes (MDS). In order to gain insight in MDS angiogenesis a combined analysis in bone marrow (BM) of gene expression levels, angiogenesis-related soluble factors and functional angiogenesis-related studies was carried out. Ninety-seven MDS patients and forty-two normal BM samples were studied. The morphology of the capillary-like structures originated by two endothelial cells lines in the BM environment of patients with refractory cytopenia with multilineage dysplasia (RCMD) was different from those of the remaining MDS. In addition, the BM mononuclear cells from RCMD patients displayed over-expression of VEGF, HIF and FN1 while they showed reduced expression of ENG in contrast to the normal ENG expression of the remaining low-risk MDS and the high expression of ENG in high-risk MDS subtype. Moreover, higher soluble ENG and soluble FLT-1 levels in BM microenvironment were observed in RCMD cases, which distinguished them from other individuals. Therefore, the present study suggests that the patterns of angiogenesis are different between the MDS subtypes. The differences in angiogenesis observed in RCMD patients could be related to ENG abnormalities.


Asunto(s)
Células Endoteliales/metabolismo , Síndromes Mielodisplásicos/patología , Neovascularización Patológica/patología , Anemia Refractaria/metabolismo , Anemia Refractaria/patología , Inductores de la Angiogénesis/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Médula Ósea/metabolismo , Médula Ósea/patología , Linaje de la Célula , Proliferación Celular , Microambiente Celular , Endoglina , Células Endoteliales/patología , Humanos , Síndromes Mielodisplásicos/metabolismo , Neovascularización Patológica/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Solubilidad , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Circ Res ; 103(12): 1383-92, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18974388

RESUMEN

Senescence of endothelial cells (ECs) may contribute to age-associated cardiovascular diseases, including atherosclerosis and hypertension. The functional and gene expression changes associated with cellular senescence are poorly understood. Here, we have analyzed the expression, during EC senescence, of 2 different isoforms (L, long; S, short) of endoglin, an auxiliary transforming growth factor (TGF)-beta receptor involved in vascular remodeling and angiogenesis. As evidenced by RT-PCR, the S/L ratio of endoglin isoforms was increased during senescence of human ECs in vitro, as well as during aging of mice in vascularized tissues. Next, the effect of S-endoglin protein on the TGF-beta receptor complex was studied. As revealed by coimmunoprecipitation assays, S-endoglin was able to interact with both TGF-beta type I receptors, ALK5 and ALK1, although the interaction with ALK5 was stronger than with ALK1. S-endoglin conferred a lower proliferation rate to ECs and behaved differently from L-endoglin in relation to TGF-beta-responsive reporters with ALK1 or ALK5 specificities, mimicking the behavior of the endothelial senescence markers Id1 and plasminogen activator inhibitor-1. In situ hybridization studies demonstrated the expression of S-endoglin in the endothelium from human arteries. Transgenic mice overexpressing S-endoglin in ECs showed hypertension, decreased hypertensive response to NO inhibition, decreased vasodilatory response to TGF-beta(1) administration, and decreased endothelial nitric oxide synthase expression in lungs and kidneys, supporting the involvement of S-endoglin in the NO-dependent vascular homeostasis. Taken together, these results suggest that S-endoglin is induced during endothelial senescence and may contribute to age-dependent vascular pathology.


Asunto(s)
Antígenos CD/biosíntesis , Senescencia Celular/fisiología , Células Endoteliales/fisiología , Endotelio Vascular/patología , Endotelio Vascular/fisiología , Regulación de la Expresión Génica/fisiología , Receptores de Superficie Celular/biosíntesis , Animales , Antígenos CD/genética , Antígenos CD/fisiología , Células COS , Células Cultivadas , Chlorocebus aethiops , Endoglina , Células Endoteliales/patología , Humanos , Ratones , Ratones Transgénicos , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Enfermedades Vasculares/sangre , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología
4.
Nephrol Dial Transplant ; 23(11): 3403-11, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18567890

RESUMEN

BACKGROUND: Mesangial activation occurs in many forms of renal disease that progress to renal failure. Mitogen-activated protein kinases (MAPKs) are important mediators involved in the intracellular network of interacting proteins that transduce extracellular stimuli to intracellular responses. The extracellular signal-regulated kinases 5 (Erk5) MAPK pathway has been involved in regulating several cellular responses. Thus, we examined the expression of Erk5 in human renal tissue and the function of Erk5 in cultured human mesangial cells. METHODS: Erk5 was visualized in human renal tissue by immunohistochemistry and in mesangial cells by immunofluorescence microscopy using the anti-Erk5 C-terminus antibody. Erk5 expression and activation, and collagen I expression were determined by western blot. To generate a dominant-negative form of the Erk5 in human mesangial cells, an EcoRI fragment from wild-type pCEFL-HA-Erk5 was subcloned into the EcoRI site of pCDNA3. Cell proliferation was analysed by an MTT-based assay. Cell contraction was analysed by studying the changes in the planar cell surface area. RESULTS: Erk5 was expressed in the kidney, mainly localized at the glomerular mesangium. In cultured human mesangial cells, Erk5 was activated by foetal calf serum (FCS), high glucose, endothelin-1, platelet-activating factor (PAF), epidermal growth factor (EGF) and transforming growth factor beta-1 (TGF-beta1). The expression of a dominant-negative form of Erk5 in human mesangial cells resulted in a significant decrease in proliferation, EGF-induced cell contraction and TGF-beta1-induced collagen I expression. CONCLUSIONS: These results suggest that Erk5 is involved in agonist-induced mesangial cell contraction, proliferation and ECM accumulation and point to a multifunctional role of Erk5 in the pathophysiology of glomerular mesangial cells.


Asunto(s)
Proliferación Celular , MAP Quinasa Quinasa 5/metabolismo , Células Mesangiales/metabolismo , Células Cultivadas , Colágeno Tipo I/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Matriz Extracelular/metabolismo , Humanos , Células Mesangiales/citología , Células Mesangiales/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología
5.
J Cell Sci ; 121(Pt 6): 913-9, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18303046

RESUMEN

TGFbeta regulates cellular processes by binding to type I and type II TGFbeta receptors (TbetaRI and TbetaRII, respectively). In addition to these signaling receptors, endoglin is an accessory TGFbeta receptor that regulates TGFbeta signaling. Although there are two different alternatively spliced isoforms of endoglin, L-endoglin (L, long) and S-endoglin (S, short), little is known about the effects of S-endoglin isoform on TGFbeta signaling. Here, we have analyzed the TGFbeta1 signaling pathways and the effects of L- and S-endoglin in endoglin-deficient L6E9 cells. We found that TGFbeta activates two distinct TbetaRI-Smad signaling pathways: ALK1-Smad1-Id1 and ALK5-Smad2-PAI1, in these cells. Interestingly, L-endoglin enhanced the ALK1-Id1 pathway, while S-endoglin promoted the ALK5-PAI1 route. These effects on signaling are supported by biological effects on TGFbeta1-induced collagen I expression and inhibition of cell proliferation. Thus, while L-endoglin decreased TGFbeta1-induced collagen I and CTGF expression and increased TGFbeta1-induced proliferation, S-endoglin strongly increased TGFbeta1-induced collagen I and CTGF expression, and reduced TGFbeta1-induced cell proliferation.


Asunto(s)
Receptores de Activinas/metabolismo , Antígenos CD/metabolismo , Mioblastos/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/farmacología , Animales , Línea Celular , Proliferación Celular , Colágeno Tipo I/metabolismo , Endoglina , Humanos , Mioblastos/efectos de los fármacos , Proteínas Serina-Treonina Quinasas , Ratas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Smad/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...