Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Stem Cell ; 3(6): 625-36, 2008 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19041779

RESUMEN

Hematopoietic stem cells (HSCs) originate within the aortic-gonado-mesonephros (AGM) region of the midgestation embryo, but the cell type responsible for their emergence is unknown since critical hematopoietic factors are expressed in both the AGM endothelium and its underlying mesenchyme. Here we employ a temporally restricted genetic tracing strategy to selectively label the endothelium, and separately its underlying mesenchyme, during AGM development. Lineage tracing endothelium, via an inducible VE-cadherin Cre line, reveals that the endothelium is capable of HSC emergence. The endothelial progeny migrate to the fetal liver, and later to the bone marrow, and are capable of expansion, self-renewal, and multilineage hematopoietic differentiation. HSC capacity is exclusively endothelial, as ex vivo analyses demonstrate lack of VE-cadherin Cre induction in circulating and fetal liver hematopoietic populations. Moreover, AGM mesenchyme, as selectively traced via a myocardin Cre line, is incapable of hematopoiesis. Our genetic tracing strategy therefore reveals an endothelial origin of HSCs.


Asunto(s)
Linaje de la Célula/genética , Desarrollo Embrionario/genética , Células Endoteliales/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Diferenciación Celular/genética , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Células Endoteliales/citología , Regulación del Desarrollo de la Expresión Génica/genética , Estratos Germinativos/embriología , Integrasas/metabolismo , Mesodermo/fisiología , Ratones , Ratones Transgénicos , Biología Molecular/métodos , Coloración y Etiquetado/métodos
2.
Hepatology ; 45(5): 1218-28, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17464995

RESUMEN

UNLABELLED: Liver neo-angiogenesis plays a fundamental role in physiological and pathological processes such as regeneration, cirrhosis, autoimmune hepatitis, and alcoholic liver disease. How liver parenchymal cells influence angiogenesis is largely unknown. We studied the influence of soluble factors released by hepatocytes on hematopoietic and endothelial cell differentiation. Human CD34+ cells cultured for several weeks in a hepatocyte-conditioned medium gradually decrease the expression of CD34 and CD133 markers (i.e. after 4 weeks from 85% and 69%, respectively, to 6% and 3%, respectively), whereas expression of CD144 and CD14 cell markers increased (from 2% and 8%, respectively, to 54% and 55%, respectively). The cells' capacity to form hematopoietic colonies in methylcellulose declined with time, whereas they acquired endothelial morphology, expressed endothelial markers, and incorporated into newly forming vascular structures both in vitro and in vivo. Cultured single CD34+ cells formed colonies expressing both hematopoietic (CD45+) and endothelial (CD144+) markers, suggesting they constitute a bona fide hemangioblast population. CONCLUSION: This system allowed subsequent stages of differentiation of hematopoietic cells to endothelial cells to be defined, underlining the strict interrelationship between endothelial and hematopoietic cells in a hepatocyte environment.


Asunto(s)
Diferenciación Celular/fisiología , Medios de Cultivo Condicionados/farmacología , Endotelio Vascular/citología , Células Madre Hematopoyéticas/citología , Hepatocitos/fisiología , Antígenos CD/análisis , Antígenos CD34/análisis , Cadherinas/análisis , Proliferación Celular , Células Cultivadas , Citometría de Flujo , Humanos , Antígenos Comunes de Leucocito/análisis
3.
J Exp Med ; 201(11): 1825-35, 2005 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-15928198

RESUMEN

The regulation of acetylation is central for the epigenetic control of lineage-specific gene expression and determines cell fate decisions. We provide evidence that the inhibition of histone deacetylases (HDACs) blocks the endothelial differentiation of adult progenitor cells. To define the mechanisms by which HDAC inhibition prevents endothelial differentiation, we determined the expression of homeobox transcription factors and demonstrated that HoxA9 expression is down-regulated by HDAC inhibitors. The causal involvement of HoxA9 in the endothelial differentiation of adult progenitor cells is supported by the finding that HoxA9 overexpression partially rescued the endothelial differentiation blockade induced by HDAC inhibitors. Knockdown and overexpression studies revealed that HoxA9 acts as a master switch to regulate the expression of prototypical endothelial-committed genes such as endothelial nitric oxide synthase, VEGF-R2, and VE-cadherin, and mediates the shear stress-induced maturation of endothelial cells. Consistently, HoxA9-deficient mice exhibited lower numbers of endothelial progenitor cells and showed an impaired postnatal neovascularization capacity after the induction of ischemia. Thus, HoxA9 is regulated by HDACs and is critical for postnatal neovascularization.


Asunto(s)
Diferenciación Celular/fisiología , Células Endoteliales/fisiología , Regulación de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/fisiología , Histona Desacetilasas/metabolismo , Proteínas de Homeodominio/biosíntesis , Animales , Antígenos CD , Cadherinas/metabolismo , Células Cultivadas , Células Endoteliales/citología , Sangre Fetal/citología , Sangre Fetal/fisiología , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/genética , Humanos , Isquemia/metabolismo , Ratones , Ratones Noqueados , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Estrés Mecánico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
4.
Thromb Haemost ; 93(6): 1041-6, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15968386

RESUMEN

The pathogenesis of vascular tumors such as angiosarcomas is poorly understood. Cadherin expression inversely correlates with tumor malignancy and the endothelial specific VE-cadherin is low or absent in angiosarcomas, suggesting an inhibitory role for this protein in tumor progression. In this paper we report that PmyT VE-cadherin null (VEC null) endothelial cells form larger vascular tumors in nude mice when injected subcutaneously as compared to isogenic VE-cadherin positive (VEC pos) cells. This effect requires the association of beta-catenin to VEcadherin, since a VE-cadherin mutant lacking the domain responsible for beta-catenin binding (Deltabetacat) cannot rescue the phenotype. In VEC null cells beta-catenin is phosphorylated and partly degraded. N-cadherin is increased and detected at junctions. VEC null cells also present an altered fibrinolytic activity with increases in tPA, uPA, uPAR and a strong reduction in PAI-1, which may be correlated to the high incidence of abrupt hemorrhages in VEC null tumors. Overall, these data strongly suggest that downregulation of VE-cadherin in endothelial tumors may have important consequences for tumor growth and bleeding complications.


Asunto(s)
Cadherinas/genética , Hemangiosarcoma/etiología , Hemorragia/etiología , Animales , Antígenos CD , Cadherinas/fisiología , Línea Celular , Regulación hacia Abajo , Endotelio Vascular/fisiopatología , Fibrinólisis , Hemangiosarcoma/genética , Hemangiosarcoma/patología , Hemangiosarcoma/fisiopatología , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Inhibidor 1 de Activador Plasminogénico/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 25(4): 692-7, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15662021

RESUMEN

OBJECTIVE: To test the potential of mesoangioblasts (Mabs) in reducing postischemic injury in comparison with bone marrow progenitor cells (BMPCs), fibroblasts (Fbs), and embryonic stem cell-derived endothelial cells (ECs), and to identify putative cellular protective mechanisms. METHODS AND RESULTS: Cells were injected percutaneously in the left ventricular (LV) chamber of C57BL/6 mice, 3 to 6 hours after coronary ligation, and detected in the hearts 2 days and 6 weeks later. Echocardiographic examinations were performed at 6 weeks. LV dilation was reduced and LV shortening fraction was improved with Mabs and BMPCs but not with ECs and Fbs. Donor cell colonization of the host myocardium was modest and predominantly in the smooth muscle layer of vessels. Capillary density was higher in the peripheral infarct area and apoptotic cardiomyocytes were fewer with Mabs and BMPCs. Mabs and BMPCs, but not Fbs or ECs, promoted survival of cultured cardiocytes under low-oxygen in culture. This activity was present in Mab-conditioned medium and could be replaced by a combination of basic fibroblast growth factor (bFGF), insulin-like growth factor (IGF)-1, and hepatocyte growth factor (HGF), all of which are produced by these cells. Conditioned medium from Mabs, but not from Fbs, stimulated proliferation of smooth muscle cells in vitro. CONCLUSIONS: Mabs appear as effective as BMPCs in reducing postinfarction LV dysfunction, likely through production of antiapoptotic and angiogenic factors.


Asunto(s)
Endotelio Vascular/trasplante , Fibroblastos/trasplante , Células Madre Hematopoyéticas/citología , Células Madre Multipotentes/citología , Infarto del Miocardio/terapia , Trasplante de Células Madre , Animales , Apoptosis , Vasos Sanguíneos/citología , Ecocardiografía , Endotelio Vascular/citología , Fibroblastos/citología , Supervivencia de Injerto , Células Madre Hematopoyéticas/fisiología , Mesodermo/citología , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/fisiología , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/fisiopatología , Miocardio/citología , Neovascularización Fisiológica , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
6.
Blood ; 105(7): 2771-6, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15604224

RESUMEN

We investigated the role of vascular endothelial (VE)-cadherin in blood vessel morphogenesis and established a temporal correlation linking the failure in vessel morphogenesis in VE-cadherin null embryos to a specific step in vasculogenesis. We showed that the sequence in which blood vessels failed followed the order in which they had formed (ie, those forming first--yolk sac, allantoic and endocardial vessels--were the first to display morphologic abnormalities). We next showed that in place of normal reticulated networks of blood vessels, clusters of platelet endothelial cell adhesion molecule-positive (PECAM+) cells formed within cultured allantois explants from VE-cadherin null embryos. Similarly, a function-blocking VE-cadherin antibody, BV13, caused PECAM+ cell clusters to form in cultured allantois explants from normal mice. Finally, we demonstrated that formation of PECAM+ cell clusters in response to BV13 was not due to a disruption in the formation of nascent vessels but was due to the actual disassembly of nascent vessels. Based on these findings, we conclude that the events of de novo blood vessel formation up to the point at which a vascular epithelium forms (ie, nascent vessels with lumens) are not dependent on VE-cadherin and that VE-cadherin, whose expression is up-regulated following vascular epithelialization, is required to prevent the disassembly of nascent blood vessels.


Asunto(s)
Vasos Sanguíneos/embriología , Vasos Sanguíneos/fisiología , Cadherinas/genética , Cadherinas/metabolismo , Neovascularización Fisiológica/fisiología , Animales , Anticuerpos Monoclonales , Antígenos CD , Cadherinas/inmunología , Membrana Corioalantoides/embriología , Membrana Corioalantoides/fisiología , Endotelio/fisiología , Regulación del Desarrollo de la Expresión Génica , Mesodermo/fisiología , Ratones , Técnicas de Cultivo de Órganos , Saco Vitelino/embriología , Saco Vitelino/fisiología
7.
Am J Pathol ; 165(1): 181-9, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15215174

RESUMEN

Survivin is strongly expressed in embryonic organs and in tumor cells but is low or absent in differentiated normal tissues. Resting endothelium expresses low levels of survivin but can up-regulate its synthesis on activation to proliferate. The mechanisms responsible for survivin down-regulation in resting conditions are still unknown. We report here that confluence and vascular endothelial-cadherin (VE-cadherin) expression induce contact inhibition of cell growth and survivin down-regulation in the endothelium. Using beta-catenin null and positive isogenic endothelial cell lines we found that the effect requires beta-catenin expression and its association to VE-cadherin cytoplasmic tail. Furthermore, in allantois organ cultures, survivin expression is up-regulated in areas of growing vessels where VE-cadherin is partially dismantled from junctions or in VE-cadherin -/- specimens. Overall, these data indicate that VE-cadherin and beta-catenin may negatively regulate survivin synthesis in endothelial cells. Consistently, in epidermal and pancreatic cell lines or ovarian tumors, epithelial-cadherin (E-cadherin) and survivin expression is inversely related, suggesting a non-cell-specific role of cadherins in reducing survivin synthesis.


Asunto(s)
Cadherinas/metabolismo , Endotelio Vascular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Alantoides/citología , Animales , Antígenos CD , Western Blotting , Cadherinas/genética , División Celular , Línea Celular , Línea Celular Tumoral , Análisis por Conglomerados , Proteínas del Citoesqueleto , Regulación hacia Abajo , Embrión de Mamíferos , Endotelio Vascular/citología , Fibronectinas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes , Humanos , Proteínas Inhibidoras de la Apoptosis , Proteínas Luminiscentes , Ratones , Ratones Noqueados , Microscopía Confocal , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias , Neovascularización Fisiológica/fisiología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Células Madre/citología , Survivin , Transactivadores , Venas Umbilicales/citología , Regulación hacia Arriba , beta Catenina
8.
Blood ; 103(8): 3005-12, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15070677

RESUMEN

The junctional membrane protein vascular endothelial (VE)-cadherin mediates contact inhibition of growth and inhibits apoptosis of endothelial cells. In this article we show that VE-cadherin induces expression of growth arrest-specific 1 (Gas1), an integral membrane protein up-regulated in nonproliferating cells. By comparing syngenic endothelial cell lines, we found that Gas1 mRNA was increased by 3-fold in VE-cadherin-positive cells in comparison to VE-cadherin-null cells. Ectopic expression of Gas1 in endothelial or 293 cells strongly reduced apoptosis without affecting cell growth. Addition of vascular endothelial growth factor (VEGF) also up-regulated Gas1 and this effect was augmented more so in confluent nonproliferating cells than in sparse cultures. VE-cadherin-blocking antibody partially inhibited VEGF-induced Gas1, suggesting that VE-cadherin clustering is required for an optimal response to this stimulus. Inhibition of phosphoinositole-3-OH kinase (PI3-kinase) pathway by Wortmannin prevented Gas1 synthesis and the antiapoptotic effect of VEGF, but, in cells ectopically expressing Gas1, Wortmannin was ineffective. Furthermore, inhibition of Gas1 expression by short interfering RNA (siRNA) both in vitro and in allantois organ cultures made endothelial cells refractory to the antiapoptotic effect of VEGF. Overall these data indicate that Gas1 induction by VE-cadherin and VEGF in endothelial cells requires activation of PI3-kinase. Gas1 expression positively correlates with inhibition of endothelial cell apoptosis and may contribute to the integrity of resting endothelium.


Asunto(s)
Cadherinas/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Proteínas de la Membrana/biosíntesis , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Antígenos CD , Apoptosis/efectos de los fármacos , Cadherinas/genética , Proteínas de Ciclo Celular , Línea Celular , Endotelio Vascular/efectos de los fármacos , Activación Enzimática , Proteínas Ligadas a GPI , Humanos , Proteínas de la Membrana/genética , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo
9.
Exp Cell Res ; 290(2): 207-16, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14567980

RESUMEN

We previously showed that clonable skeletal myogenic cells can be derived from the embryonic aorta but become very rare in the more mature and structured fetal aorta. The aim of this study was to investigate whether, during fetal and postnatal development, these myogenic progenitors progressively disappear or may rather associate with the microvascular district, being thus distributed to virtually all tissues. To test this hypothesis, we used F1 embryos (or mice) from a transgenic line expressing a striated muscle-specific reporter gene (LacZ) crossed with a transgenic line expressing a different endothelial-specific reporter genes (GFP). Endothelial cells were isolated from yolk sac (at E11) and lung (at E11, E17, P1, P10, and P60), two organs embryologically unrelated to paraxial mesoderm, rich in vessels, and devoid of skeletal muscle. Endothelial cells, purified by magnetic bead selection (CD31/PECAM-1(+)) or cell sorting (Tie2-GFP(+)) were then challenged for their skeletal myogenic potential in vitro and in vivo. The results demonstrated that both yolk sac and lung contain progenitor cells, which express endothelial markers and are endowed with a skeletal myogenic potential that they reveal when in the presence of differentiating myoblasts, in vitro, and regenerating muscle, in vivo. The number (or potency to generate skeletal muscle) of these vessels associated cells decreases rapidly with age and is very low in mature animals, possibly correlating with reduced regenerative capacity of adult mammalian tissues.


Asunto(s)
Endotelio Vascular/citología , Pulmón/embriología , Músculo Esquelético/citología , Células Madre/citología , Saco Vitelino/embriología , Envejecimiento , Animales , Animales Recién Nacidos , Biomarcadores/análisis , Línea Celular , Endotelio Vascular/metabolismo , Femenino , Separación Inmunomagnética , Pulmón/metabolismo , Ratones , Ratones SCID , Ratones Transgénicos , Desarrollo de Músculos , Músculo Esquelético/metabolismo , Técnicas de Cultivo de Órganos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Saco Vitelino/metabolismo
10.
Blood ; 100(3): 905-11, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12130501

RESUMEN

Vascular endothelial cadherin (VE-cadherin) is an endothelial-specific, trans-membrane protein that promotes homophilic cell adhesion. Inhibition of VE-cadherin by the blocking monoclonal antibody (mAb) BV13 inhibited angiogenesis and tumor growth in vivo. However, this effect was accompanied by a marked increase in lung and heart permeability. In the present paper, we characterize a different VE-cadherin mAb (BV14) that is able to inhibit angiogenesis without affecting vascular permeability. In vitro studies show that BV14, in contrast to BV13, did not increase paracellular permeability of endothelial monolayers and did not disrupt VE-cadherin clusters at junctions. However, both antibodies could inhibit formation of vascularlike structures in collagen gels and increase migration of endothelial cells into wounded areas. In vivo, BV14 and BV13 were equally active in inhibiting angiogenesis in the mouse cornea and in reducing the growth of hemangioma and C6 glioma. In contrast to BV13, BV14 did not change vascular permeability in all the organs tested and at any dose used. BV14 and BV13 bind to VE-cadherin extracellular repeats EC4 and EC1, respectively. We propose that, in resting vessels, where junctions are stable and well-structured, antibody binding to EC1 but not EC4 disrupts their organization and increases permeability. In contrast, in growing vessels, where endothelial cells are migrating and junctions are weaker, antibody binding to EC4 may be sufficient to disrupt cell-to-cell adhesion and inhibit assembly of new vascular structures.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Cadherinas/fisiología , Permeabilidad Capilar/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Neovascularización Patológica/prevención & control , Animales , Antígenos CD , Antineoplásicos/farmacología , Cadherinas/inmunología , Cadherinas/metabolismo , División Celular/efectos de los fármacos , Endotelio Vascular/química , Endotelio Vascular/citología , Mapeo Epitopo , Uniones Intercelulares/efectos de los fármacos , Ratones , Ratones Noqueados , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neovascularización Patológica/tratamiento farmacológico , Células Tumorales Cultivadas/trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...