Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros











Intervalo de año de publicación
1.
PLoS One ; 7(12): e51245, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23251467

RESUMEN

BACKGROUND: Endothelial cell junctions control blood vessel permeability. Altered permeability can be associated with vascular fragility that leads to vessel weakness and haemorrhage formation. In vivo studies on the function of genes involved in the maintenance of vascular integrity are essential to better understand the molecular basis of diseases linked to permeability defects. Ve-ptp (Vascular Endothelial-Protein Tyrosine Phosphatase) is a transmembrane protein present at endothelial adherens junctions (AJs). METHODOLOGY/PRINCIPAL FINDINGS: We investigated the role of Ve-ptp in AJ maturation/stability and in the modulation of endothelial permeability using zebrafish (Danio rerio). Whole-mount in situ hybridizations revealed zve-ptp expression exclusively in the developing vascular system. Generation of altered zve-ptp transcripts, induced separately by two different splicing morpholinos, resulted in permeability defects closely linked to vascular wall fragility. The ultrastructural analysis revealed a statistically significant reduction of junction complexes and the presence of immature AJs in zve-ptp morphants but not in control embryos. CONCLUSIONS/SIGNIFICANCE: Here we show the first in vivo evidence of a potentially critical role played by Ve-ptp in AJ maturation, an important event for permeability modulation and for the development of a functional vascular system.


Asunto(s)
Uniones Adherentes/fisiología , Vasos Sanguíneos/fisiología , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Uniones Adherentes/enzimología , Animales , Secuencia de Bases , Vasos Sanguíneos/enzimología , Western Blotting , Células Cultivadas , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Humanos , Hibridación in Situ , Neovascularización Fisiológica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Pez Cebra
2.
Arterioscler Thromb Vasc Biol ; 32(9): e104-16, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22837470

RESUMEN

OBJECTIVE: Endothelial sphingosine-1-phosphate (S1P) receptor-1 (S1P(1)) affects different vascular functions, including blood vessel maturation and permeability. Here, we characterized the role of the zS1P(1) ortholog in vascular development in zebrafish. METHODS AND RESULTS: zS1P(1) is expressed in dorsal aorta and posterior cardinal vein of zebrafish embryos at 24 to 30 hours postfertilization. zS1P(1) downregulation by antisense morpholino oligonucleotide injection causes early pericardial edema, lack of blood circulation, alterations of posterior cardinal vein structure, and late generalized edema. Also, zS1P(1) morphants are characterized by downregulation of vascular endothelial cadherin (VE-cadherin) and Eph receptor EphB4a expression and by disorganization of zonula occludens 1 junctions in posterior cardinal vein endothelium, with no alterations of dorsal aorta endothelium. VE-cadherin knockdown results in similar vascular alterations, whereas VE-cadherin overexpression is sufficient to rescue venous vascular integrity defects and EphB4a downregulation in zS1P(1) morphants. Finally, S1P(1) small interfering RNA transfection and the S1P(1) antagonist (R)-3-amino-(3-hexylphenylamino)-4-oxobutylphosphonic acid (W146) cause EPHB4 receptor down-modulation in human umbilical vein endothelial cells and the assembly of zonula occludens 1 intercellular contacts is prevented by the EPHB4 antagonist TNYL-RAW peptide in these cells. CONCLUSIONS: The data demonstrate a nonredundant role of zS1P(1) in the regulation of venous endothelial barrier in zebrafish and identify a S1P(1)/VE-cadherin/EphB4a genetic pathway that controls venous vascular integrity.


Asunto(s)
Permeabilidad Capilar , Células Endoteliales/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Venas/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Anilidas/farmacología , Animales , Animales Modificados Genéticamente , Antígenos CD/metabolismo , Células CHO , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Cricetinae , Cricetulus , Células Endoteliales/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Morfolinos/metabolismo , Oligonucleótidos Antisentido/metabolismo , Organofosfonatos/farmacología , Fosfoproteínas/metabolismo , Interferencia de ARN , Receptor EphB4/metabolismo , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/genética , Receptores de Esfingosina-1-Fosfato , Uniones Estrechas/metabolismo , Transfección , Venas/efectos de los fármacos , Venas/embriología , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética , Proteína de la Zonula Occludens-1
3.
Arterioscler Thromb Vasc Biol ; 32(7): 1563-72, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22516065

RESUMEN

OBJECTIVE: The goal of this study was to determine the in vivo functions of the synaptic proteins neurexins and neuroligins in embryonic vascular system development using zebrafish as animal model. METHODS AND RESULTS: In the present study, we show that the knockdown of the α-form of neurexin 1a induces balance defects and reduced locomotory activity, whereas ß-neurexin 1a and neuroligin 1 morphants present defects in sprouting angiogenesis and vascular remodeling, in particular in the caudal plexus and subintestinal vessels. Coinjection of low doses of morpholinos for ß-neurexin 1a and neuroligin 1 together or in combination with morpholinos targeting the -heparin--binding isoforms of vascular endothelial growth factor A (encoded by the VEGFAb gene) recapitulates the observed abnormalities, suggesting synergistic activity of these molecules. Similar coinjection experiments with morpholinos, targeting the enzyme heparan sulfate 6-O-sulfotransferase 2, confirm the presence of a functional correlation between extracellular matrix maturation and ß-neurexin 1a or neuroligin 1. CONCLUSIONS: Our data represent the first in vivo evidence of the role of neurexin and neuroligin in embryonic blood vessel formation and provide insights into their mechanism of action.


Asunto(s)
Vasos Sanguíneos/embriología , Moléculas de Adhesión Celular Neuronal/fisiología , Glicoproteínas/fisiología , Heparina/metabolismo , Neovascularización Fisiológica , Neuropéptidos/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Pez Cebra/embriología , Animales , Matriz Extracelular/fisiología , Sulfotransferasas/fisiología
4.
PLoS One ; 5(12): e14296, 2010 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-21179188

RESUMEN

BACKGROUND: Notch signaling is an evolutionarily conserved regulatory circuitry implicated in cell fate determination in various developmental processes including hematopoietic stem cell self-renewal and differentiation of blood lineages. Known endogenous inhibitors of Notch activity are Numb-Nb and Numblike-Nbl, which play partially redundant functions in specifying and maintaining neuronal differentiation. Nb and Nbl are expressed in most tissues including embryonic and adult hematopoietic tissues in mice and humans, suggesting possible roles for these proteins in hematopoiesis. METHODOLOGY AND PRINCIPAL FINDINGS: We employed zebrafish to investigate the possible functional role of Numb and Numblike during hematopoiesis, as this system allows a detailed analysis even in embryos with severe defects that would be lethal in other organisms. Here we describe that nb/nbl knockdown results in severe reduction or absence of embryonic erythrocytes in zebrafish. Interestingly, nb/nbl knocked-down embryos present severe downregulation of the erythroid transcription factor gata1. This results in erythroblasts which fail to mature and undergo apoptosis. Our results indicate that Notch activity is increased in embryos injected with nb/nbl morpholino, and we show that inhibition of Notch activation can partially rescue the hematopoietic phenotype. CONCLUSIONS AND SIGNIFICANCE: Our results provide the first in vivo evidence of an involvement of Numb and Numblike in zebrafish erythroid differentiation during primitive hematopoiesis. Furthermore, we found that, at least in part, the nb/nbl morphant phenotype is due to enhanced Notch activation within hematopoietic districts, which in turn results in primitive erythroid differentiation defects.


Asunto(s)
Eritrocitos/citología , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Proteínas de Pez Cebra/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Relación Dosis-Respuesta a Droga , Evolución Molecular , Hematopoyesis , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Fenotipo , Plásmidos/metabolismo , Biosíntesis de Proteínas , Receptores Notch/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA