Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4405, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38782923

RESUMEN

Zonula occludens-1 (ZO-1) is involved in the regulation of cell-cell junctions between endothelial cells (ECs). Here we identify the ZO-1 protein interactome and uncover ZO-1 interactions with RNA-binding proteins that are part of stress granules (SGs). Downregulation of ZO-1 increased SG formation in response to stress and protected ECs from cellular insults. The ZO-1 interactome uncovered an association between ZO-1 and Y-box binding protein 1 (YB-1), a constituent of SGs. Arsenite treatment of ECs decreased the interaction between ZO-1 and YB-1, and drove SG assembly. YB-1 expression is essential for SG formation and for the cytoprotective effects induced by ZO-1 downregulation. In the developing retinal vascular plexus of newborn mice, ECs at the front of growing vessels express less ZO-1 but display more YB-1-positive granules than ECs located in the vascular plexus. Endothelial-specific deletion of ZO-1 in mice at post-natal day 7 markedly increased the presence of YB-1-positive granules in ECs of retinal blood vessels, altered tip EC morphology and vascular patterning, resulting in aberrant endothelial proliferation, and arrest in the expansion of the retinal vasculature. Our findings suggest that, through its interaction with YB-1, ZO-1 controls SG formation and the response of ECs to stress during angiogenesis.


Asunto(s)
Células Endoteliales , Proteína 1 de Unión a la Caja Y , Proteína de la Zonula Occludens-1 , Animales , Proteína 1 de Unión a la Caja Y/metabolismo , Proteína 1 de Unión a la Caja Y/genética , Proteína de la Zonula Occludens-1/metabolismo , Proteína de la Zonula Occludens-1/genética , Ratones , Humanos , Células Endoteliales/metabolismo , Gránulos de Estrés/metabolismo , Neovascularización Fisiológica , Vasos Retinianos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Angiogénesis , Factores de Transcripción
2.
Cell Mol Life Sci ; 79(1): 37, 2021 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-34971428

RESUMEN

The roles of nitric oxide (NO) and endothelial NO synthase (eNOS) in the regulation of angiogenesis are well documented. However, the involvement of eNOS in the sprouting of endothelial tip-cells at the vascular front during sprouting angiogenesis remains poorly defined. In this study, we show that downregulation of eNOS markedly inhibits VEGF-stimulated migration of endothelial cells but increases their polarization, as evidenced by the reorientation of the Golgi in migrating monolayers and by the fewer filopodia on tip cells at ends of sprouts in endothelial cell spheroids. The effect of eNOS inhibition on EC polarization was prevented in Par3-depleted cells. Importantly, downregulation of eNOS increased the expression of polarity genes, such as PARD3B, PARD6A, PARD6B, PKCΖ, TJP3, and CRB1 in endothelial cells. In retinas of eNOS knockout mice, vascular development is retarded with decreased vessel density and vascular branching. Furthermore, tip cells at the extremities of the vascular front have a marked reduction in the number of filopodia per cell and are more oriented. In a model of oxygen-induced retinopathy (OIR), eNOS deficient mice are protected during the initial vaso-obliterative phase, have reduced pathological neovascularization, and retinal endothelial tip cells have fewer filopodia. Single-cell RNA sequencing of endothelial cells from OIR retinas revealed enrichment of genes related to cell polarity in the endothelial tip-cell subtype of eNOS deficient mice. These results indicate that inhibition of eNOS alters the polarity program of endothelial cells, which increases cell polarization, regulates sprouting angiogenesis and normalizes pathological neovascularization during retinopathy.


Asunto(s)
Neovascularización Patológica , Neovascularización Fisiológica , Óxido Nítrico Sintasa de Tipo III/fisiología , Retina/metabolismo , Neovascularización Retiniana , Vasos Retinianos , Animales , Bovinos , Línea Celular , Movimiento Celular , Polaridad Celular , Células Endoteliales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/citología , Retina/patología , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Vasos Retinianos/citología , Vasos Retinianos/patología
3.
Cells ; 9(1)2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31936361

RESUMEN

Angiopoietin-1 (Ang-1) is an important proangiogenic factor also involved in the maintenance of endothelial-barrier integrity. The small GTPase Rap1 is involved in the regulation of adherens junctions through VE-cadherin-mediated adhesion, and in endothelial permeability. While many studies established that Rap1 activation is critical for endothelial cell-cell adhesions, its roles in the antipermeability effects of Ang-1 are ill-defined. Thus, we determined the contribution of Rap1 to Ang-1-stimulated angiogenic effects on endothelial cells (ECs). We found that Rap1 is activated following Ang-1 stimulation and is required for the antipermeability effects of Ang-1 on EC monolayers. Our results also revealed that Rap1 is necessary for EC sprouting stimulated by Ang-1 but had no significant effect on Ang-1-induced EC migration and adhesion. In contrast, downregulation of VE-cadherin markedly increased the adhesiveness of ECs to the substratum, which resulted in inhibition of Ang-1-stimulated migration. These results revealed that Rap1 is central to the effects of Ang-1 at intercellular junctions of ECs, whereas VE-cadherin is also involved in the adhesion of ECs to the extracellular matrix.


Asunto(s)
Angiopoyetina 1/farmacología , Aorta/fisiología , Adhesión Celular , Comunicación Celular , Endotelio Vascular/fisiología , Proteínas de Unión al GTP rap1/metabolismo , Animales , Aorta/citología , Aorta/efectos de los fármacos , Bovinos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Transducción de Señal , Proteínas de Unión al GTP rap1/genética
4.
Mol Biol Cell ; 30(17): 2227-2239, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31141452

RESUMEN

Binding of angiopoietin-1 (Ang-1) to its receptor Tie2 on endothelial cells (ECs) promotes vessel barrier integrity and angiogenesis. Here, we identify PAK2 and paxillin as critical targets of Ang-1 responsible for EC migration, polarization, and sprouting. We found that Ang-1 increases PAK2-dependent paxillin phosphorylation and remodeling of focal adhesions and that PAK2 and paxillin are required for EC polarization, migration, and angiogenic sprouting in response to Ang-1. Our findings show that Ang-1 triggers Cdc42 activation at the leading edges of migrating ECs, which is dependent on PAK2 and paxillin expression. We also established that the polarity protein Par3 interacts with Cdc42 in response to Ang-1 in a PAK2- and paxillin-dependent manner. Par3 is recruited at the leading edges of migrating cells and in focal adhesion, where it forms a signaling complex with PAK2 and paxillin in response to Ang-1. These results show that Ang-1 triggers EC polarization and angiogenic sprouting through PAK2-dependent paxillin activation and remodeling of focal adhesions, which are necessary for local activation of Cdc42 and the associated polarity complex. We have shown that PAK2 controls a signaling pathway important for angiogenic sprouting that links focal adhesions to polarity signaling in ECs.


Asunto(s)
Angiopoyetina 1/metabolismo , Paxillin/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Quinasas p21 Activadas/metabolismo , Animales , Aorta/metabolismo , Bovinos , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Adhesiones Focales/metabolismo , Neovascularización Fisiológica , Fosforilación , Receptor TIE-2 , Transducción de Señal , Quinasas p21 Activadas/fisiología
5.
Proc Natl Acad Sci U S A ; 116(10): 4538-4547, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30787185

RESUMEN

Diabetic macular edema is a major complication of diabetes resulting in loss of central vision. Although heightened vessel leakiness has been linked to glial and neuronal-derived factors, relatively little is known on the mechanisms by which mature endothelial cells exit from a quiescent state and compromise barrier function. Here we report that endothelial NOTCH1 signaling in mature diabetic retinas contributes to increased vascular permeability. By providing both human and mouse data, we show that NOTCH1 ligands JAGGED1 and DELTA LIKE-4 are up-regulated secondary to hyperglycemia and activate both canonical and rapid noncanonical NOTCH1 pathways that ultimately disrupt endothelial adherens junctions in diabetic retinas by causing dissociation of vascular endothelial-cadherin from ß-catenin. We further demonstrate that neutralization of NOTCH1 ligands prevents diabetes-induced retinal edema. Collectively, these results identify a fundamental process in diabetes-mediated vascular permeability and provide translational rational for targeting the NOTCH pathway (primarily JAGGED1) in conditions characterized by compromised vascular barrier function.


Asunto(s)
Permeabilidad Capilar , Retinopatía Diabética/patología , Receptor Notch1/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Proteínas de Unión al Calcio/biosíntesis , Activación Enzimática , Hiperglucemia/metabolismo , Proteína Jagged-1/biosíntesis , Ratones , Óxido Nítrico/biosíntesis , Vasos Retinianos/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Familia-src Quinasas/metabolismo
6.
Mol Cell Biol ; 37(12)2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28320874

RESUMEN

Nitric oxide (NO) produced by endothelial NO synthase (eNOS) modulates many functions in endothelial cells. S-nitrosylation (SNO) of cysteine residues on ß-catenin by eNOS-derived NO has been shown to influence intercellular contacts between endothelial cells. However, the implication of SNO in the regulation of ß-catenin transcriptional activity is ill defined. Here, we report that NO inhibits the transcriptional activity of ß-catenin and endothelial cell proliferation induced by activation of Wnt/ß-catenin signaling. Interestingly, induction by Wnt3a of ß-catenin target genes, such as the axin2 gene, is repressed in an eNOS-dependent manner by vascular endothelial growth factor (VEGF). We identified Cys466 of ß-catenin as a target for SNO by eNOS-derived NO and as the critical residue for the repressive effects of NO on ß-catenin transcriptional activity. Furthermore, we observed that Cys466 of ß-catenin, located at the binding interface of the ß-catenin-TCF4 transcriptional complex, is essential for disruption of this complex by NO. Importantly, Cys466 of ß-catenin is necessary for the inhibitory effects of NO on Wnt3a-stimulated proliferation of endothelial cells. Thus, our data define the mechanism responsible for the repressive effects of NO on the transcriptional activity of ß-catenin and link eNOS-derived NO to the modulation by VEGF of Wnt/ß-catenin-induced endothelial cell proliferation.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Proteína Wnt3A/farmacología , beta Catenina/metabolismo , Animales , Bovinos , Proliferación Celular/efectos de los fármacos , Cisteína/metabolismo , Células Endoteliales/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Óxido Nítrico/metabolismo , Nitrosación/efectos de los fármacos , Transcripción Genética , Factor A de Crecimiento Endotelial Vascular/farmacología , Vía de Señalización Wnt
7.
Mol Cell Proteomics ; 15(5): 1511-25, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26846344

RESUMEN

VEGF and angiopoietin-1 (Ang-1) are essential factors to promote angiogenesis through regulation of a plethora of signaling events in endothelial cells (ECs). Although pathways activated by VEGF and Ang-1 are being established, the unique signaling nodes conferring specific responses to each factor remain poorly defined. Thus, we conducted a large-scale comparative phosphoproteomic analysis of signaling pathways activated by VEGF and Ang-1 in ECs using mass spectrometry. Analysis of VEGF and Ang-1 networks of regulated phosphoproteins revealed that the junctional proteins ZO-1, ZO-2, JUP and p120-catenin are part of a cluster of proteins phosphorylated following VEGF stimulation that are linked to MAPK1 activation. Down-regulation of these junctional proteins led to MAPK1 activation and accordingly, increased proliferation of ECs stimulated specifically by VEGF, but not by Ang-1. We identified ZO-1 as the central regulator of this effect and showed that modulation of cellular ZO-1 levels is necessary for EC proliferation during vascular development of the mouse postnatal retina. In conclusion, we uncovered ZO-1 as part of a signaling node activated by VEGF, but not Ang-1, that specifically modulates EC proliferation during angiogenesis.


Asunto(s)
Angiopoyetina 1/metabolismo , Células Endoteliales/citología , Proteómica/métodos , Retina/crecimiento & desarrollo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Animales , Bovinos , Línea Celular , Proliferación Celular , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Espectrometría de Masas/métodos , Ratones , Neovascularización Fisiológica , Fosfoproteínas/metabolismo , Retina/metabolismo , Transducción de Señal
8.
Arterioscler Thromb Vasc Biol ; 32(10): 2484-92, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22859491

RESUMEN

OBJECTIVE: Vascular endothelial growth factor (VEGF) signaling to endothelial NO synthase (eNOS) plays a central role in angiogenesis. In endothelial cells (ECs), heat-shock protein 90 (Hsp90) is also a regulator of eNOS activity. Our study is designed to determine whether modulation of the activator of Hsp90 ATPase 1 (AHA1) regulates the function of Hsp90 in ECs. METHODS AND RESULTS: We show that eNOS phosphorylation on Ser-1179 after VEGF stimulation is significantly reduced in ECs transfected with a small interfering RNA against AHA1. Accordingly, VEGF-stimulated NO production, endothelial permeability, cell migration, and EC invasion in Matrigel implants in mice are reduced in small interfering RNA against AHA1-treated conditions. Furthermore, the induction of eNOS association with Hsp90 after VEGF stimulation is decreased in AHA1-downregulated cells. We also demonstrate that modulation of Hsp90 activity by AHA1 regulates phosphorylation of Hsp90 on Tyr-300. Interestingly, the association of AHA1 with Hsp90 is increased after c-Src-mediated phosphorylation of Hsp90 on Tyr-300. Finally, we show that overexpression of AHA1 in ECs promotes association of eNOS and Hsp90, phosphorylation of Ser-1179 of eNOS, increases NO production, and cell migration. CONCLUSIONS: These results reveal that modulation of Hsp90 activity by AHA1 regulates VEGF signaling to eNOS and angiogenesis.


Asunto(s)
Endotelio Vascular/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células COS , Bovinos , Movimiento Celular/fisiología , Células Cultivadas , Chlorocebus aethiops , Regulación hacia Abajo/fisiología , Endotelio Vascular/citología , Humanos , Técnicas In Vitro , Modelos Animales , Neovascularización Fisiológica/fisiología , Óxido Nítrico/metabolismo , Fosforilación
9.
Mol Cell ; 39(3): 468-76, 2010 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-20705246

RESUMEN

Disruption of adherens junctions between endothelial cells results in compromised endothelial barrier function and in altered angiogenesis. Nitric oxide (NO) produced by endothelial NO synthase (eNOS) is essential for increased vascular permeability induced by vascular endothelial growth factor (VEGF). However, the molecular mechanisms by which NO modulates endothelial permeability remain elusive. Here, we show that, within adherens junctions, beta-catenin is a substrate for S-nitrosylation by NO. Stimulation of endothelial cells with VEGF induces S-nitrosylation of beta-catenin, which is dependent on expression and activity of eNOS. Furthermore, VEGF-induced S-nitrosylation of beta-catenin is inhibited in eNOS(-/-) mice. We identify Cys619, located within the VE-cadherin interaction site, as the major S-nitrosylation locus in response to VEGF. Inhibition of S-nitrosylation at Cys619 prevents NO-dependent dissociation of beta-catenin from VE-cadherin and disassembly of adherens junction complexes and inhibits VEGF-stimulated endothelial permeability. Thus, we identify S-nitrosylation of beta-catenin as a modulator of intercellular contacts between endothelial cells.


Asunto(s)
Permeabilidad Capilar/fisiología , Células Endoteliales/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , beta Catenina/metabolismo , Uniones Adherentes/genética , Uniones Adherentes/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Cisteína/genética , Cisteína/metabolismo , Ratones , Ratones Noqueados , Óxido Nítrico/genética , Óxido Nítrico Sintasa de Tipo III/genética , Factor A de Crecimiento Endotelial Vascular/farmacología , beta Catenina/genética
10.
FASEB J ; 20(2): 340-2, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16322129

RESUMEN

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that acts both as an extracellular ligand for endothelial differentiation gene receptor family and as an intracellular second messenger. Cellular levels of S1P are low and tightly regulated by sphingosine kinase (SPK). Recent studies have suggested that eNOS pathway may function as a downstream target for the biological effects of receptor-mediated S1P. Here we have studied the possible interplay between intracellular SIP generation and the eNOS activation pathway. S1P causes an endothelium-dependent vasorelaxation in rat aorta that is PTX sensitive, inhibited by L-NAME that involves eNOS phosphorylation, and mainly dependent on hsp90. When rat aorta rings were incubated with the SPK inhibitor DL-threo-dihydrosphingosine (DTD), there was a concentration-dependent reduction of Ach-induced vasorelaxation, implying a consistent contribution of sphingolipid pathway through intracellular sphingosine release and phosphorylation. Co-immunoprecipitation experiments consistently showed increased association of hsp90 with eNOS after exposure of cells to S1P as well to BK or calcium ionophore A-23187. Interestingly, as opposite to A-23187, BK and S1P effect were significantly inhibited by pretreatment with the SPK inhibitor DTD. In conclusion, our data demonstrate that an interplay exists among eNOS, hsp90, and intracellularly generated S1P where eNOS coupling to hsp90 is a major determinant for NO release as confirmed by our functional and molecular studies.


Asunto(s)
Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Vasodilatación/fisiología , Animales , Aorta/metabolismo , Calcio/metabolismo , Bovinos , Células Cultivadas , Células Endoteliales/enzimología , Proteínas HSP90 de Choque Térmico/metabolismo , Lisofosfolípidos/metabolismo , Masculino , Ratas , Ratas Wistar , Esfingosina/análogos & derivados , Esfingosina/metabolismo
11.
J Biol Chem ; 278(22): 20091-7, 2003 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-12649282

RESUMEN

Ligand-stimulated degradation of receptor tyrosine kinase (RTK) is an important regulatory step of signal transduction. The vascular endothelial growth factor (VEGF) receptor Flk-1/KDR is responsible for the VEGF-stimulated nitric oxide (NO) production from endothelial cells. Cellular mechanisms mediating the negative regulation of Flk-1 signaling in endothelial cells have not been investigated. Here we show that Flk-1 is rapidly down-regulated following VEGF stimulation of bovine aortic endothelial cells (BAECs). Consequently, VEGF pretreatment of endothelial cells prevents any further stimulation of Flk-1, resulting in decreased NO production from subsequent VEGF challenges. Ubiquitination of RTKs targets them for degradation; we demonstrate that activation of Flk-1 by VEGF leads to its polyubiquitination in BAECs. Furthermore, VEGF stimulation of BAECs or COS-7 cells transiently transfected with Flk-1 results in the phosphorylation of the ubiquitin ligase Cbl, the enhanced association of Cbl with Flk-1, and the relocalization of Cbl to vesicular structures in BAECs. Overexpression of Cbl in COS-7 cells enhances VEGF-induced ubiquitination of Flk-1, whereas a Cbl mutant lacking the ubiquitin ligase RING finger domain, 70Z/3-Cbl, does not. Moreover, expression of Cbl in contrast to 70Z/3-Cbl inhibits the Flk-1-dependent activation of eNOS and, thus, NO release. In BAEC overexpressing Cbl, the degradation of Flk-1 upon VEGF stimulation is accelerated compared with cells transfected with a control vector (green fluorescent protein). Our findings demonstrate that Flk-1 is rapidly down-regulated following sustained VEGF stimulation and identify Cbl as a negative regulator of Flk-1 signaling to eNOS. Cbl thus plays a role in the regulation of VEGF signaling by mediating the stimulated ubiquitination and, consequently, degradation of Flk-1 in endothelial cells.


Asunto(s)
Regulación hacia Abajo/fisiología , Factores de Crecimiento Endotelial/fisiología , Endotelio Vascular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Linfocinas/fisiología , Óxido Nítrico/biosíntesis , Proteínas Oncogénicas de Retroviridae/fisiología , Ubiquitina/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Animales , Bovinos , Células Cultivadas , Endotelio Vascular/citología , Proteína Oncogénica v-cbl , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...