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










Base de datos
Intervalo de año de publicación
1.
Mol Cell Proteomics ; 18(10): 2058-2077, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31427368

RESUMEN

Vascular endothelial protein tyrosine phosphatase (VE-PTP, PTPRB) is a receptor type phosphatase that is crucial for the regulation of endothelial junctions and blood vessel development. We and others have shown recently that VE-PTP regulates vascular integrity by dephosphorylating substrates that are key players in endothelial junction stability, such as the angiopoietin receptor TIE2, the endothelial adherens junction protein VE-cadherin and the vascular endothelial growth factor receptor VEGFR2. Here, we have systematically searched for novel substrates of VE-PTP in endothelial cells by utilizing two approaches. First, we studied changes in the endothelial phosphoproteome on exposing cells to a highly VE-PTP-specific phosphatase inhibitor followed by affinity isolation and mass-spectrometric analysis of phosphorylated proteins by phosphotyrosine-specific antibodies. Second, we used a substrate trapping mutant of VE-PTP to pull down phosphorylated substrates in combination with SILAC-based quantitative mass spectrometry measurements. We identified a set of substrate candidates of VE-PTP, of which a remarkably large fraction (29%) is related to cell junctions. Several of those were found in both screens and displayed very high connectivity in predicted functional interaction networks. The receptor protein tyrosine kinase EPHB4 was the most prominently phosphorylated protein on VE-PTP inhibition among those VE-PTP targets that were identified by both proteomic approaches. Further analysis revealed that EPHB4 forms a ternary complex with VE-PTP and TIE2 in endothelial cells. VE-PTP controls the phosphorylation of each of these two tyrosine kinase receptors. Despite their simultaneous presence in a ternary complex, stimulating each of the receptors with their own specific ligand did not cross-activate the respective partner receptor. Our systematic approach has led to the identification of novel substrates of VE-PTP, of which many are relevant for the control of cellular junctions further promoting the importance of VE-PTP as a key player of junctional signaling.


Asunto(s)
Proteómica/métodos , Receptor EphB4/metabolismo , Receptor TIE-2/metabolismo , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Compuestos de Anilina/farmacología , Cromatografía Liquida , Células Endoteliales , Células Endoteliales de la Vena Umbilical Humana , Humanos , Uniones Intercelulares , Mutación , Fosforilación/efectos de los fármacos , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Receptor EphB4/química , Receptor TIE-2/química , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/química , Especificidad por Sustrato , Ácidos Sulfónicos/farmacología , Espectrometría de Masas en Tándem
2.
J Exp Med ; 208(8): 1721-35, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21788407

RESUMEN

Neutrophil extravasation and the regulation of vascular permeability require dynamic actin rearrangements in the endothelium. In this study, we analyzed in vivo whether these processes require the function of the actin nucleation-promoting factor cortactin. Basal vascular permeability for high molecular weight substances was enhanced in cortactin-deficient mice. Despite this leakiness, neutrophil extravasation in the tumor necrosis factor-stimulated cremaster was inhibited by the loss of cortactin. The permeability defect was caused by reduced levels of activated Rap1 (Ras-related protein 1) in endothelial cells and could be rescued by activating Rap1 via the guanosine triphosphatase (GTPase) exchange factor EPAC (exchange protein directly activated by cAMP). The defect in neutrophil extravasation was caused by enhanced rolling velocity and reduced adhesion in postcapillary venules. Impaired rolling interactions were linked to contributions of ß(2)-integrin ligands, and firm adhesion was compromised by reduced ICAM-1 (intercellular adhesion molecule 1) clustering around neutrophils. A signaling process known to be critical for the formation of ICAM-1-enriched contact areas and for transendothelial migration, the ICAM-1-mediated activation of the GTPase RhoG was blocked in cortactin-deficient endothelial cells. Our results represent the first physiological evidence that cortactin is crucial for orchestrating the molecular events leading to proper endothelial barrier function and leukocyte recruitment in vivo.


Asunto(s)
Actinas/metabolismo , Permeabilidad Capilar/inmunología , Cortactina/deficiencia , Cortactina/inmunología , Neutrófilos/inmunología , Transducción de Señal/inmunología , Animales , Western Blotting , Adhesión Celular/fisiología , Cortactina/genética , Cortactina/metabolismo , Células Endoteliales/metabolismo , GTP Fosfohidrolasas/metabolismo , Genotipo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Fluorescente , Neutrófilos/metabolismo , Oligonucleótidos/genética , ARN Interferente Pequeño/genética , Venas Umbilicales/citología , Proteínas de Unión al GTP rap1/metabolismo , Proteínas de Unión al GTP rho
3.
J Exp Med ; 205(12): 2929-45, 2008 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19015309

RESUMEN

We have shown recently that vascular endothelial protein tyrosine phosphatase (VE-PTP), an endothelial-specific membrane protein, associates with vascular endothelial (VE)-cadherin and enhances VE-cadherin function in transfected cells (Nawroth, R., G. Poell, A. Ranft, U. Samulowitz, G. Fachinger, M. Golding, D.T. Shima, U. Deutsch, and D. Vestweber. 2002. EMBO J. 21:4885-4895). We show that VE-PTP is indeed required for endothelial cell contact integrity, because down-regulation of its expression enhanced endothelial cell permeability, augmented leukocyte transmigration, and inhibited VE-cadherin-mediated adhesion. Binding of neutrophils as well as lymphocytes to endothelial cells triggered rapid (5 min) dissociation of VE-PTP from VE-cadherin. This dissociation was only seen with tumor necrosis factor alpha-activated, but not resting, endothelial cells. Besides leukocytes, vascular endothelial growth factor also rapidly dissociated VE-PTP from VE-cadherin, indicative of a more general role of VE-PTP in the regulation of endothelial cell contacts. Dissociation of VE-PTP and VE-cadherin in endothelial cells was accompanied by tyrosine phoshorylation of VE-cadherin, beta-catenin, and plakoglobin. Surprisingly, only plakoglobin but not beta-catenin was necessary for VE-PTP to support VE-cadherin adhesion in endothelial cells. In addition, inhibiting the expression of VE-PTP preferentially increased tyrosine phosphorylation of plakoglobin but not beta-catenin. In conclusion, leukocytes interacting with endothelial cells rapidly dissociate VE-PTP from VE-cadherin, weakening endothelial cell contacts via a mechanism that requires plakoglobin but not beta-catenin.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Células Endoteliales/metabolismo , Endotelio , Leucocitos/metabolismo , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , gamma Catenina/metabolismo , Animales , Antígenos CD/genética , Cadherinas/genética , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Endosomas/metabolismo , Células Endoteliales/citología , Endotelio/citología , Endotelio/metabolismo , Humanos , Uniones Intercelulares/metabolismo , Leucocitos/citología , Linfocitos/citología , Linfocitos/metabolismo , Ratones , Neutrófilos/citología , Neutrófilos/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Factor de Necrosis Tumoral alfa/metabolismo , beta Catenina/metabolismo , gamma Catenina/genética
4.
J Biol Chem ; 282(39): 29059-29066, 2007 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-17681949

RESUMEN

The family of SLPs (Src homology 2 domain-containing leukocyte adaptor proteins) are cytoplasmic signal effectors of lymphocyte antigen receptors. A main function of SLP is to orchestrate the assembly of Ca(2+)-mobilizing enzymes at the inner leaflet of the plasma membrane. For this purpose, SLP-76 in T cells utilizes the transmembrane adaptor LAT, but the mechanism of SLP-65 membrane anchoring in B cells remains an enigma. We now employed two genetic reconstitution systems to unravel structural requirements of SLP-65 for the initiation of Ca(2+) mobilization and subsequent activation of gene transcription. First, mutational analysis of SLP-65 in DT40 B cells revealed that its C-terminal Src homology 2 domain controls efficient tyrosine phosphorylation by the kinase Syk, plasma membrane recruitment, as well as downstream signaling to NFAT activation. Second, we dissected these processes by expressing SLP-65 in SLP-76-deficient T cells and found that a kinase-independent adaptor function of Syk is required to link phosphorylated SLP-65 to Ca(2+) mobilization. These approaches unmask a mechanistic complexity of SLP-65 activation and coupling to signaling cascades in that Syk is upstream as well as downstream of SLP-65. Moreover, membrane anchoring of the SLP-65-assembled Ca(2+) initiation complex, which appears to be fundamentally different from that of closely related SLP-76, does not necessarily involve a B cell-specific component.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Linfocitos B/metabolismo , Señalización del Calcio/fisiología , Membrana Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Calcio/metabolismo , Membrana Celular/genética , Activación Enzimática/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Células Jurkat , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Tirosina Quinasas/genética , Quinasa Syk , Linfocitos T/metabolismo , Dominios Homologos src/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA