Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Circ Res ; 113(11): 1206-18, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24025447

RESUMEN

RATIONALE: The formation of novel blood vessels is initiated by vascular endothelial growth factor. Subsequently, DLL4-Notch signaling controls the selection of tip cells, which guide new sprouts, and trailing stalk cells. Notch signaling in stalk cells is induced by DLL4 on the tip cells. Moreover, DLL4 and DLL1 are expressed in the stalk cell plexus to maintain Notch signaling. Notch loss-of-function causes formation of a hyperdense vascular network with disturbed blood flow. OBJECTIVE: This study was aimed at identifying novel modifiers of Notch signaling that interact with the intracellular domains of DLL1 and DLL4. METHODS AND RESULTS: Synaptojanin-2 binding protein (SYNJ2BP, also known as ARIP2) interacted with the PDZ binding motif of DLL1 and DLL4, but not with the Notch ligand Jagged-1. SYNJ2BP was preferentially expressed in stalk cells, enhanced DLL1 and DLL4 protein stability, and promoted Notch signaling in endothelial cells. SYNJ2BP induced expression of the Notch target genes HEY1, lunatic fringe (LFNG), and ephrin-B2, reduced phosphorylation of ERK1/2, and decreased expression of the angiogenic factor vascular endothelial growth factor (VEGF)-C. It inhibited the expression of genes enriched in tip cells, such as angiopoietin-2, ESM1, and Apelin, and impaired tip cell formation. SYNJ2BP inhibited endothelial cell migration, proliferation, and VEGF-induced angiogenesis. This could be rescued by blockade of Notch signaling or application of angiopoietin-2. SYNJ2BP-silenced human endothelial cells formed a functional vascular network in immunocompromised mice with significantly increased vascular density. CONCLUSIONS: These data identify SYNJ2BP as a novel inhibitor of tip cell formation, executing its functions predominately by promoting Delta-Notch signaling.


Asunto(s)
Proteínas Portadoras/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas de la Membrana/fisiología , Neovascularización Fisiológica/fisiología , Receptores Notch/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Movimiento Celular/fisiología , Proliferación Celular , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Femenino , Humanos , Ratones , Ratones SCID , Modelos Animales , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
2.
Cancer Res ; 73(16): 5080-9, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23774209

RESUMEN

Extracellular RNA (eRNA) released from injured cells promotes tissue permeability, thrombosis, and inflammation in vitro and in vivo, and RNase1 pretreatment can reduce all these effects. In this study, we investigated the role of the eRNA/RNase1 system in tumor progression and metastasis. Under quiescent and stimulatory conditions, tumor cells released much higher levels of endogenous extracellular RNA (eRNA) than nontumor cells. In glioblastomas, eRNA was detected at higher levels in tumors than nontumor tissue. eRNA induced tumor cells to adhere to and migrate through human cerebral microvascular endothelial cells (HCMEC/D3), in a manner requiring activation of VEGF signaling. In addition, eRNA liberated TNF-α from macrophages in a manner requiring activation of the TNF-α-converting enzyme TACE. Accordingly, supernatants derived from eRNA-treated macrophages enhanced tumor cell adhesion to HCMEC/D3. TNF-α release evoked by eRNA relied upon signaling activation of mitogen-activated protein kinases and the NF-κB pathway. In subcutaneous xenograft models of human cancer, administration of RNase1 but not DNase decreased tumor volume and weight. Taken together, these results suggest that eRNA released from tumor cells has the capacity to promote tumor cell invasion through endothelial barriers by both direct and indirect mechanisms, including through a mechanism involving TNF-α release from tumor-infiltrating monocytes/macrophages. Our findings establish a crucial role for eRNA in driving tumor progression, and they suggest applications for extracellular RNase1 as an antiinvasive regimen for cancer treatment.


Asunto(s)
Movimiento Celular/genética , Neoplasias/genética , Neoplasias/patología , ARN/genética , ARN/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animales , Adhesión Celular/genética , Línea Celular Tumoral , Células Cultivadas , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Células HT29 , Xenoinjertos , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Microvasos/metabolismo , Microvasos/patología , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Monocitos/metabolismo , Monocitos/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Ribonucleasas/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 33(3): 533-43, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23288173

RESUMEN

OBJECTIVE: Histone deacetylases (HDACs) modulate gene expression by deacetylation of histone and nonhistone proteins. Several HDACs control angiogenesis, but the role of HDAC9 is unclear. METHODS AND RESULTS: Here, we analyzed the function of HDAC9 in angiogenesis and its involvement in regulating microRNAs. In vitro, silencing of HDAC9 reduces endothelial cell tube formation and sprouting. Furthermore, HDAC9 silencing decreases vessel formation in a spheroid-based Matrigel plug assay in mice and disturbs vascular patterning in zebrafish embryos. Genetic deletion of HDAC9 reduces retinal vessel outgrowth and impairs blood flow recovery after hindlimb ischemia. Consistently, overexpression of HDAC9 increases endothelial cell sprouting, whereas mutant constructs lacking the catalytic domain, the nuclear localization sequence, or sumoylation site show no effect. To determine the mechanism underlying the proangiogenic effect of HDAC9, we measured the expression of the microRNA (miR)-17-92 cluster, which is known for its antiangiogenic activity. We demonstrate that silencing of HDAC9 in endothelial cells increases the expression of miR-17-92. Inhibition of miR-17-20a rescues the sprouting defects induced by HDAC9 silencing in vitro and blocking miR-17 expression partially reverses the disturbed vascular patterning of HDAC9 knockdown in zebrafish embryos. CONCLUSIONS: We found that HDAC9 promotes angiogenesis and transcriptionally represses the miR-17-92 cluster.


Asunto(s)
Histona Desacetilasas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/enzimología , Isquemia/enzimología , MicroARNs/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Proteínas Represoras/metabolismo , Neovascularización Retiniana/enzimología , Proteínas de Pez Cebra/metabolismo , Animales , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Células HEK293 , Miembro Posterior , Histona Desacetilasas/deficiencia , Histona Desacetilasas/genética , Humanos , Isquemia/genética , Isquemia/fisiopatología , Ratones , Ratones Noqueados , MicroARNs/genética , Mutación , Neovascularización Fisiológica/genética , Interferencia de ARN , ARN Largo no Codificante , Flujo Sanguíneo Regional , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Neovascularización Retiniana/genética , Neovascularización Retiniana/fisiopatología , Transfección , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
4.
EMBO J ; 30(20): 4142-56, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21847094

RESUMEN

Histone deacetylases (HDACs) deacetylate histones and non-histone proteins, thereby affecting protein activity and gene expression. The regulation and function of the cytoplasmic class IIb HDAC6 in endothelial cells (ECs) is largely unexplored. Here, we demonstrate that HDAC6 is upregulated by hypoxia and is essential for angiogenesis. Silencing of HDAC6 in ECs decreases sprouting and migration in vitro and formation of functional vascular networks in matrigel plugs in vivo. HDAC6 regulates zebrafish vessel formation, and HDAC6-deficient mice showed a reduced formation of perfused vessels in matrigel plugs. Consistently, overexpression of wild-type HDAC6 increases sprouting from spheroids. HDAC6 function requires the catalytic activity but is independent of ubiquitin binding and deacetylation of α-tubulin. Instead, we found that HDAC6 interacts with and deacetylates the actin-remodelling protein cortactin in ECs, which is essential for zebrafish vessel formation and which mediates the angiogenic effect of HDAC6. In summary, we show that HDAC6 is necessary for angiogenesis in vivo and in vitro, involving the interaction and deacetylation of cortactin that regulates EC migration and sprouting.


Asunto(s)
Movimiento Celular , Cortactina/metabolismo , Histona Desacetilasas/metabolismo , Neovascularización Fisiológica , Tubulina (Proteína)/metabolismo , Proteínas de Pez Cebra/metabolismo , Acetilación , Animales , Carcinoma Pulmonar de Lewis/enzimología , Células Cultivadas , Células Endoteliales/metabolismo , Femenino , Histona Desacetilasa 6 , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Noqueados
5.
Blood ; 117(9): 2735-44, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21224470

RESUMEN

The HLX gene encoding a diverged homeobox transcription factor has been found to be up-regulated by vascular endothelial growth factor-A (VEGF-A) in endothelial cells. We have now investigated the gene repertoire induced by HLX and its potential biologic function. HLX strongly increased the transcripts for several repulsive cell-guidance proteins including UNC5B, plexin-A1, and semaphorin-3G. In addition, genes for transcriptional repressors such as HES-1 were up-regulated. In line with these findings, adenoviral overexpression of HLX inhibited endothelial cell migration, sprouting, and vessel formation in vitro and in vivo, whereas proliferation was unaffected. This inhibition of sprouting was caused to a significant part by HLX-mediated up-regulation of UNC5B as shown by short hairpin RNA (shRNA)-mediated down-modulation of the respective mRNA. VEGF-A stimulation of endothelial cells induced elevated levels of HLX over longer time periods resulting in especially high up-regulation of UNC5B mRNA as well as an increase in cells displaying UNC5B at their surface. However, induction of HLX was strongly reduced and UNC5B up-regulation completely abrogated when cells were exposed to hypoxic conditions. These data suggest that HLX may function to balance attractive with repulsive vessel guidance by up-regulating UNC5B and to down-modulate sprouting under normoxic conditions.


Asunto(s)
Movimiento Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Proteínas de Homeodominio/metabolismo , Neovascularización Fisiológica , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Hipoxia de la Célula/genética , Movimiento Celular/genética , Proliferación Celular , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Ratones , Ratones SCID , Receptores de Netrina , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/genética , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Factores de Transcripción/genética , Transcripción Genética , Trasplante Heterólogo , Regulación hacia Arriba/genética
6.
Cancer Res ; 66(14): 7083-94, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16849554

RESUMEN

Expression of the tetraspanin CO-029 is associated with poor prognosis in patients with gastrointestinal cancer. In a pancreatic tumor line, overexpression of the rat homologue, D6.1A, induces lethally disseminated intravascular coagulation, suggesting D6.1A engagement in angiogenesis. D6.1A-overexpressing tumor cells induce the greatest amount of angiogenesis in vivo, and tumor cells as well as exosomes derived thereof strikingly increase endothelial cell branching in vitro. Tumor cell-derived D6.1A stimulates angiogenic factor transcription, which includes increased matrix metalloproteinase and urokinase-type plasminogen activator secretion, pronounced vascular endothelial growth factor expression in fibroblasts, vascular endothelial growth factor receptor expression, and strong D6.1A up-regulation in sprouting endothelium. Thus, D6.1A initiates an angiogenic loop that, probably due to the abundance of D6.1A in tumor-derived exosomes, reaches organs distant from the tumor. Most importantly, because of the strong D6.1A up-regulation on sprouting capillaries, angiogenesis could be completely inhibited by a D6.1A-specific antibody, irrespective of whether or not the tumor expresses D6.1A. Tetraspanins have been suggested to be involved in morphogenesis. This is the first report that a tetraspanin, CO-029/D6.1A, promotes tumor growth by its capacity to induce systemic angiogenesis that can effectively, and with high selectivity for sprouting endothelium, be blocked by a D6.1A-specific antibody.


Asunto(s)
Adenocarcinoma/irrigación sanguínea , Glicoproteínas de Membrana/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias Pancreáticas/irrigación sanguínea , Adenocarcinoma/metabolismo , Animales , Colagenasas/biosíntesis , Colagenasas/genética , Endotelio Vascular/metabolismo , Metaloproteinasa 13 de la Matriz , Glicoproteínas de Membrana/biosíntesis , Proteínas de Neoplasias/biosíntesis , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neoplasias Pancreáticas/metabolismo , Ratas , Ratas Wistar , Tetraspaninas , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
7.
Clin Cancer Res ; 11(8): 2840-52, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15837731

RESUMEN

PURPOSE: Patients with pancreatic adenocarcinoma have a poor prognosis due to the extraordinary high invasive capacity of this tumor. Altered integrin and tetraspanin expression is suggested to be an important factor. We recently reported that after protein kinase C activation, colocalization of alpha6beta4 with the tetraspanin CO-029 strongly supports migration of a rat pancreatic adenocarcinoma. The finding led us to explore whether and which integrin-tetraspanin complexes influence the motility of human pancreatic tumors. EXPERIMENTAL DESIGN: Integrin and tetraspanin expression of pancreatic and colorectal adenocarcinoma was evaluated with emphasis on colocalization and the impact of integrin-tetraspanin associations on tumor cell motility. RESULTS: The majority of pancreatic and colorectal tumors expressed the alpha2, alpha3, alpha6, beta1, and beta4 integrins and the tetraspanins CD9, CD63, CD81, CD151, and CO-029. Expression of alpha6beta4 and CO-029 was restricted to tumor cells, whereas alpha1, alpha2, alpha3, alpha6, beta1, and CD9, CD81, CD151 were also expressed by the surrounding stroma. CD63, CD81, and beta1 expression was observed at comparably high levels in healthy pancreatic tissue. alpha3beta1 frequently colocalized and coimmunoprecipitated with CD9, CD81, and CD151, whereas alpha6beta4 colocalized and coimmunoprecipitated mostly with CD151 and CO-029. Notably, protein kinase C activation strengthened only the colocalization of CD151 and CO-029 with beta4 and was accompanied by internalization of the integrin-tetraspanin complex, decreased laminin 5 adhesion, and increased cell migration. CONCLUSION: alpha6beta4 is selectively up-regulated in pancreatic and colorectal cancer. The association of alpha6beta4 with CD151 and CO-029 correlates with increased tumor cell motility.


Asunto(s)
Adenocarcinoma/patología , Proteínas de la Membrana/análisis , Neoplasias Pancreáticas/patología , Adenocarcinoma/metabolismo , Antígenos CD/análisis , Antígenos de Neoplasias/análisis , Línea Celular Tumoral , Movimiento Celular , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunoprecipitación , Integrinas/análisis , Glicoproteínas de Membrana/análisis , Neoplasias Pancreáticas/metabolismo , Tetraspanina 24 , Tetraspaninas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...