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.
Arterioscler Thromb Vasc Biol ; 37(9): 1710-1721, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28729362

RESUMEN

OBJECTIVE: Molecular pathways governing blood vessel patterning are vital to vertebrate development. Because of their ability to counteract proangiogenic factors, antiangiogenic secreted Sema3 (class 3 semaphorins) control embryonic vascular morphogenesis. However, if and how Sema3 may play a role in the control of extraembryonic vascular development is presently unknown. APPROACH AND RESULTS: By characterizing genetically modified mice, here, we show that surprisingly Sema3F acts instead as a selective extraembryonic, but not intraembryonic proangiogenic cue. Both in vivo and in vitro, in visceral yolk sac epithelial cells, Sema3F signals to inhibit the phosphorylation-dependent degradation of Myc, a transcription factor that drives the expression of proangiogenic genes, such as the microRNA cluster 17/92. In Sema3f-null yolk sacs, the transcription of Myc-regulated microRNA 17/92 cluster members is impaired, and the synthesis of Myc and microRNA 17/92 foremost antiangiogenic target Thbs1 (thrombospondin 1) is increased, whereas Vegf (vascular endothelial growth factor) signaling is inhibited in yolk sac endothelial cells. Consistently, exogenous recombinant Sema3F inhibits the phosphorylation-dependent degradation of Myc and the synthesis of Thbs1 in mouse F9 teratocarcinoma stem cells that were in vitro differentiated in visceral yolk sac epithelial cells. Sema3f-/- mice placentas are also highly anemic and abnormally vascularized. CONCLUSIONS: Sema3F functions as an unconventional Sema3 that promotes extraembryonic angiogenesis by inhibiting the Myc-regulated synthesis of Thbs1 in visceral yolk sac epithelial cells.


Asunto(s)
Células Epiteliales/metabolismo , Proteínas de la Membrana/metabolismo , Neovascularización Fisiológica , Proteínas del Tejido Nervioso/metabolismo , Placenta/irrigación sanguínea , Saco Vitelino/irrigación sanguínea , Animales , Línea Celular Tumoral , Células Madre de Carcinoma Embrionario/metabolismo , Células Endoteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Genotipo , Edad Gestacional , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Fenotipo , Fosforilación , Embarazo , Proteolisis , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Trombospondina 1/genética , Trombospondina 1/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Cell Adh Migr ; 10(6): 641-651, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27439112

RESUMEN

Secreted class 3 semaphorins (Sema3), which signal through holoreceptor complexes that are formed by different subunits, such as neuropilins (Nrps), proteoglycans, and plexins, were initially characterized as fundamental regulators of axon guidance during embryogenesis. Subsequently, Sema3A, Sema3C, Sema3D, and Sema3E were discovered to play crucial roles in cardiovascular development, mainly acting through Nrp1 and Plexin D1, which funnels the signal of multiple Sema3 in vascular endothelial cells. Mechanistically, Sema3 proteins control cardiovascular patterning through the enzymatic GTPase-activating-protein activity of the cytodomain of Plexin D1, which negatively regulates the function of Rap1, a small GTPase that is well-known for its ability to drive vascular morphogenesis and to elicit the conformational activation of integrin adhesion receptors.


Asunto(s)
Sistema Cardiovascular/embriología , Sistema Cardiovascular/metabolismo , Semaforinas/metabolismo , Animales , Humanos , Modelos Biológicos , Mutación/genética , Fenotipo , Transducción de Señal
3.
J Clin Invest ; 122(5): 1832-48, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22484816

RESUMEN

Cancer development, progression, and metastasis are highly dependent on angiogenesis. The use of antiangiogenic drugs has been proposed as a novel strategy to interfere with tumor growth, but cancer cells respond by developing strategies to escape these treatments. In particular, animal models show that antiangiogenic drugs currently used in clinical settings reduce tumor tissue oxygenation and trigger molecular events that foster cancer resistance to therapy. Here, we show that semaphorin 3A (Sema3A) expression overcomes the proinvasive and prometastatic resistance observed upon angiogenesis reduction by the small-molecule tyrosine inhibitor sunitinib in both pancreatic neuroendocrine tumors (PNETs) in RIP-Tag2 mice and cervical carcinomas in HPV16/E2 mice. By improving cancer tissue oxygenation and extending the normalization window, Sema3A counteracted sunitinib-induced activation of HIF-1α, Met tyrosine kinase receptor, epithelial-mesenchymal transition (EMT), and other hypoxia-dependent signaling pathways. Sema3A also reduced tumor hypoxia and halted cancer dissemination induced by DC101, a specific inhibitor of the VEGF pathway. As a result, reexpressing Sema3A in cancer cells converts metastatic PNETs and cervical carcinomas into benign lesions. We therefore suggest that this strategy could be developed to safely harnesses the therapeutic potential of the antiangiogenic treatment.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Indoles/uso terapéutico , Neoplasias Hepáticas/secundario , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Pirroles/uso terapéutico , Semaforina-3A/genética , Inhibidores de la Angiogénesis/farmacología , Animales , Hipoxia de la Célula/efectos de los fármacos , Terapia Combinada , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Femenino , Terapia Genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Indoles/farmacología , Metástasis Linfática , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Invasividad Neoplásica , Neovascularización Fisiológica , Tumores Neuroendocrinos/irrigación sanguínea , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/terapia , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Pericitos/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Pirroles/farmacología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Semaforina-3A/biosíntesis , Sunitinib , Carga Tumoral , Neoplasias del Cuello Uterino/irrigación sanguínea , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/terapia , Neoplasias del Cuello Uterino/virología
4.
J Immunol ; 188(8): 4081-92, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22442441

RESUMEN

The axon guidance cues semaphorins (Semas) and their receptors plexins have been shown to regulate both physiological and pathological angiogenesis. Sema4A plays an important role in the immune system by inducing T cell activation, but to date, the role of Sema4A in regulating the function of macrophages during the angiogenic and inflammatory processes remains unclear. In this study, we show that macrophage activation by TLR ligands LPS and polyinosinic-polycytidylic acid induced a time-dependent increase of Sema4A and its receptors PlexinB2 and PlexinD1. Moreover, in a thioglycollate-induced peritonitis mouse model, Sema4A was detected in circulating Ly6C(high) inflammatory monocytes and peritoneal macrophages. Acting via PlexinD1, exogenous Sema4A strongly increased macrophage migration. Of note, Sema4A-activated PlexinD1 enhanced the expression of vascular endothelial growth factor-A, but not of inflammatory chemokines. Sema4A-stimulated macrophages were able to activate vascular endothelial growth factor receptor-2 and the PI3K/serine/threonine kinase Akt pathway in endothelial cells and to sustain their migration and in vivo angiogenesis. Remarkably, in an in vivo cardiac ischemia/reperfusion mouse model, Sema4A was highly expressed in macrophages recruited at the injured area. We conclude that Sema4A activates a specialized and restricted genetic program in macrophages able to sustain angiogenesis and participates in their recruitment and activation in inflammatory injuries.


Asunto(s)
Macrófagos Peritoneales/inmunología , Neovascularización Fisiológica , Semaforinas/fisiología , Factor A de Crecimiento Endotelial Vascular/inmunología , Animales , Movimiento Celular , Quimiocinas/biosíntesis , Quimiocinas/inmunología , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lipopolisacáridos/farmacología , Activación de Macrófagos , Macrófagos Peritoneales/patología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Miocardio/patología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Peritonitis/inmunología , Peritonitis/metabolismo , Peritonitis/patología , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Semaforinas/farmacología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA