Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Prostate ; 76(1): 97-113, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26417683

RESUMEN

BACKGROUND: Previous results from our lab indicate a tumor suppressor role for the transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) in prostate cancer (PCa). Here, we further characterize this role and uncover new functions for TMEFF2 in cancer and adult prostate regeneration. METHODS: The role of TMEFF2 was examined in PCa cells using Matrigel(TM) cultures and allograft models of PCa cells. In addition, we developed a transgenic mouse model that expresses TMEFF2 from a prostate specific promoter. Anatomical, histological, and metabolic characterizations of the transgenic mouse prostate were conducted. The effect of TMEFF2 in prostate regeneration was studied by analyzing branching morphogenesis in the TMEFF2-expressing mouse lobes and alterations in branching morphogenesis were correlated with the metabolomic profiles of the mouse lobes. The role of TMEFF2 in prostate tumorigenesis in whole animals was investigated by crossing the TMEFF2 transgenic mice with the TRAMP mouse model of PCa and analyzing the histopathological changes in the progeny. RESULTS: Ectopic expression of TMEFF2 impairs growth of PCa cells in Matrigel or allograft models. Surprisingly, while TMEFF2 expression in the TRAMP mouse did not have a significant effect on the glandular prostate epithelial lesions, the double TRAMP/TMEFF2 transgenic mice displayed an increased incidence of neuroendocrine type tumors. In addition, TMEFF2 promoted increased branching specifically in the dorsal lobe of the prostate suggesting a potential role in developmental processes. These results correlated with data indicating an alteration in the metabolic profile of the dorsal lobe of the transgenic TMEFF2 mice. CONCLUSIONS: Collectively, our results confirm the tumor suppressor role of TMEFF2 and suggest that ectopic expression of TMEFF2 in mouse prostate leads to additional lobe-specific effects in prostate regeneration and tumorigenesis. This points to a complex and multifunctional role for TMEFF2 during PCa progression.


Asunto(s)
Adenocarcinoma , Carcinogénesis/metabolismo , Proteínas de la Membrana/metabolismo , Tumores Neuroendocrinos , Próstata , Neoplasias de la Próstata , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Trasplante de Neoplasias/patología , Trasplante de Neoplasias/fisiología , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Próstata/patología , Próstata/fisiología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Regeneración , Células Tumorales Cultivadas
2.
Biochim Biophys Acta ; 1843(6): 1216-24, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24632071

RESUMEN

Cell adhesion and migration play important roles in physiological and pathological states, including embryonic development and cancer invasion and metastasis. The type I transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) is expressed mainly in brain and prostate and its expression is deregulated in prostate cancer. We have previously shown that TMEFF2 can function as a tumor suppressor by inhibiting cell migration and invasion of prostate cells. However, the molecular mechanisms involved in this inhibition are not clear. In this study we demonstrate that TMEFF2 affects cell adhesion and migration of prostate cancer cells and that this effect correlates with changes in integrin expression and RhoA activation. Deletion of a 13 basic-rich amino acid region in the cytoplasmic domain of TMEFF2 prevented these effects. Overexpression of TMEFF2 reduced cell attachment and migration on vitronectin and caused a concomitant decrease in RhoA activation, stress fiber formation and expression of αv, ß1 and ß3 integrin subunits. Conversely, TMEFF2 interference in 22Rv1 prostate cancer cells resulted in an increased integrin expression. Results obtained with a double TRAMP/TMEFF2 transgenic mouse also indicated that TMEFF2 expression reduced integrin expression in the mouse prostate. In summary, the data presented here indicate an important role of TMEFF2 in regulating cell adhesion and migration that involves integrin signaling and is mediated by its cytoplasmic domain.


Asunto(s)
Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Integrina alfaV/metabolismo , Integrina beta3/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteína de Unión al GTP rhoA/metabolismo , Animales , Apoptosis , Western Blotting , Proliferación Celular , Forma de la Célula , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Adhesiones Focales , Humanos , Integrina alfaV/genética , Integrina beta3/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias de la Próstata/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína de Unión al GTP rhoA/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...