Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
J Cell Biol ; 178(3): 425-36, 2007 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-17664334

RESUMEN

Hypoxia activates genetic programs that facilitate cell survival; however, in cancer, it may promote invasion and metastasis. In this study, we show that breast cancer cells cultured in 1.0% O(2) demonstrate changes consistent with epithelial-mesenchymal transition (EMT). Snail translocates to the nucleus, and E-cadherin is lost from plasma membranes. Vimentin expression, cell migration, Matrigel invasion, and collagen remodeling are increased. Hypoxia-induced EMT is accompanied by increased expression of the urokinase-type plasminogen activator receptor (uPAR) and activation of cell signaling factors downstream of uPAR, including Akt and Rac1. Glycogen synthase kinase-3beta is phosphorylated, and Snail expression is increased. Hypoxia-induced EMT is blocked by uPAR gene silencing and mimicked by uPAR overexpression in normoxia. Antagonizing Rac1 or phosphatidylinositol 3-kinase also inhibits development of cellular properties associated with EMT in hypoxia. Breast cancer cells implanted on chick chorioallantoic membranes and treated with CoCl(2), to model hypoxia, demonstrate increased dissemination. We conclude that in hypoxia, uPAR activates diverse cell signaling pathways that cooperatively induce EMT and may promote cancer metastasis.


Asunto(s)
Neoplasias de la Mama , Epitelio/fisiología , Hipoxia , Mesodermo/fisiología , Receptores de Superficie Celular/metabolismo , Transducción de Señal/fisiología , Animales , Antimutagênicos/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular/fisiología , Cobalto/metabolismo , Colágeno/metabolismo , Activación Enzimática , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Mesodermo/citología , Invasividad Neoplásica , Metástasis de la Neoplasia , Oxígeno/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Superficie Celular/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Proteína de Unión al GTP rac1/metabolismo
2.
J Biol Chem ; 284(34): 22825-33, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19546228

RESUMEN

Hypoxia induces expression of the urokinase receptor (uPAR) and activates uPAR-dependent cell signaling in cancer cells. This process promotes epithelial-mesenchymal transition (EMT). uPAR overexpression in cancer cells also promotes EMT. In this study, we tested whether uPAR may be targeted to reverse cancer cell EMT. When MDA-MB 468 breast cancer cells were cultured in 1% O(2), uPAR expression increased, as anticipated. Cell-cell junctions were disrupted, vimentin expression increased, and E-cadherin was lost from cell surfaces, indicating EMT. Transferring these cells back to 21% O(2) decreased uPAR expression and reversed the signs of EMT. In uPAR-overexpressing MDA-MB 468 cells, EMT was reversed by silencing expression of endogenously produced urokinase-type plasminogen activator (uPA), which is necessary for uPAR-dependent cell signaling, or by targeting uPAR-activated cell signaling factors, including phosphatidylinositol 3-kinase, Src family kinases, and extracellular signal-regulated kinase. MDA-MB 231 breast cancer cells express high levels of uPA and uPAR and demonstrate mesenchymal cell morphology under normoxic culture conditions (21% O(2)). Silencing uPA expression in MDA-MB-231 cells decreased expression of vimentin and Snail, and induced changes in morphology characteristic of epithelial cells. These results demonstrate that uPAR-initiated cell signaling may be targeted to reverse EMT in cancer.


Asunto(s)
Epitelio/patología , Mesodermo/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Transducción de Señal , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Movimiento Celular , Embrión de Pollo , Pollos , Cromonas/farmacología , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Flavonoides/farmacología , Humanos , Immunoblotting , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Microscopía Fluorescente , Morfolinas/farmacología , Oxígeno/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tubulina (Proteína)/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Vimentina/genética
3.
Cancer Res ; 67(20): 9817-24, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17942912

RESUMEN

Low-density lipoprotein receptor-related protein-1 (LRP-1) is a multifunctional receptor involved in receptor-mediated endocytosis and cell signaling. In this study, we show that LRP-1 is abundantly expressed in severe combined immunodeficient (SCID) mouse xenografts by various human cancer cell lines that express very low or undetectable levels of LRP-1 when cultured in 21% O2 in vitro (standard cell culture conditions). To test whether LRP-1 expression in vivo may be explained by hypoxia in the xenografts, CL16 cells, which are derived from the MDA-MB-435 cell line, were cultured in 1.0% O2. A substantial increase in LRP-1 expression was observed. To test the activity of LRP-1 in cancer progression in vivo, LRP-1 expression was silenced in CL16 cells with short hairpin RNA. These cells formed tumors in SCID mice, in which LRP-1 expression remained silenced. Although LRP-1 gene silencing did not inhibit CL16 cell dissemination from the primary tumors to the lungs, the pulmonary metastases failed to enlarge, suggesting compromised survival or growth at the implantation site. In cell culture experiments, significantly increased cell death was observed when LRP-1-silenced CL16 cells were exposed to CoCl2, which models changes that occur in hypoxia. Furthermore, LRP-1-silenced cells expressed decreased levels of vascular endothelial growth factor in response to 1.0% O2. These results suggest mechanisms by which LRP-1 may facilitate the development and growth of cancer metastases in vivo.


Asunto(s)
Neoplasias de la Mama/patología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/biosíntesis , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Cobalto/farmacología , Silenciador del Gen , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/deficiencia , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones SCID , Trasplante de Neoplasias , Oxígeno/administración & dosificación , Oxígeno/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/biosíntesis
4.
PLoS One ; 4(8): e6713, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19696929

RESUMEN

Tumor-associated macrophages are known to influence cancer progression by modulation of immune function, angiogenesis, and cell metastasis, however, little is known about the chemokine signaling networks that regulate this process. Utilizing CT26 colon cancer cells and RAW 264.7 macrophages as a model cellular system, we demonstrate that treatment of CT26 cells with RAW 264.7 conditioned medium induces cell migration, invasion and metastasis. Inflammatory gene microarray analysis indicated CT26-stimulated RAW 264.7 macrophages upregulate SDF-1alpha and VEGF, and that these cytokines contribute to CT26 migration in vitro. RAW 264.7 macrophages also showed a robust chemotactic response towards CT26-derived chemokines. In particular, microarray analysis and functional testing revealed CSF-1 as the major chemoattractant for RAW 264.7 macrophages. Interestingly, in the chick CAM model of cancer progression, RAW 264.7 macrophages localized specifically to the tumor periphery where they were found to increase CT26 tumor growth, microvascular density, vascular disruption, and lung metastasis, suggesting these cells home to actively invading areas of the tumor, but not the hypoxic core of the tumor mass. In support of these findings, hypoxic conditions down regulated CSF-1 production in several tumor cell lines and decreased RAW 264.7 macrophage migration in vitro. Together our findings suggest a model where normoxic tumor cells release CSF-1 to recruit macrophages to the tumor periphery where they secrete motility and angiogenic factors that facilitate tumor cell invasion and metastasis.


Asunto(s)
Movimiento Celular , Factores Quimiotácticos/metabolismo , Macrófagos/metabolismo , Metástasis de la Neoplasia , Neoplasias Experimentales/metabolismo , Neovascularización Patológica , Transducción de Señal , Animales , Línea Celular , Embrión de Pollo , Medios de Cultivo Condicionados , Perfilación de la Expresión Génica , Macrófagos/citología , Ratones , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa
5.
Proc Natl Acad Sci U S A ; 104(44): 17406-11, 2007 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-17954920

RESUMEN

Cell metastasis is a highly dynamic process that occurs in multiple steps. Understanding this process has been limited by the inability to visualize tumor cell behavior in real time by using animal models. Here, we employ translucent zebrafish and high-resolution confocal microscopy to study how human cancer cells invade in tissues, induce angiogenesis, and interact with newly formed vessels. We use this system to study how the human metastatic gene RhoC promotes the initial steps of metastasis. We find that RhoC expression induces a primitive amoeboid-like cell invasion characterized by the formation of dynamic membrane protrusions and blebs. Surprisingly, these structures penetrate the blood vessel wall exclusively at sites of vascular remodeling and not at regions of existing intact vessels. This process requires tumor cells to secrete VEGF, which induces vascular openings, which in turn, serve as portholes allowing access of RhoC-expressing cells to the blood system. Our results support a model in which the early steps in intravasation and metastasis require two independent events: (i) dynamic regulation of the actin/myosin cytoskeleton within the tumor cell to form protrusive structures and (ii) vascular permeablization and vessel remodeling. The integration of zebrafish transgenic technology with human cancer biology may aid in the development of cancer models that target specific organs, tissues, or cell types within the tumors. Zebrafish could also provide a cost-effective means for the rapid development of therapeutic agents directed at blocking human cancer progression and tumor-induced angiogenesis.


Asunto(s)
Imagenología Tridimensional/métodos , Neoplasias/irrigación sanguínea , Neoplasias/patología , Pez Cebra , Animales , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Humanos , Invasividad Neoplásica , Neoplasias/metabolismo , Fenotipo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína rhoC de Unión a GTP
6.
J Biol Chem ; 280(47): 39273-7, 2005 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-16207704

RESUMEN

Recent studies indicate that cancer cells express erythropoietin receptor (EpoR). In this study, we have shown that erythropoietin (Epo) activates the mitogen-activated protein kinase, extracellular signal-regulated kinase (ERK), and promotes migration in MCF-7 breast cancer cells. Epo-stimulated MCF-7 cell migration was blocked by the MEK inhibitor PD098059 and by dominant negative MEK-1, indicating an essential role for ERK. When MCF-7 cells were exposed to hypoxia (1.0% O(2)) for 3 h, the Epo mRNA level increased 2.4 +/- 0.5-fold, the basal level of ERK activation increased, and cell migration increased 2.0 +/- 0.1-fold. Soluble EpoR and Epo-neutralizing antibody significantly inhibited hypoxia-induced MCF-7 cell migration, suggesting a major role for autocrine EpoR cell signaling. MCF-7 cell migration under hypoxic conditions was also inhibited by PD098059. These experiments identify a novel pathway by which exogenously administered Epo, and Epo that is produced locally by cancer cells under hypoxic conditions, may stimulate cancer cell migration.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Eritropoyetina/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias de la Mama/patología , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Eritropoyetina/antagonistas & inhibidores , Eritropoyetina/genética , Eritropoyetina/metabolismo , Femenino , Flavonoides/farmacología , Humanos , Pruebas de Neutralización , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Receptores de Eritropoyetina/metabolismo , Proteínas Recombinantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA