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.
Cancers (Basel) ; 13(19)2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34638342

RESUMEN

Malignant melanoma is one of the most aggressive skin cancers and is characterized by early lymph node metastasis and the capacity to develop resistance to therapies. Hence, understanding the regulation of lymphangiogenesis through mechanisms contributing to lymphatic vessel formation represents a treatment strategy for metastatic cancer. We have previously shown that CD147, a transmembrane glycoprotein overexpressed in melanoma, regulates the angiogenic process in endothelial cells. In this study, we show a correlation between high CD147 expression levels and the number of lymphatic vessels expressing LYVE-1, Podoplanin, and VEGFR-3 in human melanoma lymph nodes. CD147 upregulates in vitro lymphangiogenesis and its related mediators through the PROX-1 transcription factor. In vivo studies in a melanoma model confirmed that CD147 is involved in metastasis through a similar mechanism as in vitro. This study, demonstrating the paracrine role of CD147 in the lymphangiogenesis process, suggests that CD147 could be a promising target for the inhibition of melanoma-associated lymphangiogenesis.

2.
Oncotarget ; 6(12): 9766-80, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25825981

RESUMEN

EMMPRIN/CD147 is mainly known for its protease inducing function but a role in promoting tumor angiogenesis has also been demonstrated. This study provides evidence that EMMPRIN is a new coreceptor for the VEGFR-2 tyrosine kinase receptor in both endothelial and tumor cells, as it directly interacts with it and regulates its activation by its VEGF ligand, signalling and functional consequences both in vitro and in vivo. Computational docking analyses and mutagenesis studies identified a molecular binding site in the extracellular domain of EMMPRIN located close to the cell membrane and containing the amino acids 195/199. EMMPRIN is overexpressed in cancer and hence is able to further potentiate VEGFR-2 activation, suggesting that a combinatory therapy of an antiangiogenic drug together with an inhibitor of EMMPRIN/VEGFR-2 interaction may have a greater impact on inhibiting angiogenesis and malignancy.


Asunto(s)
Basigina/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/química , Animales , Sitios de Unión , Línea Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular , Sistema Libre de Células , Simulación por Computador , Femenino , Silenciador del Gen , Humanos , Ligandos , Ratones , Ratones Desnudos , Microvasos/citología , Mutagénesis , Mutagénesis Sitio-Dirigida , Trasplante de Neoplasias , Neovascularización Patológica , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal
3.
Exp Dermatol ; 24(6): 443-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25807898

RESUMEN

EMMPRIN is known to promote tumor invasion through extracellular matrix (ECM) degradation. Here we report that EMMPRIN can regulate melanoma cell adhesion to the ECM through an interaction with ß1 integrin involving kindlin-3. In this study, EMMPRIN knockdown in the human melanoma cell line M10 using siRNA decreased cell invasion and significantly increased cell adhesion and spreading. A morphological change from a round to a spread shape was observed associated with enhanced phalloidin-labelled actin staining. In situ proximity ligation assay and co-immunoprecipitation revealed that EMMPRIN silencing increased the interaction of ß1 integrin with kindlin-3, a focal adhesion protein. This was associated with an increase in ß1 integrin activation and a decrease in the phosphorylation of the downstream integrin kinase FAK. Moreover, the expression at both the transcript and protein level of kindlin-3 and of ß1 integrin was inversely regulated by EMMPRIN. EMMPRIN did not regulate either talin expression or its interaction with ß1 integrin. These results are consistent with our in vivo demonstration that EMMPRIN inhibition increased ß1 integrin activation and its interaction with kindlin-3. To conclude, these findings reveal a new role of EMMPRIN in tumor cell migration through ß1 integrin/kindlin-3-mediated adhesion pathway.


Asunto(s)
Basigina/fisiología , Adhesión Celular/fisiología , Integrina beta1/fisiología , Melanoma/patología , Proteínas de la Membrana/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias Cutáneas/patología , Animales , Basigina/efectos de los fármacos , Basigina/genética , Línea Celular Tumoral , Forma de la Célula/fisiología , Matriz Extracelular/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Técnicas In Vitro , Melanoma/fisiopatología , Ratones , Ratones Desnudos , ARN Interferente Pequeño/farmacología , Transducción de Señal/fisiología , Neoplasias Cutáneas/fisiopatología
4.
Oncotarget ; 5(19): 8970-85, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25344860

RESUMEN

Kindlin-3 (FERMT-3) is known to be central in hemostasis and thrombosis control and its deficiency disrupts platelet aggregation and causes Leukocyte Adhesion Deficiency disease. Here we report that Kindlin-3 has a tumor suppressive role in solid cancer. Our present genetic and functional data show that Kindlin-3 is downregulated in several solid tumors by a mechanism involving gene hypermethylation and deletions. In vivo experiments demonstrated that Kindlin-3 knockdown in 2 tumor cell models (breast cancer and melanoma) markedly increases metastasis formation, in accord with the in vitro increase of tumor cell malignant properties. The metastatic phenotype was supported by a mechanism involving alteration in ß3-integrin activation including decreased phosphorylation, interaction with talin and the internalization of its active form leading to less cell attachment and more migration/invasion. These data uncover a novel and unexpected tumor suppressor role of Kindlin-3 which can influence integrins targeted therapies development.


Asunto(s)
Neoplasias de la Mama/patología , Genes Supresores de Tumor , Melanoma/patología , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Adhesión Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Metilación de ADN , Decitabina , Femenino , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Integrina beta3/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/genética , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/metabolismo , Ratones , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Fosforilación , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Talina/genética
5.
Methods Mol Biol ; 1066: 103-11, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23955737

RESUMEN

Communication between the epithelial and stromal tissue layers, separated by basement membrane, is known to provide the information necessary for development, differentiation, and homeostasis. These interactions are altered in benign or malignant diseases, in particular when the basement membrane barrier is disrupted allowing a greater proximity between the two cell layers that triggers tissue remodeling. Epithelial-stromal interactions (ESI) have been examined in vitro by various approaches that can be broadly divided into interactions arising from secreted diffusible factors and interactions through direct cell-cell contact. Here we describe a method for the study of direct ESI through CD147, an adhesion molecule present on the epithelial cell surface and which is known to interact with stromal cells, such as fibroblasts and endothelial cells, and signal them to increase production of matrix metalloproteinases. This method can be extended to other adhesion molecules involved in ESI.


Asunto(s)
Basigina/genética , Basigina/metabolismo , Comunicación Celular/fisiología , Vesículas Citoplasmáticas/fisiología , Animales , Membrana Basal , Células CHO , Línea Celular , Cricetulus , Células Epiteliales/metabolismo , Epitelio/metabolismo , Fibroblastos/metabolismo , Células del Estroma/metabolismo , Transfección
6.
BMC Cancer ; 12: 115, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22443116

RESUMEN

BACKGROUNDS: An elevated level of EMMPRIN in cancer tissues have been correlated with tumor invasion in numerous cancers including oral cavity and larynx. Although EMMPRIN's effect has been generally attributed to its MMP inducing activity, we have previously demonstrated in breast cancer model that EMMPRIN can also enhance invasion by upregulating uPA. In this study, the role of EMMPRIN in regulating uPA and invasion was investigated in oral squamous cell carcinoma (OSCC) progression. METHODS: Precancerous and invasive oral tumoral tissues were used as well as the corresponding cell lines, DOK and SCC-9 respectively. The paracrine regulation of uPA by EMMPRIN was investigated by treating culture cells with EMMPRIN-enriched membrane vesicles. UPA expression was analyzed by qPCR and immunostaining and the consequence on the invasion capacity was studied using modified Boyden chamber assay, in the presence or absence of EMMPRIN blocking antibody, the uPA inhibitor amiloride or the MMP inhibitor marimastat. RESULTS: OSCC tumors were shown to express more EMMPRIN and uPA compared to dysplastic lesions. The corresponding cell models, SCC-9 and DOK cells, displayed similar expression pattern. In both cell types EMMPRIN upregulated the expression of uPA as well as that of MMP-2 and MMP-9. EMMPRIN treatment led to a significant increase in cell invasion both in the invasive SCC-9 and in the less invasive dysplastic DOK cells, in an MMP and uPA dependent manner. CONCLUSIONS: Our results suggest that the upregulation of uPA contributes to EMMPRIN's effect in promoting oral tumor invasion.


Asunto(s)
Basigina/fisiología , Carcinoma de Células Escamosas/metabolismo , Neoplasias de la Boca/metabolismo , Proteínas de Neoplasias/fisiología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Western Blotting , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Progresión de la Enfermedad , Humanos , Inmunohistoquímica , Neoplasias de la Boca/patología , Invasividad Neoplásica/patología , Lesiones Precancerosas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba
7.
PLoS One ; 5(8): e12265, 2010 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-20824203

RESUMEN

EMMPRIN's expression in melanoma tissue was reported to be predictive of poor prognosis. Here we demonstrate that EMMPRIN up-regulated VEGF receptor-2 (VEGFR-2) in two different primary melanoma cell lines and consequently increased migration and proliferation of these cells while inhibiting their apoptosis. SiRNA inhibition of VEGFR-2 expression abrogated these EMMPRIN effects. EMMPRIN regulation of VEGFR-2 was mediated through the over-expression of HIF-2alpha and its translocation to the nucleus where it forms heterodimers with HIF-1beta. These results were supported by an in vivo correlation between the expression of EMMPRIN with that of VEGFR-2 in human melanoma tissues as well as with the extent of HIF-2alpha localization in the nucleus. They demonstrate a novel mechanism by which EMMPRIN promotes tumor progression through HIF-2alpha/VEGFR-2 mediated mechanism, with an autocrine role in melanoma cell malignancy. The inhibition of EMMPRIN in cancer may thus simultaneously target both the VEGFR-2/VEGF system and the matrix degrading proteases to block tumor cell growth and invasion.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Basigina/metabolismo , Melanoma/patología , Regulación hacia Arriba , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Transporte Activo de Núcleo Celular , Animales , Apoptosis/genética , Comunicación Autocrina/genética , Basigina/genética , Células CHO , Línea Celular Tumoral , Movimiento Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Cricetinae , Cricetulus , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Invasividad Neoplásica/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA