Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 461(1): 47-53, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25866182

RESUMEN

Wound healing is critical for normal development and pathological processes including cancer cell metastasis. MAPK, Rho-GTPases and NFκB are important regulators of wound healing, but mechanisms for their integration are incompletely understood. Annexin-A1 (ANXA1) is upregulated in invasive breast cancer cells resulting in constitutive activation of NFκB. We show here that silencing ANXA1 increases the formation of stress fibers and focal adhesions, which may inhibit wound healing. ANXA1 regulated wound healing is dependent on the activation of ERK1/2. ANXA1 increases the activation of RhoA, which is dependent on ERK activation. Furthermore, active RhoA is important in NF-κB activation, where constitutively active RhoA potentiates NFκB activation, while dominant negative RhoA inhibits NFκB activation in response to CXCL12 stimulation and active MEKK plasmids. These findings establish a central role for ANXA1 in the cell migration through the activation of NFκB, ERK1/2 and RhoA.


Asunto(s)
Anexina A1/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Cicatrización de Heridas/fisiología , Proteína de Unión al GTP rhoA/metabolismo , Movimiento Celular , Femenino , Humanos , Células MCF-7 , Células Tumorales Cultivadas
2.
Int J Biol Sci ; 17(15): 4254-4270, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34803496

RESUMEN

Rationale: MicroRNAs (miRNAs) are endogenous ~22nt RNAs that play critical regulatory roles in various biological and pathological processes, including various cancers. Their function in renal cancer has not been fully elucidated. It has been reported that miR-196a can act as oncogenes or as tumor suppressors depending on their target genes. However, the molecular target for miR-196a and the underlying mechanism in miR-196a promoted cell migration and invasion in renal cancer is still not clear. Methods: The expression, survival and correlation between miR-196a and BRAM1 were investigated using TCGA analysis and validated by RT-PCR and western blot. To visualize the effect of Bram1 on tumor metastasis in vivo, NOD-SCID gamma (NSG) mice were intravenously injected with RCC4 cells (106 cells/mouse) or RCC4 overexpressing Bram1. In addition, cell proliferation assays, migration and invasion assays were performed to examine the role of miR-196a in renal cells in vitro. Furthermore, immunoprecipitation was done to explore the binding targets of Bram1. Results: TCGA gene expression data from renal clear cell carcinoma patients showed a lower level of Bram1 expression in patients' specimens compared to adjacent normal tissues. Moreover, Kaplan­Meier survival data clearly show that high expression of Bram1correlates to poor prognosis in renal carcinoma patients. Our mouse metastasis model confirmed that Bram1 overexpression resulted in an inhibition in tumor metastasis. Target-prediction analysis and dual-luciferase reporter assay demonstrated that Bram1 is a direct target of miR-196a in renal cells. Further, our in vitro functional assays revealed that miR-196a promotes renal cell proliferation, migration, and invasion. Rescue of Bram1 expression reversed miR-196a-induced cell migration. MiR-196a promotes renal cancer cell migration by directly targeting Bram1 and inhibits Smad1/5/8 phosphorylation and MAPK pathways through BMPR1A and EGFR. Conclusions: Our findings thus provide a new mechanism on the oncogenic role of miR-196a and the tumor-suppressive role of Bram1 in renal cancer cells. Dysregulated miR-196a and Bram1 represent potential prognostic biomarkers and may have therapeutic applications in renal cancer.


Asunto(s)
Carcinoma de Células Renales/patología , Proteínas de Ciclo Celular/metabolismo , Proteínas Co-Represoras/metabolismo , Proteínas de Unión al ADN/metabolismo , MicroARNs/metabolismo , Animales , Carcinoma de Células Renales/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Proteínas Co-Represoras/genética , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , MicroARNs/genética , Metástasis de la Neoplasia , Neoplasias Experimentales
3.
Oncotarget ; 7(19): 27007-20, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27105503

RESUMEN

MiRNAs are endogenous ~22 nt RNAs which play critical regulatory roles in a wide range of biological and pathological processes, which can act as oncogenes or tumor suppressor genes depending on their target genes. We have recently shown that ANXA1 inhibits the expression of miRNAs including miR196a. Here, we show that miR196a was highly expressed in ER+ MCF-7 breast cancer cells when compared to normal mammary gland cells, with expression levels negatively correlating to ANXA1. ANXA1 inhibits the biogenesis of oncogenic miR-196a by suppressing primary-miR196a indirectly through the stimulation of c-myc and NFkB expression and activity in breast cancer cells. In a negative feedback loop, miR-196a directly inhibits ANXA1 and enhances breast cancer cell proliferation in vitro. Finally, miR196a promotes breast tumor growth in vivo. This study reports a novel regulatory circuit between ANXA1, NF-kB, c-myc and miR-196a which regulates breast cancer cell proliferation and tumor growth.


Asunto(s)
Anexina A1/genética , Neoplasias de la Mama/genética , Proliferación Celular/genética , MicroARNs/genética , FN-kappa B/genética , Proteínas Proto-Oncogénicas c-myc/genética , Animales , Anexina A1/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Retroalimentación Fisiológica , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Ratones Endogámicos BALB C , Ratones Desnudos , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , Trasplante Heterólogo , Carga Tumoral/genética
4.
PLoS One ; 9(12): e114507, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25536365

RESUMEN

Annexin 1 (ANXA1) is an endogenous anti-inflammatory protein implicated in cancer. ANXA1 was previously shown to be regulated by hsa-miR-196a. However, whether ANXA1 itself regulates microRNA (miR) expression is unknown. Therefore, we investigated the regulation of miR by ANXA1 in MCF7 breast cancer cells. MCF7-EV (Empty vector) and MCF7-V5 (ANXA1-V5 expressing cells) were subjected to a miR microarray. Microarray analysis revealed a number of miRNAs which were dysregulated in MCF7-V5 cells. 2 novel miRNAs (miR562 and miR26b*) were validated, cloned and functionally characterized. As ANXA1 constitutively activates NF-κB activity to modulate breast cancer metastasis, we found that miR26b* and miR562 directly targeted the canonical NF-κB pathway by targeting the 3' UTR and inhibiting expression of Rel A (p65) and NF-κB1 (p105) respectively. MiR562 inhibited wound healing, which was reversed when ANXA1 was overexpressed. Overexpression of either miR562 or miR26b* in MCF-7 cells enhanced endothelial tube formation when cocultured with human umbilical cord endothelial cells while conversely, treatment of MCF7 cells with either anti-miR562 or anti-miR26b* inhibited endothelial tube formation after co-culture. Further analysis of miR562 revealed that miR562-transfected cell conditioned media enhances endothelial cell tube formation, indicating that miR562 increased angiogenic secreted factors from MCF-7 breast tumor cells. TNFα was increased upon overexpression of miR562, which was reversed when ANXA1 was co-transfected In conclusion, this data suggests that ANXA1-regulated miR26b* and miR562 may play a role in wound healing and tumor-induced endothelial cell tube formation by targeting NF-κB expression and point towards a potential therapeutic target for breast cancer.


Asunto(s)
Anexina A1/metabolismo , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/genética , MicroARNs/genética , FN-kappa B/metabolismo , Neovascularización Patológica/genética , Regiones no Traducidas 3'/genética , Secuencia de Bases , Neoplasias de la Mama/patología , Clonación Molecular , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Células MCF-7 , MicroARNs/metabolismo , Datos de Secuencia Molecular , Neovascularización Patológica/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reproducibilidad de los Resultados , Factor de Transcripción ReIA/metabolismo , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA