Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 31(1): 78-89, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36045587

RESUMEN

Androgen receptor signaling inhibitors (ARSIs) are standard of care for advanced prostate cancer (PCa) patients. Eventual resistance to ARSIs can include the expression of androgen receptor (AR) splice variant, AR-V7, expression as a recognized means of ligand-independent androgen signaling. We demonstrated that interleukin (IL)-6-mediated AR-V7 expression requires bone morphogenic protein (BMP) and CD105 receptor activity in both PCa and associated fibroblasts. Chromatin immunoprecipitation supported CD105-dependent ID1- and E2F-mediated expression of RBM38. Further, RNA immune precipitation demonstrated RBM38 binds the AR-cryptic exon 3 to enable AR-V7 generation. The forced expression of AR-V7 by primary prostatic fibroblasts diminished PCa sensitivity to ARSI. Conversely, downregulation of AR-V7 expression in cancer epithelia and associated fibroblasts was achieved by a CD105-neutralizing antibody, carotuximab. These compelling pre-clinical findings initiated an interventional study in PCa patients developing ARSI resistance. The combination of carotuximab and ARSI (i.e., enzalutamide or abiraterone) provided disease stabilization in four of nine assessable ARSI-refractory patients. Circulating tumor cell evaluation showed AR-V7 downregulation in the responsive subjects on combination treatment and revealed a three-gene panel that was predictive of response. The systemic antagonism of BMP/CD105 signaling can support ARSI re-sensitization in pre-clinical models and subjects that have otherwise developed resistance due to AR-V7 expression.


Asunto(s)
Antagonistas de Receptores Androgénicos , Endoglina , Neoplasias de la Próstata Resistentes a la Castración , Receptores Androgénicos , Humanos , Masculino , Resistencia a Antineoplásicos , Células Neoplásicas Circulantes/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Isoformas de Proteínas , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Proteínas de Unión al ARN , Endoglina/antagonistas & inhibidores , Antagonistas de Receptores Androgénicos/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico
3.
Mol Cell Biol ; 37(5)2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27956696

RESUMEN

The epithelial mesenchymal transition (EMT) promotes tumor migration and invasion by downregulating epithelial markers such as E-cadherin and upregulating mesenchymal markers such as vimentin. Cathepsin L (Cat L) is a cysteine protease that can proteolytically activate CCAAT displacement protein/cut homeobox transcription factor (CUX1). We hypothesized that nuclear Cat L may promote EMT via CUX1 and that this could be antagonized with the Cat L-specific inhibitor Z-FY-CHO. Mesenchymal prostate (ARCaP-M and ARCaP-E overexpressing Snail) and breast (MDA-MB-468, MDA-MB-231, and MCF-7 overexpressing Snail) cancer cells expressed lower E-cadherin activity, higher Snail, vimentin, and Cat L activity, and a p110/p90 active CUX1 form, compared to epithelial prostate (ARCaP-E and ARCaP-Neo) and breast (MCF-7 and MCF-7 Neo) cancer cells. There was increased binding of CUX1 to Snail and the E-cadherin promoter in mesenchymal cells compared to epithelial prostate and breast cells. Treatment of mesenchymal cells with the Cat L inhibitor Z-FY-CHO led to nuclear-to-cytoplasmic relocalization of Cat L, decreased binding of CUX1 to Snail and the E-cadherin promoter, reversed EMT, and decreased cell migration/invasion. Overall, our novel data suggest that a positive feedback loop between Snail-nuclear Cat L-CUX1 drives EMT, which can be antagonized by Z-FY-CHO. Therefore, Z-FY-CHO may be an important therapeutic tool to antagonize EMT and cancer progression.


Asunto(s)
Neoplasias de la Mama/patología , Catepsina L/metabolismo , Núcleo Celular/enzimología , Dipéptidos/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/patología , Inhibidores de Proteasas/farmacología , Proteínas Represoras/metabolismo , Antígenos CD , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Retroalimentación Fisiológica/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Mesodermo/efectos de los fármacos , Mesodermo/patología , Modelos Biológicos , Invasividad Neoplásica , Regiones Promotoras Genéticas/genética , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Unión Proteica/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Factores de Transcripción , Transcripción Genética/efectos de los fármacos
4.
J Clin Med ; 5(2)2016 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-26828526

RESUMEN

Epithelial-mesenchymal transition (EMT) is a complex molecular program that regulates changes in cell morphology and function during embryogenesis and tissue development. EMT also contributes to tumor progression and metastasis. Cells undergoing EMT expand out of and degrade the surrounding microenvironment to subsequently migrate from the primary site. The mesenchymal phenotype observed in fibroblasts is specifically important based on the expression of smooth muscle actin (α-SMA), fibroblast growth factor (FGF), fibroblast-specific protein-1 (FSP1), and collagen to enhance EMT. Although EMT is not completely dependent on EMT regulators such as Snail, Twist, and Zeb-1/-2, analysis of upstream signaling (i.e., TGF-ß, EGF, Wnt) is necessary to understand tumor EMT more comprehensively. Tumor epithelial-fibroblast interactions that regulate tumor progression have been identified during prostate cancer. The cellular crosstalk is significant because these events influence therapy response and patient outcome. This review addresses how canonical EMT signals originating from prostate cancer fibroblasts contribute to tumor metastasis and recurrence after therapy.

5.
Carcinogenesis ; 36(9): 1019-27, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26069256

RESUMEN

To develop new and effective chemopreventive agents against bone metastasis, we assessed the effects of muscadine grape skin extract (MSKE), whose main bioactive component is anthocyanin, on bone turnover, using prostate and breast cancer cell models overexpressing Snail transcription factor. MSKE has been shown previously to promote apoptosis in prostate cancer cells without affecting normal prostate epithelial cells. Snail is overexpressed in prostate and breast cancer, and is associated with increased invasion, migration and bone turnover/osteoclastogenesis. Cathepsin L (CatL) is a cysteine cathepsin protease that is overexpressed in cancer and involved in bone turnover. Snail overexpression in prostate (LNCaP, ARCaP-E) and breast (MCF-7) cancer cells led to increased CatL expression/activity and phosphorylated STAT-3 (pSTAT-3), compared to Neo vector controls, while the reverse was observed in C4-2 (the aggressive subline of LNCaP) cells with Snail knockdown. Moreover, CatL expression was higher in prostate and breast tumor tissue compared to normal tissue. MSKE decreased Snail and pSTAT3 expression, and abrogated Snail-mediated CatL activity, migration and invasion. Additionally, Snail overexpression promoted osteoclastogenesis, which was significantly inhibited by the MSKE as effectively as Z-FY-CHO, a CatL-specific inhibitor, or osteoprotegerin, a receptor activator of nuclear factor kappa B ligand (RANKL) antagonist. Overall, these novel findings suggest that Snail regulation of CatL may occur via STAT-3 signaling and can be antagonized by MSKE, leading to decreased cell invasion, migration and bone turnover. Therefore, inhibition using a natural product such as MSKE could potentially be a promising bioactive compound for bone metastatic cancer.


Asunto(s)
Anticarcinógenos/farmacología , Neoplasias Óseas/prevención & control , Neoplasias de la Mama/patología , Catepsina L/antagonistas & inhibidores , Extractos Vegetales/farmacología , Neoplasias de la Próstata/patología , Factores de Transcripción/antagonistas & inhibidores , Vitis/química , Animales , Anticarcinógenos/uso terapéutico , Apoptosis/efectos de los fármacos , Neoplasias Óseas/secundario , Catepsina L/biosíntesis , Catepsina L/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Quimioprevención/métodos , Femenino , Humanos , Células MCF-7 , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica , Osteoclastos/citología , Osteogénesis/efectos de los fármacos , Osteoprotegerina/farmacología , Extractos Vegetales/uso terapéutico , Ligando RANK/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción de la Familia Snail , Factores de Transcripción/biosíntesis
6.
PLoS One ; 9(8): e104987, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25122124

RESUMEN

Snail transcription factor is up-regulated in several cancers and associated with increased tumor migration and invasion via induction of epithelial-to-mesenchymal transition (EMT). MAPK (ERK1/2) signaling regulates cellular processes including cell motility, adhesion, and invasion. We investigated the regulation of ERK1/2 by Snail in breast cancer cells. ERK1/2 activity (p-ERK) was higher in breast cancer patient tissue as compared to normal tissue. Snail and p-ERK were increased in several breast cancer cell lines as compared to normal mammary epithelial cells. Snail knockdown in MDA-MB-231 and T47-D breast cancer cells decreased or re-localized p-ERK from the nuclear compartment to the cytoplasm. Snail overexpression in MCF-7 breast cancer cells induced EMT, increased cell migration, decreased cell adhesion and also increased tumorigenicity. Snail induced nuclear translocation of p-ERK, and the activation of its subcellular downstream effector, Elk-1. Inhibiting MAPK activity with UO126 or knockdown of ERK2 isoform with siRNA in MCF-7 Snail cells reverted EMT induced by Snail as shown by decreased Snail and vimentin expression, decreased cell migration and increased cell adhesion. Overall, our data suggest that ERK2 isoform activation by Snail in aggressive breast cancer cells leads to EMT associated with increased cell migration and decreased cell adhesion. This regulation is enhanced by positive feedback regulation of Snail by ERK2. Therefore, therapeutic targeting of ERK2 isoform may be beneficial for breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Núcleo Celular/enzimología , Transición Epitelial-Mesenquimal/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Factores de Transcripción/fisiología , Animales , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Complejo de la Endopetidasa Proteasomal/metabolismo , Transporte de Proteínas , Factores de Transcripción de la Familia Snail
7.
Cell Adh Migr ; 6(5): 433-41, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23076049

RESUMEN

Prostate cancer is the second most frequently diagnosed cancer and the sixth leading cause of death from cancer in men. Epithelial-mesenchymal transition (EMT) is a process by which cancer cells invade and migrate, and is characterized by loss of cell-cell adhesion molecules such as E-cadherin and increased expression of mesenchymal proteins such as vimentin; EMT is also associated with resistance to therapy. Snail, a master regulator of EMT, has been extensively studied and reported in cancers such as breast and colon; however, its role in prostate cancer is not as widely reported. The purpose of this review is to put together recent facts that summarize Snail signaling in human prostate cancer. Snail is overexpressed in prostate cancer and its expression and activity is controlled via phosphorylation and growth factor signaling. Snail is involved in its canonical role of inducing EMT in prostate cancer cells; however, it plays a role in non-canonical pathways that do not involve EMT such regulation of bone turnover and neuroendocrine differentiation. Thus, studies indicate that Snail signaling contributes to prostate cancer progression and metastasis and therapeutic targeting of Snail in prostate cancer holds promise in future.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/patología , Factores de Transcripción/metabolismo , Antineoplásicos/farmacología , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Humanos , Masculino , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factores de Transcripción de la Familia Snail , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética
8.
BMC Cancer ; 12: 336, 2012 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-22857708

RESUMEN

BACKGROUND: Maspin, a putative tumor suppressor that is down-regulated in breast and prostate cancer, has been associated with decreased cell motility. Snail transcription factor is a zinc finger protein that is increased in breast cancer and is associated with increased tumor motility and invasion by induction of epithelial-mesenchymal transition (EMT). We investigated the molecular mechanisms by which Snail increases tumor motility and invasion utilizing prostate cancer cells. METHODS: Expression levels were analyzed by RT-PCR and western blot analyses. Cell motility and invasion assays were performed, while Snail regulation and binding to maspin promoter was analyzed by luciferase reporter and chromatin immunoprecipitation (ChIP) assays. RESULTS: Snail protein expression was higher in different prostate cancer cells lines as compared to normal prostate epithelial cells, which correlated inversely with maspin expression. Snail overexpression in 22Rv1 prostate cancer cells inhibited maspin expression and led to increased migration and invasion. Knockdown of Snail in DU145 and C4-2 cancer cells resulted in up-regulation of maspin expression, concomitant with decreased migration. Transfection of Snail into 22Rv1 or LNCaP cells inhibited maspin promoter activity, while stable knockdown of Snail in C4-2 cells increased promoter activity. ChIP analysis showed that Snail is recruited to the maspin promoter in 22Rv1 cells. CONCLUSIONS: Overall, this is the first report showing that Snail can negatively regulate maspin expression by directly repressing maspin promoter activity, leading to increased cell migration and invasion. Therefore, therapeutic targeting of Snail may be useful to re-induce expression of maspin tumor suppressor and prevent prostate cancer tumor progression.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Serpinas/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Línea Celular Tumoral , Movimiento Celular , Células Epiteliales/metabolismo , Expresión Génica , Silenciador del Gen , Humanos , Masculino , Regiones Promotoras Genéticas , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...