Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 46(4): 1895-1911, 2018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29309643

RESUMEN

Androgen receptor (AR) splice variants (ARVs) are implicated in development of castration-resistant prostate cancer (CRPC). Upregulation of ARVs often correlates with persistent AR activity after androgen deprivation therapy (ADT). However, the genomic and epigenomic characteristics of ARV-dependent cistrome and the disease relevance of ARV-mediated transcriptome remain elusive. Through integrated chromatin immunoprecipitation coupled sequencing (ChIP-seq) and RNA sequencing (RNA-seq) analysis, we identified ARV-preferential-binding sites (ARV-PBS) and a set of genes preferentially transactivated by ARVs in CRPC cells. ARVs preferentially bind to enhancers located in nucleosome-depleted regions harboring the full AR-response element (AREfull), while full-length AR (ARFL)-PBS are enhancers resided in closed chromatin regions containing the composite FOXA1-nnnn-AREhalf motif. ARV-PBS exclusively overlapped with AR binding sites in castration-resistant (CR) tumors in patients and ARV-preferentially activated genes were up-regulated in abiraterone-resistant patient specimens. Expression of ARV-PBS target genes, such as oncogene RAP2A and cell cycle gene E2F7, were significantly associated with castration resistance, poor survival and tumor progression. We uncover distinct genomic and epigenomic features of ARV-PBS, highlighting that ARVs are useful tools to depict AR-regulated oncogenic genome and epigenome landscapes in prostate cancer. Our data also suggest that the ARV-preferentially activated transcriptional program could be targeted for effective treatment of CRPC.


Asunto(s)
Androstenos/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/metabolismo , Animales , Sitios de Unión , Línea Celular Tumoral , Cromatina/metabolismo , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Epigénesis Genética , Genómica , Humanos , Masculino , Ratones SCID , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Isoformas de Proteínas/metabolismo , Empalme del ARN , Receptores Androgénicos/genética , Proteínas de Unión al GTP rap/fisiología
2.
J Urol ; 193(2): 690-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25132238

RESUMEN

PURPOSE: Spliced variant forms of androgen receptor were recently identified in castration resistant prostate cancer cell lines and clinical samples. We identified the cistrome and gene signature of androgen receptor splice variants in castration resistant prostate cancer cell lines and determined the clinical significance of androgen receptor splice variant regulated genes. MATERIALS AND METHODS: The castration resistant prostate cancer cell line 22Rv1, which expresses full-length androgen receptor and androgen receptor splice variants endogenously, was used as the research model. We established 22Rv1-ARFL(-)/ARV(+) and 22Rv1-ARFL(-)/ARV(-) through RNA interference. Chromatin immunoprecipitation coupled with next generation sequencing and microarray techniques were used to identify the cistrome and gene expression profiles of androgen receptor splice variants in the absence of androgen. RESULTS: Androgen receptor splice variant binding sites were identified in 22Rv1-ARFL(-)/ARV(+). A gene set was regulated uniquely by androgen receptor splice variants but not by full-length androgen receptor in the absence of androgen. Integrated analysis revealed that some genes were directly modulated by androgen receptor splice variants. Unsupervised clustering analysis showed that the androgen receptor splice variant gene signature differentiated benign from malignant prostate tissue as well as localized prostate cancer from metastatic castration resistant prostate cancer specimens. Some genes that were modulated uniquely by androgen receptor splice variants also correlated with histological grade and biochemical failure. CONCLUSIONS: Androgen receptor splice variants can bind to DNA independent of full-length androgen receptor in the absence of androgen and modulate a unique set of genes that is not regulated by full-length androgen receptor. The androgen receptor splice variant gene signature correlates with disease progression. It distinguishes primary cancer from castration resistant prostate cancer specimens and benign from malignant prostate specimens.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/genética , Transcriptoma , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Isoformas de Proteínas , Células Tumorales Cultivadas
3.
Nat Cell Biol ; 10(9): 1076-82, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19160488

RESUMEN

Proper control of entry into and progression through mitosis is essential for normal cell proliferation and the maintenance of genome stability. The mammalian mitotic kinase Polo-like kinase 1 (Plk1) is involved in multiple stages of mitosis5. Here we report that Forkhead Box M1 (FoxM1), a substrate of Plk1, controls a transcriptional programme that mediates Plk1-dependent regulation of cell-cycle progression. The carboxy-terminal domain of FoxM1 binds Plk1, and phosphorylation of two key residues in this domain by Cdk1 is essential for Plk1-FoxM1 interaction. Formation of the Plk1-FoxM1 complex allows for direct phosphorylation of FoxM1 by Plk1 at G2/M and the subsequent activation of FoxM1 activity, which is required for expression of key mitotic regulators, including Plk1 itself. Thus, Plk1-dependent regulation of FoxM1 activity provides a positive-feedback loop ensuring tight regulation of transcriptional networks essential for orderly mitotic progression.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Factores de Transcripción Forkhead/metabolismo , Mitosis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transcripción Genética , Secuencia de Aminoácidos , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/genética , Fase G2 , Regulación de la Expresión Génica , Células HeLa , Humanos , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Quinasa Tipo Polo 1
4.
N Engl J Med ; 362(13): 1192-202, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20357281

RESUMEN

BACKGROUND: We conducted a study to determine whether dutasteride reduces the risk of incident prostate cancer, as detected on biopsy, among men who are at increased risk for the disease. METHODS: In this 4-year, multicenter, randomized, double-blind, placebo-controlled, parallel-group study, we compared dutasteride, at a dose of 0.5 mg daily, with placebo. Men were eligible for inclusion in the study if they were 50 to 75 years of age, had a prostate-specific antigen (PSA) level of 2.5 to 10.0 ng per milliliter, and had had one negative prostate biopsy (6 to 12 cores) within 6 months before enrollment. Subjects underwent a 10-core transrectal ultrasound-guided biopsy at 2 and 4 years. RESULTS: Among 6729 men who underwent a biopsy or prostate surgery, cancer was detected in 659 of the 3305 men in the dutasteride group, as compared with 858 of the 3424 men in the placebo group, representing a relative risk reduction with dutasteride of 22.8% (95% confidence interval, 15.2 to 29.8) over the 4-year study period (P<0.001). Overall, in years 1 through 4, among the 6706 men who underwent a needle biopsy, there were 220 tumors with a Gleason score of 7 to 10 among 3299 men in the dutasteride group and 233 among 3407 men in the placebo group (P=0.81). During years 3 and 4, there were 12 tumors with a Gleason score of 8 to 10 in the dutasteride group, as compared with only 1 in the placebo group (P=0.003). Dutasteride therapy, as compared with placebo, resulted in a reduction in the rate of acute urinary retention (1.6% vs. 6.7%, a 77.3% relative reduction). The incidence of adverse events was similar to that in studies of dutasteride therapy for benign prostatic hyperplasia, except that in our study, as compared with previous studies, the relative incidence of the composite category of cardiac failure was higher in the dutasteride group than in the placebo group (0.7% [30 men] vs. 0.4% [16 men], P=0.03). CONCLUSIONS: Over the course of the 4-year study period, dutasteride reduced the risk of incident prostate cancer detected on biopsy and improved the outcomes related to benign prostatic hyperplasia. (ClinicalTrials.gov number, NCT00056407.)


Asunto(s)
Inhibidores de 5-alfa-Reductasa , Azaesteroides/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Hiperplasia Prostática/tratamiento farmacológico , Neoplasias de la Próstata/prevención & control , Anciano , Azaesteroides/efectos adversos , Biopsia , Método Doble Ciego , Dutasterida , Inhibidores Enzimáticos/efectos adversos , Disfunción Eréctil/inducido químicamente , Insuficiencia Cardíaca/inducido químicamente , Humanos , Masculino , Persona de Mediana Edad , Próstata/patología , Antígeno Prostático Específico/sangre , Hiperplasia Prostática/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Isoformas de Proteínas , Riesgo , Resultado del Tratamiento
5.
Int J Mol Sci ; 14(7): 14833-59, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23863692

RESUMEN

The androgen receptor (AR) is important in the development of the prostate by regulating transcription, cellular proliferation, and apoptosis. AR undergoes posttranslational modifications that alter its transcription activity, translocation to the nucleus and stability. The posttranslational modifications that regulate these events are of utmost importance to understand the functional role of AR and its activity. The majority of these modifications occur in the activation function-1 (AF1) region of the AR, which contains the transcriptional activation unit 1 (TAU1) and 5 (TAU5). Identification of the modifications that occur to these regions may increase our understanding of AR activation in prostate cancer and the role of AR in the progression from androgen-dependent to castration-resistant prostate cancer (CRPC). Most of the posttranslational modifications identified to date have been determined using the full-length AR in androgen dependent cells. Further investigations into the role of posttranslational modifications in androgen-independent activation of full-length AR and constitutively active splicing variants are warranted, findings from which may provide new therapeutic options for CRPC.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Acetilación , Humanos , Masculino , Metilación , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Neoplasias de la Próstata/patología , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Receptores Androgénicos/química , Receptores Androgénicos/genética , Sumoilación , Ubiquitinación
6.
Biochim Biophys Acta ; 1813(11): 1961-4, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21238503

RESUMEN

Forkhead box O-class (FOXO) proteins are evolutionally conserved transcription factors. They belong to a family of proteins consisting of FOXO1, FOXO3a, FOXO4 and FOXO6 in humans. Increasing evidence suggests that FOXO proteins function as tumor suppressors by transcriptionally regulating expression of genes involved in cell cycle arrest, apoptosis, DNA repair and oxidative stress resistance. Activation of various protein kinases, including Akt, IκB kinase (IKK) and ERK, leads to phosphorylation of FOXO proteins and their ubiquitination mediated by E3 ligases such as SKP2 and MDM2 in human primary tumors and cancer cell lines. As a result, the tumor suppressor functions of FOXO proteins are either diminished or abrogated due to their ubiquitination-proteasome degradation, thereby favoring cell transformation, proliferation and survival. Thus, ubiquitination and proteasome degradation of FOXO proteins play an important role in tumorigenesis and represent a viable target for cancer treatment. This article is part of a Special Issue entitled: PI3K-AKT-FoxO axis in cancer and aging.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Ubiquitinación , Animales , Proteína Forkhead Box O1 , Humanos , Modelos Biológicos , Unión Proteica
7.
Prostate ; 72(2): 225-32, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21630295

RESUMEN

BACKGROUND: Androgens control homeostasis of the normal prostate and growth of prostate cancer (PCa) through the androgen receptor (AR) by regulating gene networks involving in cell proliferation, differentiation, and survival. We demonstrated previously that expression of Skp2, a key protein regulating cell entry into the S phase, is inhibited by androgens in an AR-dependent manner (Oncogene, 2004; 23(12): 2161-2176). However, the underlying mechanism of this regulation is unknown. METHODS: Using the LNCaP PCa cell line as a working model, the effect of androgens on the expression of Skp2 was examined by Western and Northern blot analyses. Cell cycle was measured by fluorescence-activated cell sorting (FACS). Gene transfection was performed by electroporation to manipulate the expression levels of proteins studied. RESULTS: At physiological levels androgens markedly repressed Skp2 expression but slightly induced Skp2 expression at subphysiological levels. Androgens modestly decreased the stability of the Skp2 protein. Androgenic repression of Skp2 expression was completely abolished by E1A-mediated inactivation of pocket proteins including RB, p130, and p107. Moreover, ectopic expression of p107 inhibited Skp2 expression, and silencing of p107 partially blocked androgenic repression of Skp2. CONCLUSIONS: Our data indicate that androgens repress Skp2 expression via p107-dependent and -independent pathways in PCa cells. These regulatory mechanisms may be targeted for the development of new therapeutics of androgen-refractory PCa.


Asunto(s)
Ciclo Celular/fisiología , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Proteína p107 Similar a la del Retinoblastoma/metabolismo , Proteínas Quinasas Asociadas a Fase-S/biosíntesis , Northern Blotting , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Masculino , Metribolona/farmacología , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Congéneres de la Testosterona/farmacología
8.
Artículo en Inglés | MEDLINE | ID: mdl-22461839

RESUMEN

Azadirachta indica (common name: neem) leaves have been found to possess immunomodulatory, anti-inflammatory and anti-carcinogenic properties. The present study evaluates anti-angiogenic potential of ethanol extract of neem leaves (EENL) in human umbilical vein endothelial cells (HUVECs). Treatment of HUVECs with EENL inhibited VEGF induced angiogenic response in vitro and in vivo. The in vitro proliferation, invasion and migration of HUVECs were suppressed with EENL. Nuclear fragmentation and abnormally small mitochondria with dilated cristae were observed in EENL treated HUVECs by transmission electron microscopy. Genome-wide mRNA expression profiling after treatment with EENL revealed differentially regulated genes. Expression changes of the genes were validated by quantitative real-time polymerase chain reaction. Additionally, increase in the expression of HMOX1, ATF3 and EGR1 proteins were determined by immunoblotting. Analysis of the compounds in the EENL by mass spectrometry suggests the presence of nimbolide, 2',3'-dehydrosalannol, 6-desacetyl nimbinene and nimolinone. We further confirmed antiproliferative activity of nimbolide and 2',3'-dehydrosalannol in HUVECs. Our results suggest that EENL by regulating the genes involved in cellular development and cell death functions could control cell proliferation, attenuate the stimulatory effects of VEGF and exert antiangiogenic effects. EENL treatment could have a potential therapeutic role during cancer progression.

9.
Endocr Rev ; 28(7): 778-808, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17940184

RESUMEN

Androgens, acting through the androgen receptor (AR), are responsible for the development of the male phenotype during embryogenesis, the achievement of sexual maturation at puberty, and the maintenance of male reproductive function and behavior in adulthood. In addition, androgens affect a wide variety of nonreproductive tissues. Moreover, aberrant androgen action plays a critical role in multiple pathologies, including prostate cancer and androgen insensitivity syndromes. The formation of a productive AR transcriptional complex requires the functional and structural interaction of the AR with its coregulators. In the last decade, an overwhelming and ever increasing number of proteins have been proposed to possess AR coactivating or corepressing characteristics. Intriguingly, a vast diversity of functions has been ascribed to these proteins, indicating that a multitude of cellular functions and signals converge on the AR to regulate its function. The current review aims to provide an overview of the AR coregulator proteins identified to date and to propose a classification of these AR coregulator proteins according to the function(s) ascribed to them. Taken together, this approach will increase our understanding of the cellular pathways that converge on the AR to ensure an appropriate transcriptional response to androgens.


Asunto(s)
Andrógenos/metabolismo , Receptores Androgénicos/metabolismo , Factores de Transcripción/metabolismo , Andrógenos/fisiología , Regulación de la Expresión Génica , Humanos , Masculino , Receptores Androgénicos/efectos de los fármacos , Receptores Androgénicos/genética
10.
J Carcinog ; 10: 20, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21886458

RESUMEN

The androgen receptor (AR) signaling axis plays a critical role in the development, function and homeostasis of the prostate. The classical action of AR is to regulate gene transcriptional processes via AR nuclear translocation, binding to androgen response elements on target genes and recruitment of, or crosstalk with, transcription factors. Prostate cancer initiation and progression is also uniquely dependent on AR. Androgen deprivation therapy remains the standard of care for treatment of advanced prostate cancer. Despite an initial favorable response, almost all patients invariably progress to a more aggressive, castrate-resistant phenotype. Considerable evidence now supports the concept that development of castrate-resistant prostate cancer (CRPC) is causally related to continued transactivation of AR. Understanding the critical events and complexities of AR signaling in the progression to CRPC is essential in developing successful future therapies. This review provides a synopsis of AR structure and signaling in prostate cancer progression, with a special focus on recent findings on the role of AR in CRPC. Clinical implications of these findings and potential directions for future research are also outlined.

11.
Prostate ; 70(9): 959-70, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20166126

RESUMEN

BACKGROUND: Deregulated androgen receptor (AR) action is critical for prostate cancer (PCa) progression. Aberrant expression of AR-associated coregulators contributes to AR activity in PCa. The mechanisms underlying coregulator expression in PCa are under intense investigation as they may lead to alternative means of targeting AR activity in PCa cells. We have recently shown that over 30% of coregulator expression in the PCa cell line LNCaP is subject to androgen regulation. METHODS: Using multiple PCa cell lines as well as xenograft models, non-malignant prostate epithelial cell lines and androgen-responsive tissues derived from a male Wistar rat model system, we explored the effect of androgen stimulation and androgen deprivation on the expression of the core coactivators SRC1, SRC2, SRC3, CBP, and p300. RESULTS: Androgen stimulation of model systems representing PCa led to a decrease in the expression of SRC1, SRC2, SRC3, CBP, and p300, whereas androgen deprivation induced the expression of these coactivators. In contrast, expression of these coregulators remained largely unaffected following changes in the androgenic milieu in AR-positive models representing non-malignant prostate cells and tissues. CONCLUSIONS: Our data indicate differences in the regulation of coregulator expression between neoplastic and normal prostate cells. These findings emphasize the important potential of targeting the mechanisms regulating coregulator expression for therapeutic intervention in PCa.


Asunto(s)
Coactivadores de Receptor Nuclear/genética , Próstata/metabolismo , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Factores de Transcripción p300-CBP/genética , Animales , Western Blotting , Fraccionamiento Celular , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Masculino , Coactivadores de Receptor Nuclear/metabolismo , Neoplasias de la Próstata/metabolismo , Ratas , Ratas Wistar , Receptores Androgénicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción p300-CBP/metabolismo
12.
Future Oncol ; 6(5): 823-36, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20465393

RESUMEN

Detection of lymph node metastases indicates poor prognosis for prostate cancer patients. Therefore, elucidation of the mechanism(s) of lymph node metastasis is important to understand the progression of prostate cancer and also to develop therapeutic interventions. In this article, the known mechanisms for lymph node metastasis are discussed and the involvement of lymphatic vessels in prostate cancer lymph node metastasis is comprehensively summarized. In addition, contradictory findings regarding the importance of lymphangiogenesis in facilitating lymph node metastasis in prostate cancer are pointed out and reconcilation is attempted.


Asunto(s)
Linfangiogénesis/fisiología , Metástasis Linfática/patología , Neoplasias de la Próstata/patología , Humanos , Masculino
13.
Mol Ther ; 17(12): 2041-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19773744

RESUMEN

Prostate cancer cells overexpress the measles virus (MV) receptor CD46. Herein, we evaluated the antitumor activity of an oncolytic derivative of the MV Edmonston (MV-Edm) vaccine strain engineered to express the human sodium iodide symporter (NIS; MV-NIS virus). MV-NIS showed significant cytopathic effect (CPE) against prostate cancer cell lines in vitro. Infected cells effectively concentrated radioiodide isotopes as measured in vitro by Iodide-125 ((125)I) uptake assays. Virus localization and spread in vivo could be effectively followed by imaging of (123)I uptake. In vivo administration of MV-NIS either locally or systemically (total dose of 9 x 10(6) TCID(50)) resulted in significant tumor regression (P < 0.05) and prolongation of survival (P < 0.01). Administration of (131)I further enhanced the antitumor effect of MV-NIS virotherapy (P < 0.05). In conclusion, MV-NIS is an oncolytic vector with significant antitumor activity against prostate cancer, which can be further enhanced by (131)I administration. The NIS transgene allows viral localization and monitoring by noninvasive imaging which can facilitate dose optimization in a clinical setting.


Asunto(s)
Diagnóstico por Imagen , Radioisótopos de Yodo/metabolismo , Vacuna Antisarampión/genética , Virus del Sarampión/genética , Viroterapia Oncolítica/métodos , Neoplasias de la Próstata/terapia , Simportadores/genética , Animales , Proliferación Celular , Chlorocebus aethiops , Efecto Citopatogénico Viral , Ingeniería Genética , Humanos , Radioisótopos de Yodo/uso terapéutico , Masculino , Virus del Sarampión/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Próstata/patología , Simportadores/metabolismo , Células Tumorales Cultivadas , Rayos Ultravioleta , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Mol Endocrinol ; 23(4): 572-83, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19164447

RESUMEN

Aberrant coregulator expression that occurs during prostate cancer (PCa) progression correlates with poor prognosis and aggressive disease. This has been attributed to the ability to regulate androgen receptor-mediated transcription. We have shown previously that the androgenic milieu regulates the expression of the coactivators p300 and FHL2, with severe consequences for PCa cell proliferation and androgen receptor transcriptional activity. To determine the extent of androgen dependency of coregulator genes, we designed a cDNA-mediated annealing, selection, extension, and ligation RNA profiling array that probes the expression of 186 coregulators. Using this assay, we demonstrated androgen control over approximately 30% of coregulator genes in PCa cells. For a subset of 15 functionally diverse coregulators, androgen regulation was confirmed using real-time RT-PCR and immunoblotting. The extent, dose dependency, and kinetics by which androgens affect coregulator expression differed widely, indicating diverse molecular mechanisms underlying these effects. Moreover, differences in coregulator expression were observed between isogenic androgen-dependent and castration-recurrent PCa cells. Small interfering RNA-mediated changes in coregulator expression had profound effects on cell proliferation, which were most pronounced in castration-recurrent cells. Taken together, our integrated approach combining expression profiling, characterization of androgen-dependent coregulator expression, and validation of the importance of altered coregulator expression for cell proliferation identified several potential novel therapeutic targets for PCa treatment.


Asunto(s)
Andrógenos/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Neoplasias de la Próstata/metabolismo , Factores de Transcripción , Línea Celular Tumoral , Proliferación Celular , Humanos , Masculino , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Endocr Relat Cancer ; 16(1): 123-37, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18845648

RESUMEN

Urothelial carcinoma (UC) of the bladder is approximately three times more common in men than women. While the etiology for this gender difference in incidence remains unknown, a role for androgen receptor (AR) signaling has been suggested. The mechanisms by which AR activity is regulated in UC cells, however, are largely elusive. Here, we explore the significance of coregulators that are critical for the formation of a functional AR transcriptional complex, in UC cells. Using two AR-positive UC cell lines, TCC-SUP and UMUC3, we demonstrate the expression of the coactivators NCOA1, NCOA2, NCOA3, CREBBP, and EP300 in UC cells. small interfering RNA-mediated knockdown of the AR or any of these coactivators markedly impacted cell viability and abrogated androgen-dependent cell proliferation. Noteworthy, contrary to AR-positive prostate cancer cells, expression of these AR-associated coactivators was not androgen regulated in UC cells. To assess the clinical relevance of coactivator expression, we performed immunohistochemistry on paraffin-embedded sections from 55 patients with UC of the bladder. We found that while 24 out of 55 (44%) of tumors expressed the AR, each of the coactivators was expressed by 85-100% of the bladder cancers. Moreover, we noted a significant downregulation of NCOA1 expression in tumors versus adjacent, non-tumor bladder urothelium, with a mean of 68% (range 0-100) of tumor cells demonstrating NCOA1 staining versus a mean of 81% (range 0-90) of non-tumor cells (P=0.03). Taken together, our data suggest an important role for AR-associated coactivators in UC and point toward differences in the regulation of AR activity between bladder and prostate cancer cells.


Asunto(s)
Histona Acetiltransferasas/genética , Coactivador 2 del Receptor Nuclear/genética , Receptores Androgénicos/metabolismo , Transactivadores/genética , Factores de Transcripción/genética , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/fisiopatología , Andrógenos/metabolismo , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Línea Celular Tumoral , Proteína p300 Asociada a E1A/genética , Proteína p300 Asociada a E1A/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Histona Acetiltransferasas/metabolismo , Humanos , Inmunohistoquímica , Masculino , Coactivador 1 de Receptor Nuclear , Coactivador 2 del Receptor Nuclear/metabolismo , Coactivador 3 de Receptor Nuclear , ARN Interferente Pequeño , Transducción de Señal/fisiología , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Urotelio/fisiología
16.
Prostate ; 69(16): 1730-43, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19676081

RESUMEN

BACKGROUND: In the prostate, androgens play a crucial role in normal and cancerous growth; hence the androgenic pathway has become a target of therapeutic intervention. Dutasteride is a 5 alpha-reductase (5AR) inhibitor currently being evaluated both for chemoprevention and treatment of prostate cancer. Dutasteride inhibits both 5AR I and II enzymes, effectively blocking conversion of testosterone to dihydrotestosterone (DHT) in the prostate. This greatly reduces the amount of the active ligand DHT available for binding to the androgen receptor (AR) and stimulating proliferation, making this a good candidate for chemoprevention of prostate cancer. In this study, we sought to determine how dutasteride is functioning at the molecular level, using a prostate cancer xenograft model. METHODS: Androgen-responsive LuCaP 35 xenograft tumors were grown in Balb/c mice. Subcutaneously implanted time-release pellets were used for drug delivery. Microarray analysis was performed using the Affymetrix HG-U133Av2 platform to examine changes in gene expression in tumors following dutasteride treatment. RESULTS: Dutasteride significantly reduced tumor growth in LuCaP 35 xenografts by affecting genes involved in apoptotic, cytoskeletal remodeling, and cell cycle pathways among others. Notably, genes in the Rho GTPase signaling pathway, shown to be important in androgen-deprivation conditions, were significantly up-regulated. CONCLUSION: We have identified multiple pathways outside of the androgenic pathway in prostate cancer xenografts affected by treatment with dutasteride. These findings provide insights into the function of dutasteride within the tumor microenvironment, potentially allowing for development of agents that can be used in combination with this drug to further enhance its effectiveness.


Asunto(s)
Azaesteroides/administración & dosificación , Colestenona 5 alfa-Reductasa/antagonistas & inhibidores , Inhibidores Enzimáticos/administración & dosificación , Trasplante de Neoplasias , Neoplasias de la Próstata/metabolismo , Trasplante Heterólogo , Andrógenos/deficiencia , Animales , División Celular/efectos de los fármacos , Preparaciones de Acción Retardada , Implantes de Medicamentos , Dutasterida , Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Análisis por Micromatrices , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba , Proteínas de Unión al GTP rho/metabolismo
17.
N Engl J Med ; 355(16): 1647-59, 2006 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-17050889

RESUMEN

BACKGROUND: Dehydroepiandrosterone (DHEA) and testosterone are widely promoted as antiaging supplements, but the long-term benefits, as compared with potential harm, are unknown. METHODS: We performed a 2-year, placebo-controlled, randomized, double-blind study involving 87 elderly men with low levels of the sulfated form of DHEA and bioavailable testosterone and 57 elderly women with low levels of sulfated DHEA. Among the men, 29 received DHEA, 27 received testosterone, and 31 received placebo. Among the women, 27 received DHEA and 30 received placebo. Outcome measures included physical performance, body composition, bone mineral density (BMD), glucose tolerance, and quality of life. RESULTS: As compared with the change from baseline to 24 months in the placebo group, subjects who received DHEA for 2 years had an increase in plasma levels of sulfated DHEA by a median of 3.4 microg per milliliter (9.2 micromol per liter) in men and by 3.8 microg per milliliter (10.3 micromol per liter) in women. Among men who received testosterone, the level of bioavailable testosterone increased by a median of 30.4 ng per deciliter (1.1 nmol per liter), as compared with the change in the placebo group. A separate analysis of men and women showed no significant effect of DHEA on body-composition measurements. Neither hormone altered the peak volume of oxygen consumed per minute, muscle strength, or insulin sensitivity. Men who received testosterone had a slight increase in fat-free mass, and men in both treatment groups had an increase in BMD at the femoral neck. Women who received DHEA had an increase in BMD at the ultradistal radius. Neither treatment improved the quality of life or had major adverse effects. CONCLUSIONS: Neither DHEA nor low-dose testosterone replacement in elderly people has physiologically relevant beneficial effects on body composition, physical performance, insulin sensitivity, or quality of life. (ClinicalTrials.gov number, NCT00254371 [ClinicalTrials.gov].).


Asunto(s)
Envejecimiento/efectos de los fármacos , Composición Corporal/efectos de los fármacos , Deshidroepiandrosterona/administración & dosificación , Terapia de Reemplazo de Hormonas , Aptitud Física , Testosterona/administración & dosificación , Adulto , Anciano , Densidad Ósea/efectos de los fármacos , Deshidroepiandrosterona/efectos adversos , Deshidroepiandrosterona/sangre , Método Doble Ciego , Estradiol/sangre , Femenino , Terapia de Reemplazo de Hormonas/efectos adversos , Humanos , Resistencia a la Insulina , Masculino , Persona de Mediana Edad , Calidad de Vida , Testosterona/efectos adversos , Testosterona/sangre , Insuficiencia del Tratamiento
18.
Cancer Invest ; 27(5): 549-60, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19229700

RESUMEN

To identify the relevant CpG sites as molecular markers, for the diagnosis and to distinguish the indolent and aggressive prostate tumors, we have determined the methylation status of 8 genes, including FLNC, EFS, ECRG4, RARB2, PITX2, GSTP1, PDLIM4, and KCNMA1 in 32 nonrecurrent, 32 recurrent primary prostate tumors, and 32 benign prostate tissues using EpiTYPER technology. Specific CpG site hypermethylation of RARB2 and GSTP1 CpG sites were useful for diagnosis of prostate cancer. Furthermore, CpG site hypermethylation of genes FLNC, EFS, ECRG4, PITX2, PDLIM4, and KCNMA1 were associated with prediction of biochemical, local, and systemic recurrence of prostate cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Islas de CpG , Metilación de ADN , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/genética , ADN de Neoplasias/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Recurrencia Local de Neoplasia/diagnóstico , Recurrencia Local de Neoplasia/genética , Reacción en Cadena de la Polimerasa , Hiperplasia Prostática/diagnóstico , Hiperplasia Prostática/genética , Factores de Riesgo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
19.
Cancer Res ; 67(20): 10067-77, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17942941

RESUMEN

Systemic prostate cancer therapy requires androgen ablation, which inhibits the production or action of androgens. Prostate cancer ultimately relapses during androgen ablation, and an androgen depletion-independent (ADI) phenotype emerges. Aberrant androgen receptor (AR) activation underlies therapy resistance at this stage of the disease, and mounting evidence implicates the large and highly disordered AR NH2-terminal domain (NTD) as a key mediator of this activity. In this study, we investigated the role of the NTD transactivation unit 5 (TAU5) domain in mediating AR transcriptional activity in cell-based models of prostate cancer progression. AR replacement and Gal4-based promoter tethering experiments revealed that AR TAU5 had a dichotomous function, inhibiting ligand-dependent AR activity in androgen-dependent prostate cancer cells, while enhancing ligand-independent AR activity in ADI prostate cancer cells. Molecular dissection of TAU5 showed that a WxxLF motif was fully responsible for its ligand-independent activity. Mechanistically, WxxLF did not rely on an interaction with the AR ligand-binding domain to mediate ligand-independent AR activity. Rather, WxxLF functioned as an autonomous transactivation domain. These data show that ligand-dependent and ligand-independent AR activation rely on fundamentally distinct mechanisms, and define WxxLF as the major transactivation motif within the AR TAU5 domain.


Asunto(s)
Andrógenos/deficiencia , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Antagonistas de Receptores Androgénicos , Animales , Secuencia de Bases , Humanos , Ligandos , Masculino , Ratones , Datos de Secuencia Molecular , Neoplasias de la Próstata/genética , Estructura Terciaria de Proteína , Receptores Androgénicos/genética , Activación Transcripcional
20.
Cancer Res ; 67(7): 3422-30, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17409453

RESUMEN

Standard therapy for nonorgan confined prostate cancer aims to block the production or action of androgens. Although initially successful, antiandrogen therapy eventually fails and androgen depletion independent (ADI) disease emerges. Remarkably, ADI prostate cancers still rely on a functional androgen receptor (AR). Aberrant expression of coregulatory proteins required for the formation of productive AR transcriptional complexes is critical for ADI AR activation. Previously, we have shown that the transcriptional coactivator p300 is required for ADI activation of the AR and is up-regulated in prostate cancer, in which its expression is associated with cell proliferation and predicts aggressive tumor features. The mechanism responsible for the deregulated expression of p300, however, remains elusive. Here, we show that p300 expression in prostate cancer cells is subject to androgen regulation. In several prostate cancer model systems, addition of synthetic and natural androgens led to decreased expression of p300 in a time-dependent and dose-dependent manner. Experiments using AR antagonists or small interfering RNA targeting the AR revealed that down-regulation of p300 depends entirely on the presence of a functional AR. It is noteworthy that androgens down-regulated p300 protein expression while leaving messenger levels unaltered. Conversely, both short-term and long-term androgen deprivation resulted in marked up-regulation of p300 expression. The androgen deprivation-induced increase in p300 expression was not affected by the addition of cytokines or growth factors or by cotreatment with antiandrogens. Moreover, increased p300 expression upon androgen starvation is crucial for prostate cancer cell proliferation, as loss of p300 expression severely reduces expression of cyclins governing G(1)-S and G(2)-M cell cycle transition and decreases 5-bromo-2'-deoxyuridine incorporation.


Asunto(s)
Andrógenos/deficiencia , Proteína p300 Asociada a E1A/biosíntesis , Neoplasias de la Próstata/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Regulación hacia Abajo , Proteína p300 Asociada a E1A/genética , Humanos , Masculino , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Androgénicos/biosíntesis , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA