Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Prostate ; 80(13): 1058-1070, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32692871

RESUMEN

BACKGROUND: Most prostate cancers express androgen receptor (AR), and our previous studies have focused on identifying transcription factors that modify AR function. We have shown that nuclear factor I/B (NFIB) regulates AR activity in androgen-dependent prostate cancer cells in vitro. However, the status of NFIB in prostate cancer was unknown. METHODS: We immunostained a tissue microarray including normal, hyperplastic, prostatic intraepithelial neoplasia, primary prostatic adenocarcinoma, and castration-resistant prostate cancer tissue samples for NFIB, AR, and synaptophysin, a marker of neuroendocrine differentiation. We interrogated publically available data sets in cBioPortal to correlate NFIB expression and AR and neuroendocrine prostate cancer (NEPCa) activity scores. We analyzed prostate cancer cell lines for NFIB expression via Western blot analysis and used nuclear and cytoplasmic fractionation to assess where NFIB is localized. We performed co-immunoprecipitation studies to determine if NFIB and AR interact. RESULTS: NFIB increased in the nucleus and cytoplasm of prostate cancer samples versus matched normal controls, independent of Gleason score. Similarly, cytoplasmic AR and synaptophysin increased in primary prostate cancer. We observed strong NFIB staining in primary small cell prostate cancer. The ratio of cytoplasmic-to-nuclear NFIB staining was predictive of earlier biochemical recurrence in prostate cancer, once adjusted for tumor margin status. Cytoplasmic AR was an independent predictor of biochemical recurrence. There was no statistically significant difference between NFIB and synaptophysin expression in primary and castration-resistant prostate cancer, but cytoplasmic AR expression was increased in castration-resistant samples. In primary prostate cancer, nuclear NFIB expression correlated with cytoplasmic NFIB and nuclear AR, while cytoplasmic NFIB correlated with synaptophysin, and nuclear and cytoplasmic AR. In castration-resistant prostate cancer samples, NFIB expression correlated positively with an AR activity score, and negatively with the NEPCa score. In prostate cancer cell lines, NFIB exists in several isoforms. We observed NFIB predominantly in the nuclear fraction of prostate cancer cells with increased cytoplasmic expression seen in castration-resistant cell lines. We observed an interaction between AR and NFIB through co-immunoprecipitation experiments. CONCLUSION: We have described the expression pattern of NFIB in primary and castration-resistant prostate cancer and its positive correlation with AR. We have also demonstrated AR interacts with NFIB.


Asunto(s)
Factores de Transcripción NFI/biosíntesis , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/biosíntesis , Línea Celular Tumoral , Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Factores de Transcripción NFI/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/genética , Análisis de Matrices Tisulares , Transcriptoma
2.
Oncotarget ; 7(38): 61955-61969, 2016 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-27542219

RESUMEN

Numerous studies indicate that androgen receptor splice variants (ARVs) play a critical role in the development of castration-resistant prostate cancer (CRPC), including the resistance to the new generation of inhibitors of androgen receptor (AR) action. Previously, we demonstrated that activation of NF-κB signaling increases ARVs expression in prostate cancer (PC) cells, thereby promoting progression to CRPC. However, it is unclear how NF-κB signaling is activated in CRPC. In this study, we report that long-term treatment with anti-androgens increases a neuroendocrine (NE) hormone - gastrin-releasing peptide (GRP) and its receptor (GRP-R) expression in PC cells. In addition, activation of GRP/GRP-R signaling increases ARVs expression through activating NF-κB signaling. This results in an androgen-dependent tumor progressing to a castrate resistant tumor. The knock-down of AR-V7 restores sensitivity to antiandrogens of PC cells over-expressing the GRP/GRP-R signaling pathway. These findings strongly indicate that the axis of Androgen-Deprivation Therapy (ADT) induces GRP/GRP-R activity, activation NF-κB and increased levels of AR-V7 expression resulting in progression to CRPC. Both prostate adenocarcinoma and small cell NE prostate cancer express GRP-R. Since the GRP-R is clinically targetable by analogue-based approach, this provides a novel therapeutic approach to treat advanced CRPC.


Asunto(s)
Péptido Liberador de Gastrina/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores de Bombesina/metabolismo , Adenocarcinoma/metabolismo , Antagonistas de Andrógenos/uso terapéutico , Andrógenos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Progresión de la Enfermedad , Variación Genética , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/cirugía , Empalme del ARN , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal , Transcripción Genética
3.
Mol Cancer Ther ; 7(11): 3519-26, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19001435

RESUMEN

Medulloblastomas are the most frequent malignant brain tumors in children. Sorafenib (Nexavar, BAY43-9006), a multikinase inhibitor, blocks cell proliferation and induces apoptosis in a variety of tumor cells. Sorafenib inhibited proliferation and induced apoptosis in two established cell lines (Daoy and D283) and a primary culture (VC312) of human medulloblastomas. In addition, sorafenib inhibited phosphorylation of signal transducer and activator of transcription 3 (STAT3) in both cell lines and primary tumor cells. The inhibition of phosphorylated STAT3 (Tyr(705)) occurs in a dose- and time-dependent manner. In contrast, AKT (protein kinase B) was only decreased in D283 and VC312 medulloblastoma cells and mitogen-activated protein kinases (extracellular signal-regulated kinase 1/2) were not inhibited by sorafenib in these cells. Both D-type cyclins (D1, D2, and D3) and E-type cyclin were down-regulated by sorafenib. Also, expression of the antiapoptotic protein Mcl-1, a member of the Bcl-2 family, was decreased and correlated with apoptosis induced by sorafenib. Finally, sorafenib suppressed the growth of human medulloblastoma cells in a mouse xenograft model. Together, our data show that sorafenib blocks STAT3 signaling as well as expression of cell cycle and apoptosis regulatory proteins, associated with inhibition of cell proliferation and induction of apoptosis in medulloblastomas. These findings provide a rationale for treatment of pediatric medulloblastomas with sorafenib.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Bencenosulfonatos/farmacología , Neoplasias Cerebelosas/metabolismo , Meduloblastoma/metabolismo , Piridinas/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal , Animales , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Ciclina D , Ciclinas/antagonistas & inhibidores , Ciclinas/metabolismo , Regulación hacia Abajo , Humanos , Meduloblastoma/tratamiento farmacológico , Ratones , Ratones Desnudos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor de Transcripción STAT3/metabolismo , Sorafenib , Transfección
4.
Differentiation ; 76(6): 682-98, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18462434

RESUMEN

Androgen receptor (AR) within prostatic mesenchymal cells, with the absence of AR in the epithelium, is still sufficient to induce prostate development. AR in the luminal epithelium is required to express the secretory markers associated with differentiation. Nkx3.1 is expressed in the epithelium in early prostatic embryonic development and expression is maintained in the adult. Induction of the mouse prostate gland by the embryonic mesenchymal cells results in the organization of a sparse basal layer below the luminal epithelium with rare neuroendocrine cells that are interdispersed within this basal layer. The human prostate shows similar glandular organization; however, the basal layer is continuous. The strong inductive nature of embryonic prostatic and bladder mesenchymal cells is demonstrated in grafts where embryonic stem (ES) cells are induced to differentiate and organize as a prostate and bladder, respectively. Further, the ES cells can be driven by the correct embryonic mesenchymal cells to form epithelium that differentiates into secretory prostate glands and differentiated bladders that produce uroplakin. This requires the ES cells to mature into endoderm that gives rise to differentiated epithelium. This process is control by transcription factors in both the inductive mesenchymal cells (AR) and the responding epithelium (FoxA1 and Nkx3.1) that allows for organ development and differentiation. In this review, we explore a molecular mechanism where the pattern of transcription factor expression controls cell determination, where the cell is assigned a developmental fate and subsequently cell differentiation, and where the assigned cell now emerges with it's own unique character.


Asunto(s)
Células Epiteliales , Modelos Biológicos , Próstata/citología , Animales , Diferenciación Celular , Humanos , Masculino , Próstata/efectos de los fármacos , Factores de Transcripción/farmacología , Vejiga Urinaria/citología , Vejiga Urinaria/efectos de los fármacos
5.
Prostate ; 66(10): 1013-28, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16001449

RESUMEN

The molecular mechanism(s) for prostate cancer progression to androgen independence are poorly understood. We have recently shown that Foxa1 and Foxa2 proteins are differentially expressed in epithelial cells during murine prostate development, growth, and adult function. Currently, the role of Foxa proteins in prostate cancer development and progression is unknown. Foxa protein expression was investigated in the LPB-Tag LADY mouse prostate cancer models, in human prostate cancer specimens, and various prostate cancer cell lines using Western blot and immunostaining analysis. In vitro transient transfection, studies were performed to investigate Foxa/prostate-specific gene regulation. Foxa1 was strongly expressed in areas of prostatic intraepithelial neoplasia (PIN) in both the androgen dependent 12T-7f and in the metastatic, androgen independent 12T-10 LADY models. Prominent Foxa1 and Foxa2 expression was observed in 12T-10 invasive undifferentiated neuroendocrine carcinomas, in the hormone independent and metastasizing 12T-10 derived, NE-10 allograft tumors, and in all metastatic lesions isolated from 12T-10 mice. Foxa1 protein expression was always observed in human prostate carcinomas, regardless of Gleason grade score, while Foxa2 was only detected in neuroendocrine small cell carcinomas and in some high Gleason score adenocarcinomas. Foxa proteins were also differentially expressed in three prostate cancer cell lines. Importantly, in vitro functional assays demonstrated that Foxa2 could activate androgen-dependent prostate-specific genes in an androgen receptor and ligand-independent manner. These results suggest that Foxa proteins are important in prostate carcinogenesis. In particular, Foxa2 may be involved in progression of prostate cancer to androgen independence. As such, Foxa proteins may represent novel targets for therapeutic intervention.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , Factor Nuclear 3-alfa del Hepatocito/fisiología , Factor Nuclear 3-beta del Hepatocito/fisiología , Neoplasias de la Próstata/fisiopatología , Adenocarcinoma/química , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma/fisiopatología , Andrógenos/fisiología , Animales , Carcinoma Neuroendocrino/química , Carcinoma Neuroendocrino/genética , Carcinoma Neuroendocrino/patología , Carcinoma Neuroendocrino/fisiopatología , Carcinoma de Células Pequeñas/química , Carcinoma de Células Pequeñas/genética , Carcinoma de Células Pequeñas/patología , Carcinoma de Células Pequeñas/fisiopatología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Epitelio/química , Epitelio/patología , Epitelio/fisiopatología , Técnica del Anticuerpo Fluorescente , Factor Nuclear 3-alfa del Hepatocito/análisis , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/análisis , Factor Nuclear 3-beta del Hepatocito/genética , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Neoplasia Intraepitelial Prostática/química , Neoplasia Intraepitelial Prostática/genética , Neoplasia Intraepitelial Prostática/patología , Neoplasia Intraepitelial Prostática/fisiopatología , Neoplasias de la Próstata/química , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Transfección , Regulación hacia Arriba
6.
Cancer Res ; 65(20): 9185-9, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16230377

RESUMEN

Src family kinases (SFK) are currently being investigated as targets for treatment strategies in various cancers. The novel SFK/Abl inhibitor, dasatinib (BMS-354825), is a promising therapeutic agent with oral bioavailability. Dasatinib has been shown to inhibit growth of Bcr-Abl-dependent chronic myeloid leukemia xenografts in nude mice. Dasatinib also has been shown to have activity against cultured human prostate and breast cancer cells. However, the molecular mechanism by which dasatinib acts on epithelial tumor cells remains unknown. In this study, we show that dasatinib blocks the kinase activities of the SFKs, Lyn, and Src, in human prostate cancer cells at low nanomolar concentrations. Moreover, focal adhesion kinase and Crk-associated substrate (p130(CAS)) signaling downstream of SFKs are also inhibited at similar concentrations of dasatinib. Consistent with inhibition of these signaling pathways, dasatinib suppresses cell adhesion, migration, and invasion of prostate cancer cells at low nanomolar concentrations. Therefore, dasatinib has potential as a therapeutic agent for metastatic prostate cancers harboring activated SFK and focal adhesion kinase signaling.


Asunto(s)
Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Proteína Sustrato Asociada a CrK/metabolismo , Dasatinib , Relación Dosis-Respuesta a Droga , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Masculino , Invasividad Neoplásica , Neoplasias de la Próstata/patología , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/metabolismo
7.
Development ; 132(15): 3431-43, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15987773

RESUMEN

We have previously shown that a forkhead transcription factor Foxa1 interacts with androgen signaling and controls prostate differentiated response. Here, we show the mouse Foxa1 expression marks the entire embryonic urogenital sinus epithelium (UGE), contrasting with Shh and Foxa2, which are restricted to the basally located cells during prostate budding. The Foxa1-deficient mouse prostate shows a severely altered ductal pattern that resembles primitive epithelial cords surrounded by thick stromal layers. Characterization of these mutant cells indicates a population of basal-like cells similar to those found in the embryonic UGE, whereas no differentiated or mature luminal epithelial cells are found in Foxa1-deficient epithelium. These phenotypic changes are accompanied with molecular aberrations, including focal epithelial activation of Shh and elevated Foxa2 and Notch1 in the null epithelium. Perturbed epithelial-stromal interactions induced by Foxa1-deficient epithelium is evident, as demonstrated by the expansion of surrounding smooth muscle and elevated levels of stromal factors (Bmp4, Fgf7, Fgf10 and Gli). The prostatic homeobox protein Nkx3.1, a known proliferation inhibitor, was downregulated in Foxa1-deficient epithelial cells, while several prostate-specific androgen-regulated markers, including a novel Foxa1 target, are absent in the null prostate. These data indicate that Foxa1 plays a pivotal role in controlling prostate morphogenesis and cell differentiation.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Células Epiteliales/fisiología , Proteínas Nucleares/fisiología , Próstata/embriología , Factores de Transcripción/fisiología , Animales , Diferenciación Celular/fisiología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Células Epiteliales/citología , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Factor Nuclear 3-alfa del Hepatocito , Hibridación in Situ , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Morfogénesis , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Próstata/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
8.
Genomics ; 85(3): 401-12, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15718107

RESUMEN

Microarray technology can be used to assess simultaneously global changes in expression of mRNA or genomic DNA copy number among thousands of genes in different biological states. In many cases, it is desirable to determine if altered patterns of gene expression correlate with chromosomal abnormalities or assess expression of genes that are contiguous in the genome. We describe a method, differential gene locus mapping (DIGMAP), which aligns the known chromosomal location of a gene to its expression value deduced by microarray analysis. The method partitions microarray data into subsets by chromosomal location for each gene interrogated by an array. Microarray data in an individual subset can then be clustered by physical location of genes at a subchromosomal level based upon ordered alignment in genome sequence. A graphical display is generated by representing each genomic locus with a colored cell that quantitatively reflects its differential expression value. The clustered patterns can be viewed and compared based on their expression signatures as defined by differential values between control and experimental samples. In this study, DIGMAP was tested using previously published studies of breast cancer analyzed by comparative genomic hybridization (CGH) and prostate cancer gene expression profiles assessed by cDNA microarray experiments. Analysis of the breast cancer CGH data demonstrated the ability of DIGMAP to deduce gene amplifications and deletions. Application of the DIGMAP method to the prostate data revealed several carcinoma-related loci, including one at 16q13 with marked differential expression encompassing 19 known genes including 9 encoding metallothionein proteins. We conclude that DIGMAP is a powerful computational tool enabling the coupled analysis of microarray data with genome location.


Asunto(s)
Mapeo Cromosómico , Expresión Génica , ADN/genética , Humanos , Masculino , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias de la Próstata/genética , ARN Mensajero/genética
9.
Ann N Y Acad Sci ; 1061: 77-93, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16467259

RESUMEN

Previous studies from our group have shown that Foxa1 is expressed in the prostate and interacts with the androgen receptor (AR) to regulate prostate-specific genes such as prostate-specific antigen (PSA) and probasin (PB). We report here that Foxa2 but not Foxa1 is expressed in the epididymis. Further, Foxa2 interacts with the AR to regulate the mouse epididymal retinoic acid binding protein (mE-RABP) gene, an epididymis-specific gene. Binding of Foxa2 to the mE-RABP promoter was confirmed by gel-shift and chromatin immunoprecipitation (ChIP) assays. Overexpression of Foxa2 suppresses androgen activation of the mE-RABP promoter while overexpression of Foxa2 with prostate-specific promoters activates gene expression in an androgen-independent manner. GST pull-down assays determined that both Foxa1 and Foxa2 physically interact with the DNA binding domain of the AR. The interaction between Foxa proteins and AR was further confirmed by gel-shift assays where Foxa protein was recruited to AR binding oligomers even when Foxa binding sites were not present, and AR was recruited to Foxa binding oligomers even in the absence of an AR binding site. Given that Foxa1 and Foxa2 proteins are expressed differentially in the prostate and epididymis, these data suggest that the Foxa proteins have distinct effects on AR-regulated genes in different male reproductive accessory organs.


Asunto(s)
Epidídimo/metabolismo , Regulación de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Próstata/metabolismo , Receptores Androgénicos/metabolismo , Andrógenos/farmacología , Animales , Secuencia de Bases , Sitios de Unión/genética , Inmunoprecipitación de Cromatina , Factor Nuclear 3-beta del Hepatocito/genética , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Neoplasias de la Próstata/metabolismo , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Proteínas Plasmáticas de Unión al Retinol , Transfección
10.
Prostate ; 62(4): 339-52, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15389796

RESUMEN

BACKGROUND: The Foxa family (a1, a2, and a3) of proteins are transcription factors that are central to endodermal development. Recently, Foxa1 has been shown to regulate the transcription of several murine and human prostate specific genes involved in differentiated function by interacting with DNA promoter sequences and androgen receptors. Currently, the developmental expression pattern of Foxa proteins in the murine prostate is unknown. METHODS: Male CD-1 mice (embryonic, prepubertal, pubertal, and adult) were used for immunohistochemical analysis of Foxa1, a2, and a3. Immunofluorescence was also performed for androgen receptor and cytokeratin 14 expression. Prostate tissue from pre-pubertal, pubertal, and adult mice were analyzed by Western blot and RT-PCR analysis for Foxa1, a2, and a3 expression. RESULTS: Strong Foxa1 immunoreactivity was observed in epithelial cells throughout prostate development, growth, and adult differentiation. Prominent Foxa2 protein expression was only observed in the early stages of prostate development and was exclusively localized to epithelial cells of the forming buds. RT-PCR analysis identified low Foxa2 mRNA expression levels in the ventral and dorsolateral lobes of the adult prostate, with Foxa2 epithelial cell expression being localized to periurethral regions of the murine adult prostatic complex. Foxa3 expression was not observed in the murine prostate. CONCLUSIONS: Foxa proteins represent epithelial cell markers in the murine prostate gland. The early expression of Foxa1 and a2 proteins in prostate formation suggests that these proteins play an important role in normal prostate development, in addition to differentiated secretory function.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Proteínas Nucleares/biosíntesis , Próstata/crecimiento & desarrollo , Factores de Transcripción/biosíntesis , Animales , Células Epiteliales , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito , Factor Nuclear 3-beta del Hepatocito , Factor Nuclear 3-gamma del Hepatocito , Masculino , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Cancer Cell ; 6(2): 185-95, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15324701

RESUMEN

The majority of cancer-related deaths are associated with metastasis; however, little is known about the mechanisms of this process. Hepsin is a cell surface serine protease that is markedly upregulated in human prostate cancer; however, the functional significance of this upregulation is unknown. We report here that hepsin overexpression in prostate epithelium in vivo causes disorganization of the basement membrane. Overexpression of hepsin in a mouse model of nonmetastasizing prostate cancer has no impact on cell proliferation, but causes disorganization of the basement membrane and promotes primary prostate cancer progression and metastasis to liver, lung, and bone. We provide in vivo evidence that upregulation of a cell surface serine protease in a primary tumor promotes cancer progression and metastasis.


Asunto(s)
Metástasis de la Neoplasia , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Serina Endopeptidasas/metabolismo , Animales , Apoptosis , Membrana Basal/metabolismo , Membrana Basal/patología , Membrana Basal/ultraestructura , Diferenciación Celular , División Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Próstata/anatomía & histología , Próstata/metabolismo , Próstata/patología , Serina Endopeptidasas/genética
12.
Mol Endocrinol ; 17(8): 1484-507, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12750453

RESUMEN

Androgens and mesenchymal factors are essential extracellular signals for the development as well as the functional activity of the prostate epithelium. Little is known of the intraepithelial determinants that are involved in prostatic differentiation. Here we found that hepatocyte nuclear factor-3 alpha (HNF-3 alpha), an endoderm developmental factor, is essential for androgen receptor (AR)-mediated prostatic gene activation. Two HNF-3 cis-regulatory elements were identified in the rat probasin (PB) gene promoter, each immediately adjacent to an androgen response element. Remarkably, similar organization of HNF-3 and AR binding sites was observed in the prostate-specific antigen (PSA) gene core enhancer, suggesting a common functional mechanism. Mutations that disrupt these HNF-3 motifs significantly abolished the maximal androgen induction of PB and PSA activities. Overexpressing a mutant HNF-3 alpha deleted in the C-terminal region inhibited the androgen-induced promoter activity in LNCaP cells where endogenous HNF-3 alpha is expressed. Chromatin immunoprecipitation revealed in vivo that the occupancy of HNF-3 alpha on PSA enhancer can occur in an androgen-depleted condition, and before the recruitment of ligand-bound AR. A physical interaction of HNF-3 alpha and AR was detected through immunoprecipitation and confirmed by glutathione-S-transferase pull-down. This interaction is directly mediated through the DNA-binding domain/hinge region of AR and the forkhead domain of HNF-3 alpha. In addition, strong HNF-3 alpha expression, but not HNF-3 beta or HNF-3 gamma, is detected in both human and mouse prostatic epithelial cells where markers (PSA and PB) of differentiation are expressed. Taken together, these data support a model in which regulatory cues from the cell lineage and the extracellular environment coordinately establish the prostatic differentiated response.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Proteínas Nucleares/fisiología , Próstata/fisiología , Receptores Androgénicos/fisiología , Factores de Transcripción/fisiología , Fosfatasa Ácida , Proteína de Unión a Andrógenos/genética , Animales , Secuencia de Bases , Sitios de Unión , Elementos de Facilitación Genéticos/genética , Células Epiteliales/metabolismo , Factor Nuclear 3-alfa del Hepatocito , Humanos , Masculino , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Mutación , Regiones Promotoras Genéticas , Próstata/citología , Antígeno Prostático Específico/genética , Neoplasias de la Próstata/genética , Estructura Terciaria de Proteína , Proteínas Tirosina Fosfatasas/genética , Ratas , Secuencias Reguladoras de Ácidos Nucleicos , Activación Transcripcional , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...