Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Prostate ; 75(9): 976-87, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25808853

RESUMEN

BACKGROUND: ELL-associated factor 2 (EAF2) is an androgen-regulated tumor suppressor in the prostate. However, the mechanisms underlying tumor suppressive function of EAF2 are still largely unknown. Identification of factors capable of modulating EAF2 function will help elucidate the mechanisms underlying EAF2 tumor suppressive function. METHODS: Using eaf-1(the ortholog of EAF2) mutant C. elegans model, RNAi screen was used to identify factors on the basis of their knockdown to synergistically enhance the reduced fertility phenotype of the eaf-1 mutant C. elegans. In human cells, the interaction of EAF2 with FOXA1 and the effect of EAF2 on the FOXA1 protein levels were determined by co-immunoprecipitation and protein stability assay. The effect of EAF2 and/or FOXA1 knockdown on the expression of AR-target genes was determined by real-time RT-PCR and luciferase reporter assays. The effect of EAF2 and/or FOXA1 knockdown on LNCaP human prostate cancer cell proliferation and migration was tested using BrdU assay and transwell migration assay. RESULTS: RNAi screen identified pha-4, the C. elegans ortholog of mammalian FOXA1, on the basis of its knockdown to synergistically enhance the reduced fertility phenotype of the eaf-1 mutant C. elegans causing sterility. EAF2 co-immunoprecipitated with FOXA1. EAF2 knockdown enhanced endogenous FOXA1 protein level, whereas transfected GFP-EAF2 down-regulated the FOXA1 protein. Also, EAF2 knockdown enhanced the expression of AR-target genes, cell proliferation, and migration in LNCaP cells. However, FOXA1 knockdown inhibited the effect of EAF2 knockdown on AR-target gene expression, cell proliferation, and migration in LNCaP cells, suggesting that FOXA1 can modulate EAF2 regulation of AR transcriptional activation, cell proliferation, and migration. CONCLUSIONS: These findings suggest that regulation of the AR signaling pathway, cell proliferation, and migration through FOXA1 represents an important mechanism of EAF2 suppression of prostate carcinogenesis.


Asunto(s)
Factor Nuclear 3-alfa del Hepatocito/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Factores de Transcripción/metabolismo , Animales , Western Blotting , Caenorhabditis elegans , Línea Celular Tumoral , Proliferación Celular/fisiología , Regulación hacia Abajo , Factor Nuclear 3-alfa del Hepatocito/genética , Humanos , Inmunoprecipitación , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Interferencia de ARN/fisiología , Receptores Androgénicos/genética , Factores de Transcripción/genética , Activación Transcripcional , Transfección/métodos
2.
Prostate ; 74(2): 113-20, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24272884

RESUMEN

BACKGROUND: The tumor suppressor ELL associated factor 2 (EAF2/U19) has been reported to induce apoptosis of LNCaP cells and suppress AT6.1 xenograft prostate tumor growth. EAF2/U19 expression level is down-regulated in advanced human prostate cancer. EAF2/U19 is also a putative transcription factor with a transactivation domain and capability of sequence-specific DNA binding. Identification of binding partners and regulators of EAF2/U19 is essential to understand its function in regulating apoptosis/survival of prostate cancer cells. METHODS: Through a yeast two-hybrid screening system, we identified Pirin as a binding partner of EAF2. We further determined the interaction between epitope-tagged EAF2/U19 and Pirin by co-immunoprecipitation in mammalian cells. The effect of Pirin on EAF2/U19 inhibition of LNCaP growth was assayed by colony formation. RESULTS: Pirin co-immunoprecipitated with EAF2/U19 and the overexpressed Pirin decreased the expression level of EAF2/U19 protein in prostate cancer cell lines LNCaP and PC3. Furthermore, overexpression of EAF2/U19 suppressed LNCaP colony formation, and co-expression of Pirin significantly blocked the growth inhibition induced by EAF2/U19 overexpression. CONCLUSION: Pirin is a newly identified binding partner of EAF2/U19 capable of down-regulating EAF2/U19 protein and alleviating its inhibition of prostate cancer cell survival/proliferation. Pirin may play an important role involved in EAF2/U19 function as an androgen-responsive gene and tumor repressor.


Asunto(s)
Adenocarcinoma/patología , Proteínas Portadoras/fisiología , Proliferación Celular , Regulación hacia Abajo/fisiología , Proteínas Nucleares/fisiología , Neoplasias de la Próstata/patología , Factores de Transcripción/fisiología , Adenocarcinoma/fisiopatología , Apoptosis/fisiología , Carcinogénesis/patología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Dioxigenasas , Humanos , Inmunoprecipitación , Masculino , Neoplasias de la Próstata/fisiopatología , Unión Proteica/fisiología
3.
Biochem Biophys Res Commun ; 435(4): 526-32, 2013 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-23643812

RESUMEN

Cell adhesion molecules (CADMs) comprise a protein family whose functions include maintenance of cell polarity and tumor suppression. In this report, we show that the CADM2 gene is repressed in human clear renal cell carcinoma by DNA promoter hypermethylation and/or loss of heterozygosity. Moreover, the loss of CADM2 expression is associated with a higher tumor pathology stage (p<0.05). The re-expression of CADM2 in the renal cancer cell line 786-O significantly suppressed tumor cell growth in vitro and in mouse xenografts by a G1 phase cell cycle arrest and the induction of apoptosis. Lentivirus-mediated CADM2 expression also significantly suppressed cancer cell anchorage-independent growth and invasion. Furthermore, the inhibition of endogenous CADM2 expression using siRNAs induced a tumorigenic phenotype in polarized non-tumorigenic MDCK cells. Thus, we conclude that CADM2 functions as a novel tumor suppressor and may serve as a potential therapeutic target for human renal cell carcinoma.


Asunto(s)
Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Moléculas de Adhesión Celular/metabolismo , Metilación de ADN/genética , Neoplasias Renales/genética , Neoplasias Renales/patología , Animales , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ratones , Ratones Endogámicos BALB C
4.
Prostate ; 72(10): 1117-23, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22161776

RESUMEN

BACKGROUND: Advanced prostate cancer is currently treated with androgen deprivation therapy (ADT). ADT initially results in tumor regression; however, all patients eventually relapse with castration-resistant prostate cancer. New approaches to delay the progression of prostate cancer to castration resistance are in desperate need. This study addresses whether targeting Heat shock protein 90 (HSP90) regulation of androgen receptor (AR) can inhibit prostate cancer progression to castration resistance. METHODS: The HSP90 inhibitor 17-AAG was injected intraperitoneally into nude mice bearing LuCaP35 xenograft tumors to determine the effect of HSP90 inhibition on prostate cancer progression to castration resistance and host survival. RESULTS: Administration of 17-AAG maintained androgen-sensitivity, delayed the progression of LuCaP35 xenograft tumors to castration resistance, and prolonged the survival of host. In addition, 17-AAG prevented nuclear localization of endogenous AR in LuCaP35 xenograft tumors in castrated nude mice. CONCLUSIONS: Targeting Hsp90 or the mechanism by which HSP90 regulates androgen-independent AR nuclear localization and activation may lead to new approaches to prevent and/or treat castration-resistant prostate cancer.


Asunto(s)
Benzoquinonas/farmacología , Benzoquinonas/uso terapéutico , Progresión de la Enfermedad , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Lactamas Macrocíclicas/farmacología , Lactamas Macrocíclicas/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Proteínas HSP90 de Choque Térmico/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Orquiectomía , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Distribución Aleatoria
5.
Angiogenesis ; 14(3): 331-43, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21638067

RESUMEN

Von Hippel-Lindau (VHL) disease results from the inactivation of the VHL gene and is characterized by highly vascular tumors. A consequence of VHL loss is the stabilization of hypoxia-inducible factor (HIF) alpha subunits and increased expression of HIF target genes, which include pro-angiogenic growth factors such as vascular endothelial growth factor (VEGF). In mice, homozygous deletion of VHL is embryonic lethal due to vascular abnormalities in the placenta; and, VHL(+/-) mice develop proliferative vascular lesions in several major organs, most prominently the liver. Loss of ELL-associated factor (EAF2) in murine models has also been shown to induce increased vascular density in the liver as well as the prostate. Previously, EAF2 was determined to be a binding partner of VHL and loss of EAF2 induced a reduction in pVHL levels and an increase in hypoxia induced factor 1α (HIF1α) levels in vitro. Here we characterized the cooperative effects of VHL- and EAF2-deficiency on angiogenesis in the liver and prostate of male mice. VHL deficiency consistently increased the incidence of hepatic vascular lesions across three mouse strains. These vascular lesions where characterized by an increase in microvessel density, and staining intensity of VHL target proteins HIF1α and VEGF. EAF2(-/-)VHL(+/-) mice had increased incidence of proliferative hepatic vascular lesions (4/4) compared to VHL(+/-) (10/18) and EAF2(-/-) (0/5) mice. Prostates of EAF2(-/-)VHL(+/-) mice also displayed an increase in microvessel density, as well as stromal inflammation and prostatic intraepithelial neoplasia. These results suggest that cooperation of VHL and EAF2 may be critical for angiogenic regulation of the liver and prostate, and concurrent loss of these two tumor suppressors may result in a pro-angiogenic phenotype.


Asunto(s)
Heterocigoto , Hígado/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Nucleares/metabolismo , Próstata/metabolismo , Transactivadores/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Proteínas Nucleares/genética , Próstata/patología , Neoplasia Intraepitelial Prostática/genética , Neoplasia Intraepitelial Prostática/metabolismo , Neoplasia Intraepitelial Prostática/patología , Transactivadores/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/patología
6.
Prostate ; 70(7): 797-805, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20166137

RESUMEN

BACKGROUND: Eleven-nineteen lysine-rich leukemia (ELL) plays an important role in tumorigenesis and animal development. HIF-1 is a transcriptional factor that functions as a master regulator of O(2) homeostasis. Our previous studies showed that a binding partner of ELL, U19/Eaf2, can modulate HIF-1alpha activity and hypoxia response, suggesting that ELL may also influence HIF-1alpha pathway and hypoxia response. METHODS: Co-localization and co-immunoprecipitation were performed to test the interaction between ELL and HIF-1alpha. PC3 cells with stable ELL knockdown and PC3 cells with stable ELL overexpression, along with their controls, were established using lentiviral expression system. Western blot and real-time PCR were performed to test the effect of ELL on HIF-1alpha protein and its down-stream gene transcription. To elucidate potential effect of hypoxia on ELL, cell growth and colony formation assays were performed using PC3 subline with stable ELL overexpression. RESULTS: ELL is associated with HIF-1alpha in transfected cells. In PC3 prostate cancer cells, ELL inhibited HIF-1alpha protein level and down-stream gene expression. As expected, ELL inhibited cell growth and colony formation under normoxia. Interestingly, the inhibition was alleviated under hypoxia. CONCLUSIONS: Our findings suggest that ELL and HIF-1alpha are binding partners and can modulate the functions of each other in hypoxia.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Próstata/metabolismo , Factores de Elongación Transcripcional/metabolismo , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Humanos , Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inmunoprecipitación , Masculino , Próstata/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Elongación Transcripcional/genética , Transfección
7.
Am J Clin Exp Urol ; 8(1): 18-27, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32211450

RESUMEN

Defining the cell of origin for prostatic carcinogenesis is fundamentally important for understanding the mechanisms leading to prostate cancer. Lineage tracing studies have demonstrated that luminal epithelial cells are capable of self-replication in multiple organs, including the adult murine prostate, and cell of prostate cancer origin studies have shown that while both the luminal and basal murine prostate epithelial cells are capable of neoplastic transformation, luminal cells are more efficient as the origin of prostate cancer. ELL-associated factor 2 (EAF2) is an androgen responsive tumor suppressive protein expressed by prostate luminal epithelial cells that is frequently down-regulated in primary prostate tumors. EAF2 knockdown induces prostate cancer cell proliferation and invasion in vitro and mice with Eaf2 deficiency develop epithelial hyperplasia and murine prostatic intraepithelial neoplasia (mPIN) lesions. Here, we utilized an Eaf2 knockout, PSA-CreERT2 transgenic model crossed with a fluorescent reporter line to show that Eaf2 deficiency induces mPIN lesions derived from the luminal cell lineage. These results suggest that PIN lesions in the Eaf2 knockout mouse were derived from prostate luminal epithelial cells, further suggesting that the prostatic luminal epithelial cell is the major origin of prostate carcinogenesis.

8.
Oncotarget ; 10(67): 7181-7182, 2019 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-31903175

RESUMEN

[This corrects the article DOI: 10.18632/oncotarget.8588.].

9.
Neoplasia ; 21(8): 752-764, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31229879

RESUMEN

ELL-associated factor 1 is a transcription elongation factor that shares significant homology and functional similarity to the androgen-responsive prostate tumor suppressor ELL-associated factor 2. EAF2 is frequently down-regulated in advanced prostate cancer and Eaf2 deletion in the mouse induced the development of murine prostatic intraepithelial neoplasia. Here we show that similar to EAF2, EAF1 is frequently down-regulated in advanced prostate cancer. Co-downregulation of EAF1 and EAF2 occurred in 40% of clinical specimens with Gleason score >7. We developed and characterized a murine model of prostate-epithelial specific deletion of Eaf1 in the prostate and crossed it with our previously generated mouse with conventional deletion of Eaf2. The prostates of Eaf1 deletion mice displayed murine prostatic intraepithelial neoplasia lesions with increased proliferation and inflammation. Combined deletion of Eaf1 and Eaf2 in the murine model induced an increased incidence in mPIN lesions characterized by increased proliferation and CD3+ T cells and CD19+ B cells infiltration compared to individual deletion of either Eaf1 or Eaf2 in the murine prostate. These results suggest that EAF1 may play a tumor suppressive role in the prostate. Cooperation between EAF1 and EAF2 may be important for prostate maintaining prostate epithelial homeostasis, and concurrent loss of these two tumor suppressors may promote prostate tumorigenesis and progression.


Asunto(s)
Eliminación de Gen , Predisposición Genética a la Enfermedad , Neoplasia Intraepitelial Prostática/genética , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Factores de Transcripción/genética , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Marcación de Gen , Sitios Genéticos , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Clasificación del Tumor , Neoplasia Intraepitelial Prostática/metabolismo , Neoplasias de la Próstata/metabolismo , Factores de Transcripción/química
10.
Cancer Lett ; 415: 198-207, 2018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29179998

RESUMEN

ELL2 is an androgen-responsive gene that is expressed by prostate epithelial cells and is frequently down-regulated in prostate cancer. Deletion of Ell2 in the murine prostate induced murine prostatic intraepithelial neoplasia and ELL2 knockdown enhanced proliferation and migration in C4-2 prostate cancer cells. Here, knockdown of ELL2 sensitized prostate cancer cells to DNA damage and overexpression of ELL2 protected prostate cancer cells from DNA damage. Knockdown of ELL2 impaired non-homologous end joining repair but not homologous recombination repair. Transfected ELL2 co-immunoprecipitated with both Ku70 and Ku80 proteins. ELL2 could bind to and co-accumulate with Ku70/Ku80 proteins at sites of DNA damage. Knockdown of ELL2 dramatically inhibited Ku70 and Ku80 recruitment and retention at DNA double-strand break sites in prostate cancer cells. The impaired recruitment of Ku70 and Ku80 proteins to DNA damage sites upon ELL2 knockdown was rescued by re-expression of an ELL2 transgene insensitive to siELL2. This study suggests that ELL2 is required for efficient NHEJ repair via Ku70/Ku80 in prostate cancer cells.


Asunto(s)
Reparación del ADN por Unión de Extremidades/genética , Reparación del ADN , Interferencia de ARN , Factores de Elongación Transcripcional/genética , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Doxorrubicina/farmacología , Rayos gamma , Células HEK293 , Células HeLa , Humanos , Autoantígeno Ku/metabolismo , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Factores de Elongación Transcripcional/metabolismo
11.
Cancer Lett ; 253(2): 265-72, 2007 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-17395368

RESUMEN

BACKGROUND: U19/EAF2 is a potential tumor suppressor exhibiting frequent down-regulation and allelic loss in advanced human prostate cancer specimens. U19/EAF2 has also been identified as ELL-associated factor 2 (EAF2) based on its binding to ELL, a fusion partner of MLL in acute myeloid leukemia. U19/EAF2 is a putative transcription factor with a transactivation domain and capability of sequence-specific DNA binding. METHODS: Yeast-two-hybrid-screening was used to identify U19/EAF2-binding partners. Co-immunoprecipitation and mammalian 1-hybrid assay were used to characterize a U19/EAF2-binding partner. RESULTS: FB1, an E2A fusion partner in childhood leukemia, was identified as a binding-partner of U19/EAF2. FB1 also binds to EAF1, the only homologue of U19/EAF2. FB1 also interacts and co-localizes with ELL in the nucleus. Interestingly, FB1 inhibited the transcriptional activity of U19/EAF2 but not EAF1. CONCLUSIONS: FB1 is an important binding partner and a functional regulator of U19/EAF2, EAF1, and/or ELL.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/análisis , Células COS , Chlorocebus aethiops , Humanos , Inmunoprecipitación , Factores de Transcripción/análisis , Transcripción Genética , Factores de Elongación Transcripcional/análisis , Factores de Elongación Transcripcional/metabolismo , Técnicas del Sistema de Dos Híbridos
12.
Mol Cancer Ther ; 16(10): 2120-2129, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28655783

RESUMEN

The androgen receptor (AR) is a ligand-dependent transcription factor that controls the expression of androgen-responsive genes. A key step in androgen action, which is amplified in castration-resistant prostate cancer (CRPC), is AR nuclear translocation. Small molecules capable of inhibiting AR nuclear localization could be developed as novel therapeutics for CRPC. We developed a high-throughput screen and identified two structurally-related pyrroloimidazoles that could block AR nuclear localization in CRPC cells. We show that these two small molecules, 3-(4-ethoxyphenyl)-6,7-dihydro-5H-pyrrolo[1,2-a]imidazole (EPPI) and 3-(4-chlorophenyl)-6,7-dihydro-5H-pyrrolo[1,2-a]imidazole (CPPI) can inhibit the nuclear localization and transcriptional activity of AR and reduce the proliferation of AR-positive but not AR-negative prostate cancer cell lines. EPPI and CPPI did not inhibit nuclear localization of the glucocorticoid receptor or the estrogen receptor, suggesting they selectively target AR. In LNCaP tumor xenografts, CPPI inhibited the proliferation of relapsed LNCaP tumors. These findings suggest that EPPI and CPPI could serve as lead structures for the development of therapeutic agents for CRPC. Mol Cancer Ther; 16(10); 2120-9. ©2017 AACR.


Asunto(s)
Núcleo Celular/genética , Imidazoles/administración & dosificación , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Andrógenos/genética , Andrógenos/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Imidazoles/química , Masculino , Ratones , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Señales de Localización Nuclear/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/química , Receptores Androgénicos/genética , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Neoplasia ; 19(3): 207-215, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28167296

RESUMEN

Elongation factor, RNA polymerase II, 2 (ELL2) is expressed and regulated by androgens in the prostate. ELL2 and ELL-associated factor 2 (EAF2) form a stable complex, and their orthologs in Caenorhabditis elegans appear to be functionally similar. In C. elegans, the EAF2 ortholog eaf-1 was reported to interact with the retinoblastoma (RB) pathway to control development and fertility in worms. Because RB loss is frequent in prostate cancer, ELL2 interaction with RB might be important for prostate homeostasis. The present study explored physical and functional interaction of ELL2 with RB in prostate cancer. ELL2 expression in human prostate cancer specimens was detected using quantitative polymerase chain reaction coupled with laser capture microdissection. Co-immunoprecipitation coupled with deletion mutagenesis was used to determine ELL2 association with RB. Functional interaction between ELL2 and RB was tested using siRNA knockdown, BrdU incorporation, Transwell, and/or invasion assays in LNCaP, C4-2, and 22Rv1 prostate cancer cells. ELL2 expression was downregulated in high-Gleason score prostate cancer specimens. ELL2 could be bound and stabilized by RB, and this interaction was mediated through the N-terminus of ELL2 and the C-terminus of RB. Concurrent siRNA knockdown of ELL2 and RB enhanced cell proliferation, migration, and invasion as compared to knockdown of ELL2 or RB alone in prostate cancer cells. ELL2 and RB can interact physically and functionally to suppress prostate cancer progression.

14.
Endocrinology ; 158(12): 4189-4205, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29029019

RESUMEN

Mutations in the p53 tumor suppressor are frequent in patients with castration-resistant prostate cancer but less so in patients with localized disease, and patients who have Li-Fraumeni with germline p53 mutations do not have an increased incidence of prostate cancer, suggesting that additional molecular and/or genetic changes are required for p53 to promote prostate carcinogenesis. ELL-associated factor 2 (EAF2) is a tumor suppressor that is frequently downregulated in advanced prostate cancer. Previous studies have suggested that p53 binds to EAF2, providing a potential mechanism for their functional interactions. In this study, we tested whether p53 and EAF2 could functionally interact in prostate cancer cells and whether concurrent inactivation of p53 and EAF2 could promote prostate carcinogenesis in a murine knockout model. Endogenous p53 coprecipitated with EAF2 in prostate cancer cells, and deletion mutagenesis indicated that this interaction was mediated through the C terminus of EAF2 and the DNA binding domain of p53. Concurrent knockdown of p53 and EAF2 induced an increase in proliferation and migration in cultured prostate cancer cells, and conventional p53 and EAF2 knockout mice developed prostate cancer. In human prostate cancer specimens, concurrent p53 nuclear staining and EAF2 downregulation was associated with high Gleason score. These findings suggest that EAF2 and p53 functionally interact in prostate tumor suppression and that simultaneous inactivation of EAF2 and p53 can drive prostate carcinogenesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas Nucleares/genética , Próstata/metabolismo , Neoplasias de la Próstata/genética , Transactivadores/genética , Proteína p53 Supresora de Tumor/genética , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transformación Celular Neoplásica/metabolismo , Células HEK293 , Humanos , Inmunohistoquímica , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Clasificación del Tumor , Proteínas Nucleares/metabolismo , Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Unión Proteica , Interferencia de ARN , Transactivadores/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
15.
Oncotarget ; 7(20): 29245-54, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27058417

RESUMEN

RNA Polymerase II Elongation Factor (ELL)-associated factor 2 (EAF2) is a tumor suppressor frequently down-regulated in human prostate cancer. We previously reported that its binding partner ELL1 can enhance EAF2 protein stability and activity. Here we show that EAF2 can be polyubiquitinated and its degradation blocked by proteasome inhibitor. Co-immunoprecipitation detected EAF2 binding to SIAH2, an E3 ligase, and SIAH2 overexpression enhanced polyubiquitination of EAF2. Co-transfection of EAF2 binding partner ELL1 blocked EAF2 ubiquitination, providing a mechanism for EAF2 stabilization. Finally, EAF2K81R mutant, which exhibits reduced polyubiquitination and increased stability, was more potent than wild-type EAF2 in apoptosis induction. These findings suggest that SIAH2 is an E3 ligase for EAF2 polyubiquitination and ELL1 can enhance EAF2 level and function by blocking its polyubiquitination.


Asunto(s)
Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Factores de Transcripción/metabolismo , Factores de Elongación Transcripcional/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Masculino , Ubiquitinación
16.
Assay Drug Dev Technol ; 14(4): 226-39, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27187604

RESUMEN

Patients with castration-resistant prostate cancer (CRPC) can be treated with abiraterone, a potent inhibitor of androgen synthesis, or enzalutamide, a second-generation androgen receptor (AR) antagonist, both targeting AR signaling. However, most patients relapse after several months of therapy and a majority of patients with relapsed CRPC tumors express the AR target gene prostate-specific antigen (PSA), suggesting that AR signaling is reactivated and can be targeted again to inhibit the relapsed tumors. Novel small molecules capable of inhibiting AR function may lead to urgently needed therapies for patients resistant to abiraterone, enzalutamide, and/or other previously approved antiandrogen therapies. Here, we describe a high-throughput high-content screening (HCS) campaign to identify small-molecule inhibitors of AR nuclear localization in the C4-2 CRPC cell line stably transfected with GFP-AR-GFP (2GFP-AR). The implementation of this HCS assay to screen a National Institutes of Health library of 219,055 compounds led to the discovery of 3 small molecules capable of inhibiting AR nuclear localization and function in C4-2 cells, demonstrating the feasibility of using this cell-based phenotypic assay to identify small molecules targeting the subcellular localization of AR. Furthermore, the three hit compounds provide opportunities to develop novel AR drugs with potential for therapeutic intervention in CRPC patients who have relapsed after treatment with antiandrogens, such as abiraterone and/or enzalutamide.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Antagonistas de Receptores Androgénicos/metabolismo , Antagonistas de Receptores Androgénicos/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Benzoquinonas/metabolismo , Benzoquinonas/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Lactamas Macrocíclicas/metabolismo , Lactamas Macrocíclicas/farmacología , Masculino
17.
Zhonghua Yi Xue Za Zhi ; 85(19): 1319-21, 2005 May 25.
Artículo en Zh | MEDLINE | ID: mdl-16029630

RESUMEN

OBJECTIVE: To investigate the association between A49T polymorphism of SRD5A2 gene and risk of prostate cancer. METHODS: PCR was used to examine the A49T polymorphisms of SRD5A2 gene in the tissues of prostate cancer resected from 112 patients (CaP group) and the specimens of benign prostate hyperplasia (BPH group) resected from 89 patients. The association of A49T polymorphism with age of onset, FPSA, TPSA, F/T, T stage, and Gleason score were analyzed. RESULTS: There was no significant difference in A49T polymorphism between the CaP and BPH groups (P > 0.05). The average age of CaP patients was significantly higher than that of the BPH patients (P < 0.05). In the CaP patients, the Gleason score was significantly higher, and the age of onset was significantly lower in the AT + TT genotype than in the AA genotype (both P < 0.05) 2. The age of onset of the AA + AT group was significantly lower than that of the AA group (P < 0.05). CONCLUSION: AA + AT genotype may be of worse prognosis, however, without significant difference. Rank scoring may reflect the relation between Gleason score and A49T genotype and estimate the prognosis better than two-level discrete evaluation.


Asunto(s)
3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo Genético , Neoplasias de la Próstata/genética , Anciano , Anciano de 80 o más Años , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Riesgo
18.
Sci China C Life Sci ; 47(2): 148-57, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15379247

RESUMEN

To isolate the over-expressed genes in human renal cell carcinoma (RCC) and analyze its molecular basis of carcinogenesis, we used the mRNA from human RCC tissues as tester and that from the matched normal kidney tissues as driver to construct the suppression subtractive hybridization library. 379 of the subtracted clones were arrayed onto a nylon membrane and the over-expressed genes were then screened by hybridizing the filter with radioactively labeled cDNA from RCC and matched normal kidney tissues. 67 clones over-expressed in RCC by a factor of 6 or more were sequenced and its identities were analyzed in GenBank database. 4 clones were previously unknown fragments and 2 clones represent KIAA genes. The rest clones were the known genes and some of them were RCC-related, including vascular endothelial growth factor, vimentin and tissue factor. Most of the known genes were the RCC-related genes previously unknown, including zinc ribbon domain-containing 1 protein (ZNRD1), pituitary tumor transforming gene1 (PTTG1). Northern blot and semi-quantitative RT-PCR confirmed that the mRNA levels of the 3 novel fragments and 1 KIAA and 3 known genes were significantly higher in RCC than in the matched normal kidney tissues. Immunohistochemical and Western blot analysis for PTTG1 and ZNRD1 revealed increased protein level in RCC. The over-expressed genes in RCC are the potential molecular targets for diagnosis and therapy and it is very important to understand the molecular mechanism of RCC through the profile of over-expressed genes.


Asunto(s)
Carcinoma de Células Renales/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Renales/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Secuencia de Bases , Cartilla de ADN , Biblioteca de Genes , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Hibridación de Ácido Nucleico , ARN Mensajero/genética , Células Tumorales Cultivadas
19.
Beijing Da Xue Xue Bao Yi Xue Ban ; 36(2): 154-8, 2004 Apr.
Artículo en Zh | MEDLINE | ID: mdl-15100733

RESUMEN

OBJECTIVE: To evaluate the effect of clusterin with and without leader sequence on overexpression preventing apoptosis in human prostate LNCaP cells. METHODS: The plasmid pIRES2-EGFP was used to generate the clusterin expression constructs with full-length or without the leader sequence (designated as pIRES2-EGFP/cluac, pIRES2-EGFP/clubc, respectively). Western blot analysis was employed to compare clusterin expression levels in the lysis and supernatant fluid of clusterin transfected LNCaP cells in vitro. The distribution of different functional domains of clusterin in cells was detected with Immunocytochemical staining. The clusterin's protective role of Na2SeO3-induced apoptosis in LNCaP cells was examined by flow cytometry (FCM) and fluorescence microscope. RESULTS: Clusterin expression was detected in the lysis and supernatant fluid of pIRES2-EGFP/cluac transfected LNCaP cells, while clusterin was found only in lysis liquid of pIRES2-EGFP/clubc transfected LNCaP cells, but not found in their supernatant fluid. The distribution of cluserin in the plasm of pIRES2-EGFP/cluac transfected cells was aggregative, and on the other hand, clusterin distributed dispersedly in pIRES2-EGFP/clubc transfected cells. Its anti-apoptotic property in LNCaP cells was proved by FCM and fluorescence microscope. CONCLUSION: It is apparent that clusterin plays an important role in preventing apoptosis in prostate cancer, and the presence of the leader sequence is necessary for clusterin's anti-apoptotic function.


Asunto(s)
Apoptosis , Glicoproteínas/fisiología , Chaperonas Moleculares/fisiología , Neoplasias de la Próstata/patología , Western Blotting , Línea Celular Tumoral , Clusterina , Glicoproteínas/genética , Humanos , Masculino , Chaperonas Moleculares/genética , Transfección
20.
Zhonghua Yi Xue Za Zhi ; 83(4): 333-7, 2003 Feb 25.
Artículo en Zh | MEDLINE | ID: mdl-12812655

RESUMEN

OBJECTIVE: To investigate the expression of human-mouse chimeric antibody ch-BD1 against human bladder cancer and its affinity to human bladder cancer in vitro and in vivo. METHODS: Three kinds of mutated fragments of dihydrofolate reductase (DHFR) gene were created by techniques of molecular biology to decrease the transcriptional activities and then cloned into pDHL-BD1 so as to construct the vectors pWSD1-BD1, pWSD2-BD1, and pWSD3-BD1 expressing the human-mouse chimeric antibody ch-BD1 against human bladder cancer with decreased expression of DHFR gene. These vectors and pDHL-BD1 were transfected into Chinese hamster ovary cell (CHO)/DHFR- cell respectively. 72 hours later Northern blotting was used to examine their DHFR gene expression. Methotrexate (MTX) of increasing concentrations was added into the culture of transfected CHO/DHFR-cells. The ch-BD1 levels in the supernatants were measured. Purified ch-BD1 was labeled by (99m)TcO(4)(-) and added into the serially diluted solutions of human bladder cancer EJ cells to examine their radioactivities and calculate their affinity constants. EJ cells were injected into the roots of hind limb of 3 Balb/C mice. Four weeks later, (99m)TcO(4)(-)-labeled antibody ch-BD1 were injected into the rats' caudal veins. Radioimmunoimaging was conducted to examine the distribution of the antibody. RESULTS: The sequence of DHFR gene expression levels from strong to weak in the constructed vectors was as follows: pDHL-BD1 > pWS1-BD1 > pWS3-BD1 > pWS2-BD1. When the concentration of MTX was 10(-6) mol/L the expression level of ch-BD1 was significantly correlated to the expression level of DHFR gene, the lower the baseline expression level of DHFR gene the higher the expression level of ch-BD1. After the serially diluted EJ cells were co-incubated with the (99m)TcO(4)(-)-labeled antibody ch-BD1 the immunoactivity ratio of ch-BD1 was 76%, and that of murine monoclonal antibody was 81%; the affinity constant of ch-BD1 was 3.56 x 10(9) M(-1), and that of murine monoclonal antibody BD1 was 1.22 x 10(9) M(-1). 6 hours after injection of the (99m)TcO(4)(-)-labeled antibody ch-BD1 into the mice body it was mainly distributed in the tumor, 22 hours later, it was specifically distributed in tumor, and 24 hours later it was still concentrated here. CONCLUSION: Decrease of the baseline expression level of DHFR gene effectively increases the amplification of MTX to exogenous gene. The human-mouse chimeric antibody ch-BD1 shows an ideal affinity activity to human bladder cancer in vivo and in vitro and has a certain clinical prospect.


Asunto(s)
Anticuerpos Monoclonales/genética , Anticuerpos Antineoplásicos/genética , Proteínas Recombinantes de Fusión/biosíntesis , Tetrahidrofolato Deshidrogenasa/genética , Neoplasias de la Vejiga Urinaria/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Antineoplásicos/inmunología , Afinidad de Anticuerpos , Células CHO , Cricetinae , Vectores Genéticos , Humanos , Metotrexato/farmacología , Ratones , Ratones Endogámicos BALB C , Tecnecio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA