Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Oncogene ; 36(24): 3417-3427, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28092670

RESUMO

Recent evidence has implicated the transmembrane co-receptor neuropilin-1 (NRP1) in cancer progression. Primarily known as a regulator of neuronal guidance and angiogenesis, NRP1 is also expressed in multiple human malignancies, where it promotes tumor angiogenesis. However, non-angiogenic roles of NRP1 in tumor progression remain poorly characterized. In this study, we define NRP1 as an androgen-repressed gene whose expression is elevated during the adaptation of prostate tumors to androgen-targeted therapies (ATTs), and subsequent progression to metastatic castration-resistant prostate cancer (mCRPC). Using short hairpin RNA (shRNA)-mediated suppression of NRP1, we demonstrate that NRP1 regulates the mesenchymal phenotype of mCRPC cell models and the invasive and metastatic dissemination of tumor cells in vivo. In patients, immunohistochemical staining of tissue microarrays and mRNA expression analyses revealed a positive association between NRP1 expression and increasing Gleason grade, pathological T score, positive lymph node status and primary therapy failure. Furthermore, multivariate analysis of several large clinical prostate cancer (PCa) cohorts identified NRP1 expression at radical prostatectomy as an independent prognostic biomarker of biochemical recurrence after radiation therapy, metastasis and cancer-specific mortality. This study identifies NRP1 for the first time as a novel androgen-suppressed gene upregulated during the adaptive response of prostate tumors to ATTs and a prognostic biomarker of clinical metastasis and lethal PCa.


Assuntos
Neuropilina-1/genética , Neuropilina-1/metabolismo , Neoplasias de Próstata Resistentes à Castração/mortalidade , Neoplasias da Próstata/tratamento farmacológico , Regulação para Cima , Antagonistas de Androgênios/uso terapêutico , Linhagem Celular Tumoral , Progressão da Doença , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Gradação de Tumores , Metástase Neoplásica , Neoplasias da Próstata/genética , Neoplasias da Próstata/mortalidade , Análise de Sobrevida
2.
Gene Ther ; 23(5): 460-8, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26905370

RESUMO

Urothelial bladder cancer is the most common malignancy of the urinary tract. Although most cases are initially diagnosed as non-muscle-invasive, more than 80% of patients will develop recurrent or metastatic tumors. No effective therapy exists currently for late-stage metastatic tumors. By intravesical application, local administration of oncolytic Herpes Simplex virus (oHSV-1) can provide a promising new therapy for this disease. However, its inherent neurotoxicity has been a perceived limitation for such application. In this study, we present a novel microRNA-regulatory approach to reduce HSV-1-induced neurotoxicity by suppressing viral replication in neurons while maintaining oncolytic selectivity toward urothelial tumors. Specifically, we designed a recombinant virus that utilizes differentially expressed endogenous microR143 (non-cancerous, ubiquitous) and microR124 (neural-specific) to regulate expression of ICP-4, a gene essential for HSV-1 replication. We found that expression of ICP-4 must be controlled by a combination of both miR143 and miR124 to achieve the most effective attenuation in HSV-1-induced toxicity while retaining maximal oncolytic capacity. These results suggest that interaction between miR143 and miR124 may be required to successfully regulate HSV-1 replication. Our resent study is the first proof-in-principle that miRNA combination can be exploited to fine-tune the replication of HSV-1 to treat human cancers.


Assuntos
Terapia Genética/métodos , Herpesvirus Humano 1/genética , MicroRNAs/genética , Neoplasias da Bexiga Urinária/terapia , Administração Intravesical , Animais , Vetores Genéticos , Humanos , Camundongos , MicroRNAs/uso terapêutico , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Neoplasias da Bexiga Urinária/genética , Urotélio/patologia , Replicação Viral/genética
3.
Cancer Gene Ther ; 19(1): 19-29, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21921943

RESUMO

In this study, we have taken advantage of over-expression of eukaryotic translation initiation factor 4E (eIF4E) in prostate cancer cells to design a viral-based targeting system of prostate cancer. Three different lengths of 5'-untranslated regions (5'UTRs) derived from either fibroblast growth factor-2 (FU-FGF2-GW) or ornithine decarboxylase (FU-ODC149-GW and FU-ODC274-GW) were inserted upstream of enhanced green fluorescent protein (GFP) gene in a lentiviral backbone. Both nonmalignant control (PNT1B and BPH-1) and neoplastic (LNCaP, C4-2, DU145 and PC-3) prostate cell lines were transfected with each plasmid or virus alone, or in the presence of siRNA against eIF4E, and their expression was monitored via GFP protein levels. Two 5'UTRs (FU-FGF2-GW and FU-ODC-GW) were selected as being most sensitive to eIF4E status. Lentiviruses containing these sequences were injected directly into the prostates of PTEN(-/-) (tumor-bearing) and control mice. Immunofluorescence data and western blot analyses determined that a lentivirus containing a 5'UTR derived from FGF-2 is the best candidate for directing selective gene expression in the prostate tumors of PTEN(-/-) mice in vivo. This study demonstrates that judicious selection of a complex 5'UTR can enhance selective targeting of viral-based gene therapies for prostate cancer.


Assuntos
Regiões 5' não Traduzidas , Fatores de Crescimento de Fibroblastos/genética , Terapia Genética/métodos , Ornitina Descarboxilase/genética , PTEN Fosfo-Hidrolase/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/terapia , Animais , Linhagem Celular Tumoral , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Ornitina Descarboxilase/biossíntese , Ornitina Descarboxilase/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/virologia , Transfecção
4.
Cancer Gene Ther ; 16(11): 820-31, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19373278

RESUMO

In this study, we took advantage of the overexpression of human epidermal growth factor receptor 2 (HER-2) in prostate cancers to design lentiviruses with modified envelope proteins that bind antibodies to specific cell-surface antigens. When bound to trastuzumab (Herceptin, Genentech, CA), lentiviruses were able to selectively infect androgen-sensitive LNCaP and castration-resistant C4-2 human prostate cancer cell lines, both of which express high levels of HER-2. To test for a therapeutic effect, we engineered our antibody-binding lentiviruses to express thymidine kinase, which can convert the non-toxic pro-drug ganciclovir (GCV) into a cytotoxic form. LNCaP and C4-2 cells infected by these viruses were sensitive to GCV killing. In vivo, C4-2 xenograft tumors treated either intratumorally or i.v. with trastuzumab-bound lentivirus expressed luciferase, although the latter route was less tumor specific. When a prostate-specific promoter for governing luciferase expression was combined with trastuzumab-mediated delivery, there was a further enrichment in targeting viral gene expression in prostate tumors. In conclusion, we found that although prostate cancers that express high levels of HER-2 are resistant to the killing effects of trastuzumab, they can be targeted for selective gene expression and destruction by viruses with envelope proteins engineered to bind this antibody.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Lentivirus/química , Neoplasias da Próstata/tratamento farmacológico , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais Humanizados , Antineoplásicos/química , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/metabolismo , Vetores Genéticos/química , Vetores Genéticos/genética , Vetores Genéticos/fisiologia , Humanos , Imuno-Histoquímica , Lentivirus/genética , Lentivirus/fisiologia , Masculino , Camundongos , Camundongos Nus , Receptor ErbB-2/metabolismo , Trastuzumab , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Urologe A ; 47(9): 1145-51, 2008 Sep.
Artigo em Alemão | MEDLINE | ID: mdl-18670747

RESUMO

Patients with high-risk bladder cancer who do not respond to bacillus Calmette-Guerin (BCG) immunotherapy represent a significant therapeutic challenge. The addition of interferon to BCG has recently evolved as a second-line treatment option; however, many high-grade tumors are nonresponsive to interferon. Thus, replication-competent oncolytic vesicular stomatitis viruses (VSV) that selectively target interferon-refractory tumors are promising intravesical agents. In vitro, wild-type VSV as well as a mutant variant (AV3) that has an impaired ability to shut down innate immunity preferentially killed undifferentiated, interferon-nonresponsive bladder cancer cells. Testing of these viruses in an orthotopic murine model of high-grade bladder cancer, which we have recently validated, revealed that both AV3 and wild-type VSV significantly inhibited orthotopic tumor growth. Despite the use of immunocompromised nude mice, there was no evidence of toxicity. In conclusion, VSV instillation therapy demonstrated strong antitumor activity and safety in an orthotopic model of high-risk disease. These findings provide preclinical proof-of-principle for the intravesical use of VSV, especially in interferon-refractory patients.


Assuntos
Carcinoma de Células de Transição/terapia , Terapia Viral Oncolítica/métodos , Vírus da Estomatite Vesicular Indiana/genética , Administração Intravesical , Animais , Carcinoma de Células de Transição/imunologia , Carcinoma de Células de Transição/patologia , Linhagem Celular Tumoral , Luciferina de Vaga-Lumes , Humanos , Interferons/metabolismo , Camundongos , Camundongos Nus , Mutação/genética , Invasividade Neoplásica , Transplante de Neoplasias , Carga Tumoral , Bexiga Urinária/imunologia , Bexiga Urinária/patologia , Vírus da Estomatite Vesicular Indiana/imunologia , Replicação Viral
6.
Cancer Gene Ther ; 14(7): 652-60, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17479106

RESUMO

The aim of the present study was to determine whether a prostate-specific amplicon, containing a probasin-derived promoter (ARR(2)PB) upstream of an essential Herpes simplex virus-1 (HSV-1) viral gene, infected-cell polypeptide 4 (ICP4), could complement an HSV-1 helper virus with this gene deleted (ICP4-) and cause lytic replication specifically in prostate cancer cells. Two amplicon constructs, CMV-ICP4 and ARR(2)PB-ICP4, were packaged by a replication-deficient ICP4- helper virus. The amplicon viruses could complement ICP4- helper viruses to efficiently replicate and cause cell lysis in prostate cancer cells. Intratumoral injection of LNCaP human prostate cancer xenografts with either amplicon/helper virus resulted in >75% reduction in tumor volume and serum prostate specific antigen (PSA). Histological and Q-PCR (quantitative PCR) analyses indicated that the toxicity in nontumor tissues was much lower with ARR(2)PB-ICP4 than with CMV-ICP4 amplicon/helper virus. In conclusion, a replication-deficient HSV-1 virus could be complemented by an amplicon virus to restore its oncolytic activity in a tissue-specific and low toxicity fashion, illustrating that this approach could be a potentially useful strategy for developing an oncolytic viral therapy for prostate cancer.


Assuntos
Herpesvirus Humano 1/fisiologia , Proteínas Imediatamente Precoces/genética , Neoplasias da Próstata/terapia , Replicação Viral/fisiologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Chlorocebus aethiops , Amplificação de Genes , Terapia Genética , Vírus Auxiliares/genética , Herpesvirus Humano 1/genética , Humanos , Masculino , Neoplasias da Próstata/patologia , RNA Viral/genética , RNA Viral/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Vero
7.
Cancer Gene Ther ; 13(1): 32-43, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16052226

RESUMO

To develop a gene therapy that would selectively kill prostate cancer cells while sparing normal cells, we have constructed lentiviral vectors that contain a therapeutic gene with a short DNA sequence in the 5'-untranslated region (UTR) that is recognized by the translation initiation factor, eIF4E, which is often overexpressed in malignant cells. Infection of cancer (LNCaP, PC-3M, DU145, and MCF-7 cells) and noncancer cell lines (BPH-1, 267-B1, Plat-E, and Huvec-c cells) with lentivirus having a CMV-promoter and EGFP reporter resulted in high levels of EGFP expression in all cells, whereas, inclusion of the eIF4E UTR recognition sequence restricted high expression to cancer cells and Plat-E cells, which also express substantial levels of eIF4E. Infection of the cells with lentiviral vectors having this UTR in front of the HSV thymidine kinase suicide gene resulted in differential sensitivity to the killing effects of ganciclovir, with at least 100-fold more drug required to kill noncancer cells than cancer cells. Furthermore, in experiments where the CMV promoter was replaced by the prostate-specific ARR(2)PB promoter, the killing effects of ganciclovir were restricted to prostate cancer cells and not seen in nonprostate cancer cells. Our results indicate that combined translational regulation, by incorporation of an eIF4E-UTR recognition sequence into a therapeutic gene, together with transcriptional regulation with a prostate-specific promoter, may provide a means to selectively destroy prostate cancer cells while sparing normal prostate cells.


Assuntos
Morte Celular/efeitos dos fármacos , Fator de Iniciação 4E em Eucariotos/metabolismo , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos/farmacologia , Lentivirus/patogenicidade , Neoplasias da Próstata/metabolismo , Relação Dose-Resposta a Droga , Fator de Iniciação 4E em Eucariotos/farmacologia , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Imuno-Histoquímica , Lentivirus/metabolismo , Masculino , Modelos Genéticos , Regiões Promotoras Genéticas/fisiologia , Timidina Quinase/metabolismo , Transfecção , Células Tumorais Cultivadas
8.
Endocrinology ; 141(6): 2257-65, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10830316

RESUMO

Although insulin-like growth factor (IGF) binding protein-5 (IGFBP-5) is highly up-regulated in normal and malignant prostate tissues after androgen withdrawal, its functional role in castration-induced apoptosis and androgen-independent progression remains undefined. To analyze the functional significance of IGFBP-5 overexpression in IGF-I-mediated mitogenesis and progression to androgen-independence, IGFBP-5-overexpressing human androgen-dependent LNCaP prostate cancer cells were generated by stable transfection. The growth rates of IGFBP-5-transfected LNCaP cells were significantly faster, compared with either the parental or vector-only transfected LNCaP cells in both the presence and absence ofdihydrotestosterone. IGFBP-5-induced increases in LNCaP cell proliferation occurs through both IGF-I-dependent and -independent pathways, with corresponding increases in the cyclin D1 messenger RNA expression and the fraction of cells in S + G2/M phases of the cell cycle. Changes in Akt/protein kinase B, a downstream component of phosphatidylinositol 3'-kinase (PI3K) pathway, in the LNCaP sublines also paralleled changes in their growth rates. Although treatment with a PI3K inhibitor induced apoptosis in both control and IGFBP-5-overexpressing LNCaP cells, this PI3K inhibitor-induced apoptosis was prevented by exogenous IGF-I treatment only in IGFBP-5 transfectants, suggesting that IGFBP-5 overexpression can potentiate the antiapoptotic effects of IGF-I. Furthermore, tumor growth and serum prostate-specific antigen levels increased several fold faster in mice bearing IGFBP-5-transfected LNCaP tumors after castration, despite having similar tumor incidence and tumor growth rates with controls when grown in intact mice before castration. Collectively, these data suggest that IGFBP-5 overexpression in prostate cancer cells after castration is an adaptive cell survival mechanism that helps potentiate the antiapoptotic and mitogenic effects of IGF-I, thereby accelerating progression to androgen independence through activation of the PI3K-Akt/ protein kinase B signaling pathway.


Assuntos
Androgênios/farmacologia , Expressão Gênica , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Próstata/patologia , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Ciclina D1/genética , Di-Hidrotestosterona/farmacologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Fator de Crescimento Insulin-Like I/farmacologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/metabolismo , RNA Mensageiro/metabolismo , Transfecção , Células Tumorais Cultivadas
9.
Cancer Res ; 60(9): 2547-54, 2000 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10811138

RESUMO

Testosterone-repressed prostate message-2 (TRPM-2) expression is highly up-regulated in normal and malignant prostate cells after androgen withdrawal. Although recent studies have suggested a protective role of TRPM-2 expression against apoptosis in several experimental models, the functional role of TRPM-2 in chemotherapy-induced apoptosis remains undefined. Here, we demonstrated that overexpression of TRPM-2 in human androgen-dependent LNCaP prostate cancer cells by stable transfection rendered them highly resistant to paclitaxel treatment than control LNCaP cells, with a 20-fold higher IC50 through the inhibition of apoptotic cell death. In mice bearing TRPM-2-overexpressing LNCaP tumors, tumor volume and serum prostate-specific antigen increased two to three times faster after castration and paclitaxel treatment compared with mice bearing control tumors. We then tested the efficacy of combined treatment with antisense TRPM-2 oligodeoxynucleotide (ODN) and paclitaxel in the mouse androgen-dependent Shionogi tumor model. Antisense TRPM-2 ODN treatment significantly enhanced paclitaxel chemosensitivity of Shionogi tumor cells in a dose-dependent manner, reducing the IC50 by 75%. Combined treatment of Shionogi cells with 500 nM antisense TRPM-2 ODN and 10 nM paclitaxel-induced apoptosis, either agent alone did not. Adjuvant administration of antisense TRPM-2 ODN and polymeric micellar paclitaxel after castration resulted in reduced TRPM-2 levels in vivo and a significant delay of emergence of androgen-independent recurrent Shionogi tumors compared with administration of either agent alone. Furthermore, combined treatment of mice bearing androgen-independent recurrent Shionogi tumors with antisense TRPM-2 ODN and micellar paclitaxel inhibited tumor growth compared with treatment with either agent alone. Collectively, these findings demonstrate that TRPM-2 overexpression helps confer a chemoresistant phenotype through inhibition of apoptosis, and that antisense TRPM-2 ODN may be useful in enhancing the effects of cytotoxic chemotherapy in hormone-refractory prostate cancer.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Glicoproteínas/metabolismo , Chaperonas Moleculares , Neoplasias da Próstata/genética , Animais , Antineoplásicos Fitogênicos/farmacologia , Northern Blotting , Western Blotting , Clusterina , Fragmentação do DNA , Progressão da Doença , Relação Dose-Resposta a Droga , Feminino , Glicoproteínas/genética , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Oligonucleotídeos Antissenso/farmacologia , Paclitaxel/farmacologia , Fenótipo , Antígeno Prostático Específico/sangue , Fatores de Tempo , Células Tumorais Cultivadas
10.
Cancer Res ; 60(1): 170-6, 2000 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-10646870

RESUMO

Although initially reported as an androgen-repressed gene in the rat prostate, the functional role of testosterone-repressed prostate message-2 (TRPM-2) in apoptosis remains undefined. Inhibition of castration-induced apoptosis by calcium channel blocker treatment in androgen-dependent Shionogi tumors resulted in the prevention of TRPM-2 gene up-regulation, suggesting that TRPM-2 is not directly androgen-repressed, but is regulated by apoptotic stimuli. The overexpression of the TRPM-2 gene in human androgen-dependent LNCaP prostate cancer cells by stable transfection rendered them highly resistant to androgen ablation in vivo. We then tested the efficacy of antisense TRPM-2 oligodeoxynucleotide (ODN) therapy in the Shionogi tumor model and demonstrated that the systemic administration of antisense TRPM-2 ODNs in mice bearing Shionogi tumors after castration resulted in a more rapid onset of apoptosis and time to complete regression, as well as a significant delay of emergence of androgen-independent recurrent tumors compared to control ODN treatment. Collectively, these findings illustrate that TRPM-2 is an antiapoptotic rather than an androgen-repressed gene that confers resistance to androgen ablation and thereby helps accelerate the progression to androgen independence.


Assuntos
Apoptose/efeitos dos fármacos , Glicoproteínas/metabolismo , Chaperonas Moleculares , Proteínas de Neoplasias/metabolismo , Neoplasias da Próstata/metabolismo , Animais , Northern Blotting , Western Blotting , Bloqueadores dos Canais de Cálcio/farmacologia , Clusterina , Progressão da Doença , Vetores Genéticos , Glicoproteínas/efeitos dos fármacos , Glicoproteínas/genética , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/genética , Neoplasias Hormônio-Dependentes/metabolismo , Oligodesoxirribonucleotídeos Antissenso , Orquiectomia , RNA Mensageiro/metabolismo , Regulação para Cima/efeitos dos fármacos
11.
J Mol Endocrinol ; 22(3): 313-25, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10343290

RESUMO

Glucocorticoid and androgen receptors have been shown to function through the same palindromic glucocorticoid response element (GRE) and yet have differential effects on gene transcription. In this study, we examined the functional and structural relationship of the androgen and glucocorticoid receptors with the androgen responsive region (ARR) of the probasin (PB) gene containing two androgen receptor binding sites, ARBS-1 and ARBS-2. Transfection studies indicated that one copy of each cis-acting DNA element was essential for maximal androgen-induced chloramphenicol acetyltransferase (CAT) activity and that androgen selectivity was maintained when multiple copies of the minimal wild type (wt) androgen responsive region containing both ARBS-1 and ARBS-2 (-244 to -96) were subcloned in front of the thymidine kinase promoter. Furthermore, replacing the androgen response region with 1, 2 or 3 copies of either ARBS-1 or ARBS-2 restored less than 4% of the biological activity seen with the wt PB ARR. Multiple copies of either ARBS-1 or ARBS-2 did not result in glucocorticoid-induced CAT gene activity. By comparison, 1 or 2 copies of the tyrosine aminotransferase (TAT) GRE, as well as the mouse mammary tumour virus GRE, were strong inducers of CAT activity in response to both androgen and glucocorticoid treatment. In addition, band shift assays demonstrated that although the synthetic glucocorticoid receptor, GR-DNA binding domain (GR-DBD), and the synthetic androgen receptor, AR2, could interact with the TAT GRE (dissociation constants Kd of 63.9 and 14.1 respectively), only AR2 but not GR-DBD binding could be detected on ARBS-1 and ARBS-2. Our findings provide further evidence that androgen-induced regulation of gene transcription can occur through androgen-specific DNA binding sites that are distinct from the common GRE.


Assuntos
Proteína de Ligação a Androgênios/metabolismo , Androgênios/farmacologia , Glucocorticoides/farmacologia , Receptores Androgênicos/metabolismo , Proteína de Ligação a Androgênios/genética , Androgênios/metabolismo , Sequência de Bases , Cloranfenicol O-Acetiltransferase/genética , Pegada de DNA , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Dados de Sequência Molecular , Receptores de Glucocorticoides/metabolismo , Células Tumorais Cultivadas
12.
Prostate ; 36(4): 256-63, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9719026

RESUMO

BACKGROUND: The purpose of this study was to determine the contribution of different transactivating regions of the androgen receptor (AR) to the induction of androgen-regulated promoters in poorly (PC3 cells) and well-differentiated (LNCaP cells) prostate cancer cell lines. METHODS: PC3 and LNCaP cells were co-transfected with plasmids expressing full-length AR or deletion mutants together with luciferase reporters linked to the probasin (PB) and PSA promoters; as well as to ARR3tk, a PB-derived recombinant promoter. RESULTS: Androgen induction of the ARR3tk promoter in the presence of AR was 8- to 10-fold higher than that seen with the PB promoter. Activation of ARR3tk was greatest with an androgen-independent construct in which the first 231 amino acids and the ligand binding domain had been removed, indicating that this promoter is more responsive to activating functions in the N-terminal domain than in the ligand binding domain. By comparison, induction of the PB promoter was greatest with the full-length AR, which suggests that the ligand binding domain also makes a major contribution to the activation of this promoter. In similar analyses with the PSA promoter, AR regions required for promoter induction was dependent on the host cell type. In PC3 cells, the predominant AR transactivation function was androgen-independent and resided in the N-terminal domain, whereas in LNCaP cells, the highest level of induction was androgen dependent and also required participation of the ligand binding domain. CONCLUSIONS: Our results indicate that the relative utilization of transactivating functions in N-terminal and ligand binding domains of the AR is promoter and cell specific.


Assuntos
Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/genética , Proteína de Ligação a Androgênios/genética , Sequência de Bases , Sítios de Ligação/genética , Diferenciação Celular , Primers do DNA/genética , Genes Reporter , Humanos , Luciferases/genética , Masculino , Regiões Promotoras Genéticas , Antígeno Prostático Específico/genética , Neoplasias da Próstata/patologia , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Deleção de Sequência , Timidina Quinase/genética , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas
13.
J Androl ; 19(2): 127-35, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9570735

RESUMO

We studied the response of the human ornithine decarboxylase (ODC) promoter to androgen in human prostate cancer cell lines. In the well-differentiated, androgen-sensitive human prostate cancer line LNCaP, a genomic ODC promoter fragment that includes putative androgen response elements was suppressed by androgen. In contrast, the androgen-regulated probasin promoter was induced by androgens. The ODC promoter was also induced by cotransfected androgen receptor in the poorly differentiated, androgen-insensitive human prostate cancer cell line PPC-1. We examined the effects of cotransfected mutant androgen receptors containing the LNCaP mutation or DNA-binding mutations. All cotransfected androgen receptors switched the ODC androgen response from suppression to induction in LNCaP cells. Gel-shift and DNA footprint assays demonstrated androgen receptor binding to an ODC sequence that does not contain a consensus androgen response element. Deletion of the sequence abolished androgen suppression of the ODC promoter. We propose a model of pleiotropic gene regulation by androgen that requires a regulatory balance between androgen receptor and a transcription factor binding to the nonconsensus androgen response element.


Assuntos
Androgênios/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Ornitina Descarboxilase/genética , Regiões Promotoras Genéticas , Neoplasias da Próstata/enzimologia , Androgênios/metabolismo , Animais , Sítios de Ligação , Diferenciação Celular , Pegada de DNA , Cães , Ativação Enzimática , Humanos , Masculino , Ornitina Descarboxilase/metabolismo , Inibidores da Ornitina Descarboxilase , Neoplasias da Próstata/patologia , Proteína Quinase C/metabolismo , Receptores Androgênicos/metabolismo , Células Tumorais Cultivadas
14.
Cancer Metastasis Rev ; 17(4): 401-9, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-10453284

RESUMO

Epigenetic mechanisms may be the main driving force for critical changes in gene expression that are responsible for progression of prostate cancers. The three most extensively characterized mechanisms for epigenetic gene-regulation are (i) changing patterns of DNA methylation, (ii) histone acetylations/deacetylations, and (iii) alterations in regulatory feedback loops for growth factors. Several studies have indicated that DNA hypermethylation is an important mechanism in prostate cancer for inactivation of key regulatory genes such as E-cadherin, pi-class glutathione S-transferase, the tumor suppressors CDKN2 and PTEN, and IGF-II. Similarly, histone acetylations and deacetylations are frequently associated respectively with transcriptional activation (e.g. IGFBP-2 and p21) and repression (e.g. Mad:Max dimers) of genes linked to prostate cancer progression. Recently, histone acetyltransferase and deacetylase activities have been shown to be intrinsic with transcriptional coregulator proteins that bind to steroid receptors (e.g. SRC-1 and PCAF). Changes in regulatory feedback loops for growth factors with prostate cancer progression tend toward shifts from paracrine to autocrine control where the receptor and ligand are produced by the same cell. While there are several examples of this progression pattern in prostate tumors such as with IGF, FGF, TGF-alpha and their respective receptors, the precise mechanism (i.e. epigenetic or mutational) is less certain. In the context of treatment options, the contribution of mutational versus epigenetic events to prostate cancer progression is an important consideration. Irreversible genetic changes are likely to be less amenable to therapeutic control than are epigenetic ones.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/genética , Acetilação , Animais , Metilação de DNA , Progressão da Doença , Retroalimentação , Impressão Genômica , Substâncias de Crescimento/metabolismo , Histonas/metabolismo , Humanos , Masculino , Camundongos , Neoplasias da Próstata/metabolismo
15.
J Biol Chem ; 272(28): 17485-94, 1997 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-9211894

RESUMO

In exploring the possible mechanisms of androgen independence of prostate-specific antigen (PSA) gene expression, we investigated the effect of elevating AP-1 by both 12-O-tetradecanoylphorbol 13-acetate (TPA) treatment and transfection of the c-Jun expression vector in LNCaP cells. Transcription of PSA is initiated when ligand-activated androgen receptor (AR) binds to a region in the PSA promoter that contains an androgen-responsive element (ARE). It was found that TPA inhibited androgen-induced PSA gene expression by a mechanism that did not alter nuclear levels of AR protein. Overexpression of AP-1 (jun and fos proteins) also inhibited androgen-induced PSA promoter activity. These observations were apparently related to the disruption of AR.ARE complexes as demonstrated by the results of electrophoretic mobility shift assays. Specifically, c-Jun inhibited the formation of AR.ARE complexes and conversely that AR-glutathione S-transferase proteins inhibited the formation of c-Jun.TPA-responsive element (TRE) complexes. Consistent with the inhibitory effect of both proteins, anti-c-Jun antibody blocked the inhibition of AR.ARE complex formation by c-Jun. A similar, but less marked, effect was obtained when anti-AR antibody was used to prevent AR inhibition of c-Jun.TRE complex formation. These findings together with results obtained from co-immunoprecipitation experiments strongly suggest that mutual repression of DNA binding activity is due to direct interaction between the two proteins and that the degree of repression may be determined by the ratio of AR to c-Jun. The mechanism of repression studied in mutant analysis experiments yielded evidence of an interaction between the DNA- and ligand-binding domains of AR and the leucine zipper region of c-Jun. Thus, the AR is similar to other nuclear receptors in its ability to interact with AP-1. This association provides a link between AP-1 and AR signal transduction pathways and may play a role in the regulation of the androgen-responsive PSA gene.


Assuntos
Androgênios/metabolismo , Expressão Gênica/efeitos dos fármacos , Antígeno Prostático Específico/genética , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fator de Transcrição AP-1/metabolismo , Carcinógenos/farmacologia , DNA de Neoplasias/metabolismo , Humanos , Masculino , Regiões Promotoras Genéticas , Antígeno Prostático Específico/biossíntese , Ligação Proteica , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , RNA Mensageiro/metabolismo , Receptores Androgênicos , Acetato de Tetradecanoilforbol/farmacologia , Células Tumorais Cultivadas
16.
Cancer Res ; 57(8): 1584-9, 1997 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9108464

RESUMO

Several metastasizing murine and human animal models for prostate cancer are available. However, these models are androgen-independent and lack differentiated features such as androgen receptor and androgen-regulated gene expression like prostate-specific antigen (PSA). The objective of this study was to develop a metastasizing prostate cancer model with differentiated features using the human LNCaP cell line. Athymic and SCID mice were injected either s.c. or intraprostatically with 1 x 10(6) LNCaP cells. Changes in serum and tumor PSA mRNA levels were determined before and after castration to assess time to androgen-independent progression. Local tumor and metastatic growth was assessed at sacrifice after 12 weeks. Reverse transcription-PCR (RT-PCR) was used to detect circulating LNCaP cells. LNCaP tumor incidence after s.c. injection was 100% (65 of 65) in SCID mice and 80% in athymic mice. No lymph node or distant metastases were observed with s.c. tumors, and RT-PCR for PSA transcripts was negative. Primary tumor incidence after intraprostatic injection was 89% (39 of 44) in SCID mice and 60% in athymic mice. In 10 SCID mice with primary tumors followed for 12 weeks, retroperitoneal or mediastinal lymph node metastases were found in 100%, and microscopic pulmonary metastases were identified in 40%. RT-PCR for PSA transcripts was positive in 3 of 10 mice tested. Serum PSA levels in mice with s.c. and intraprostatic tumors decreased by 65% to nadir levels at 7 and 4 days after castration, respectively. Serum PSA and LNCaP tumor PSA mRNA levels increased to precastration levels earlier in SCID mice with intraprostatic tumors compared to those with s.c. tumors. Intraprostatic injection of LNCaP cells in SCID mice provides a useful animal model to investigate mechanisms of metastasis and to evaluate therapies targeted toward inhibiting the metastatic cascade.


Assuntos
Metástase Linfática , Proteínas de Neoplasias/sangue , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/patologia , Neoplasias Retroperitoneais/secundário , Animais , Northern Blotting , Modelos Animais de Doenças , Humanos , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos SCID , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Orquiectomia , Reação em Cadeia da Polimerase , Antígeno Prostático Específico/genética , Neoplasias da Próstata/sangue , RNA Mensageiro/sangue , Neoplasias Retroperitoneais/sangue , Transplante Heterólogo , Células Tumorais Cultivadas
17.
J Steroid Biochem Mol Biol ; 59(5-6): 397-404, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9010345

RESUMO

The expression of 5alpha-reductase type 1 and type 2 isoenzymes in hyperplastic human prostate tissue and several human prostate cell lines was investigated by Northern blot analyses, reverse transcription-polymerase chain reaction (RT-PCR), and enzyme activity. Separation of stroma and epithelium was confirmed histologically and only preparations with no apparent contamination were employed in the subsequent studies. Poly(A)+ RNA was isolated from stromal and epithelial fractions and analysed by Northern blot and RT-PCR. Inhibition of epithelial and stromal 5alpha-reductase activities by 17beta-N,N-diethylcarbamoyl-4-methyl-4-aza-5alpha-androstan- 3-one (4MA) was assessed using a range of concentrations between 10(-13) and 10(-5) M. Results from Northern blot analyses and RT-PCR showed that the prostate stroma expressed 5alpha-reductase type 1 and type 2 isoenzymes, whereas the prostate epithelium only expressed 5alpha-reductase type 1. This was consistent with biphasic inhibition of 5alpha-reductase activity by 4MA in stroma and monophasic inhibition in epithelium. Cultured epithelial cells derived from human prostate only expressed 5alpha-reductase type 1 and had Vmax and Km values that approximated the lower end of the range reported for surgically removed prostate epithelium. The foregoing data explains the disparate activities of 5alpha-reductase, previously reported, in stroma and epithelium. The differential localization of these isoenzymes in the prostate suggests that future therapy of androgen-sensitive disease may be more successful through the use of selective inhibitors of the different 5alpha-reductase isoenzymes.


Assuntos
Oxirredutases/genética , Oxirredutases/metabolismo , Próstata/citologia , Próstata/metabolismo , Idoso , Antagonistas de Androgênios/farmacologia , Azasteroides/farmacologia , Northern Blotting , Colestenona 5 alfa-Redutase , Di-Hidrotestosterona/análogos & derivados , Di-Hidrotestosterona/farmacologia , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Epitélio/patologia , Regulação Enzimológica da Expressão Gênica , Humanos , Hiperplasia/enzimologia , Hiperplasia/genética , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Masculino , Pessoa de Meia-Idade , Oxirredutases/antagonistas & inibidores , Reação em Cadeia da Polimerase/métodos , Próstata/patologia , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Mensageiro/biossíntese , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/patologia , Células Tumorais Cultivadas
18.
J Steroid Biochem Mol Biol ; 59(5-6): 501-11, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9010356

RESUMO

The proportion of tumorigenic stem cells and the expression of the apoptosis-related gene, TRPM-2 (clusterin), were studied in populations of Shionogi carcinoma cells subjected to multiple cycles of androgen withdrawal and replacement (intermittent androgen suppression). The parent androgen-dependent cell line was initially transplanted into a male mouse which was castrated when the estimated weight of the resultant tumour became approximately 3 g. After the tumour had regressed to 40% or less of the original weight, it was transplanted into the next non-castrated male. This was repeated for four cycles of transplantation and castration-induced apoptosis before the tumour progressed to an androgen-independent state. The proportion of total stem cells in the tumour, as determined by in vivo limiting dilution assays in male mice, was constant during the first three cycles but increased 15-fold between the third and fourth cycles. In the parent androgen-dependent tumour before androgen ablation, the androgen-independent stem cell population formed 0.8% of the total stem cell compartment. After the fourth cycle this population increased to 47%; a population of similar size (33%, P = 0.8) was found in the androgen-independent recurrent form of the tumour induced by one-time castration. Whether androgen withdrawal therapy was intermittent or continuous, conversion to androgen independence thus occurred when one-third to one-half of the total stem cell compartment was populated by androgen-independent stem cells. The androgen-repressed TRPM-2 (clusterin) gene was actively expressed in regressing tumours after androgen ablation, and also became constitutively expressed in non-regressing tumours after the first and subsequent cycles of androgen withdrawal. Staining of cytoplasm and nuclei with anti-clusterin antibody was observed in androgen-dependent tumour cells after each cycle of intermittent androgen suppression; the nuclear staining was more intense in recurrent androgen-independent cells. The anomalous nuclear localization of clusterin, an anti-cytolytic TRPM-2 encoded protein, may serve to inhibit early events in the apoptotic process and thereby foster the generation and outgrowth of androgen-independent stem cells in an androgen-depleted environment.


Assuntos
Androgênios/metabolismo , Carcinoma/genética , Glicoproteínas/genética , Chaperonas Moleculares , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Animais , Anticorpos , Apoptose/genética , Carcinoma/tratamento farmacológico , Carcinoma/patologia , Divisão Celular , Clusterina , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/imunologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos , Transplante de Neoplasias , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Receptores de Glucocorticoides/genética , Supressão Genética , Testosterona/metabolismo , Testosterona/farmacologia
19.
J Steroid Biochem Mol Biol ; 59(3-4): 243-50, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9010316

RESUMO

An in vitro, cell-free transcription system, based on prostate-derived transcriptional machinery and very powerful androgen response elements (AREs), has been developed. Multiple (p(ARR3)LovTATA) AREs from the androgen-regulated probasin gene were linked to G-free cassettes and used in nuclear extracts prepared from prostate carcinoma cell lines (PC3 and LNCaP cells) to test specific induction of transcription by full-length AR and by glutathione-S-transferase (GST)-fusion peptides in which the androgen receptor (AR) DNA-binding domain alone (AR524-649), or together with the ligand-binding domain (AR524-902), or a portion of the NH2-terminal domain (AR232-649) were incorporated. In the presence of AR, nuclear extracts from PC3 cells had greater activity in supporting transcription than those from LNCaP cells; and lower background activity than those from HeLa cells. All of the AR forms correctly initiated in vitro transcription of ARE-templates in an androgen-independent manner. The amount of specific, inducible transcript was dependent on the concentration of AR peptide present. AR524-902 was the most potent transactivator tested, with the maximal level of specific transcript over 900-fold higher than the minimal level. At all concentrations this peptide was three to four times more active than either AR524-649 or AR232-649. In conclusion, we have developed a very specific and sensitive cell-free transcription system for delineating trans-activational regions of the AR.


Assuntos
Receptores Androgênicos/genética , Transcrição Gênica , Proteína de Ligação a Androgênios/genética , Animais , Carcinoma , Extratos Celulares , Núcleo Celular , Sistema Livre de Células , Escherichia coli/genética , Glutationa Transferase/genética , Células HeLa , Humanos , Masculino , Peptídeos , Neoplasias da Próstata , Ratos , Proteínas Recombinantes de Fusão , Sensibilidade e Especificidade , Ativação Transcricional , Células Tumorais Cultivadas
20.
Prog Urol ; 6(3): 375-85, 1996 Jun.
Artigo em Francês | MEDLINE | ID: mdl-8763692

RESUMO

Androgen-independent progression invariably occurs following castration of patients with prostate cancer. Animal model data suggest that androgen resistance may result from adaptive cell survival mechanisms activated by androgen withdrawal. If this is true, then re-exposure to, or low levels of, androgens may suppress or downregulate these mechanisms. The objective of this study is to determine whether intermittent androgen suppression (IAS) delays the onset of nonandrogen-regulated PSA production in the LNCaP prostate tumour model, when compared to continuous androgen suppression (CAS). Serum PSA levels correlate highly with LNCaP tumour volume and decrease rapidly following castration; beginning 3-4 weeks post-castration LN CaP tumours become androgen-independent with respect to PSA gene expression and produce PSA in amounts similar to precastrate state. In this study, IAS-treated mice were implanted with testosterone pellets beginning two weeks post-castration; cycles of testosterone replacement for 1 week and withdrawal for 2 weeks were repeated until serum PSA levels no longer returned to baseline. IAS therapy prolonged time to androgen-independent PSA production 3-fold, from an average of 26 days in the CAS group to 77 days in the IAS group. Serum and LNCaP tumour mRNA PSA levels remained below precastrate levels by 60 days post-castration in 75% of the IAS group, while serum PSA in all mice in the CAS group exceeded precastrate PSA by 28 days post-castration. Following castration, serum PSA levels increased 9-fold faster with CAS compared to IAS therapy. Observations using IAS in the LNCaP tumour model suggest that the onset of androgen-independent PSA gene regulation is prolonged 3-fold, perhaps due to androgen-induced differentiation and/or downregulation of androgen-suppressed gene expression.


Assuntos
Androgênios/biossíntese , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/sangue , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Orquiectomia , Antígeno Prostático Específico/análise , Antígeno Prostático Específico/genética , Neoplasias da Próstata/química , RNA Mensageiro/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA