Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 10(12): 1982-91, 2015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25818291

RESUMO

The role of ERß in prostate cancer is unclear, although loss of ERß is associated with aggressive disease. Given that mice deficient in ERß do not develop prostate cancer, we hypothesized that ERß loss occurs as a consequence of tumorigenesis caused by other oncogenic mechanisms and that its loss is necessary for tumorigenesis. In support of this hypothesis, we found that ERß is targeted for repression in prostate cancer caused by PTEN deletion and that loss of ERß is important for tumor formation. ERß transcription is repressed by BMI-1, which is induced by PTEN deletion and important for prostate tumorigenesis. This finding provides a mechanism for how ERß expression is regulated in prostate cancer. Repression of ERß contributes to tumorigenesis because it enables HIF-1/VEGF signaling that sustains BMI-1 expression. These data reveal a positive feedback loop that is activated in response to PTEN loss and sustains BMI-1.


Assuntos
Transformação Celular Neoplásica/metabolismo , Receptor beta de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias da Próstata/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Humanos , Masculino , Camundongos , Neoplasias da Próstata/genética , Transdução de Sinais/genética
2.
J Cell Physiol ; 228(7): 1601-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23359252

RESUMO

This study was carried out to dissect the mechanism by which ß1 integrins promote resistance to radiation. For this purpose, we conditionally ablated ß1 integrins in the prostatic epithelium of transgenic adenocarcinoma of mouse prostate (TRAMP) mice. The ability of ß1 to promote resistance to radiation was also analyzed by using an inhibitory antibody to ß1 , AIIB2, in a xenograft model. The role of ß1 integrins and of a ß1 downstream target, c-Jun amino-terminal kinase 1 (JNK1), in regulating radiation-induced apoptosis in vivo and in vitro was studied. We show that ß1 integrins promote prostate cancer (PrCa) progression and resistance to radiation in vivo. Mechanistically, ß1 integrins are shown here to suppress activation of JNK1 and, consequently apoptosis, in response to irradiation. Downregulation of JNK1 is necessary to preserve the effect of ß1 on resistance to radiation in vitro and in vivo. Finally, given the established crosstalk between ß1 integrins and type1 insulin-like growth factor receptor (IGF-IR), we analyzed the ability of IGF-IR to modulate ß1 integrin levels. We report that IGF-IR regulates the expression of ß1 integrins, which in turn confer resistance to radiation in PrCa cells. In conclusion, this study demonstrates that ß1 integrins mediate resistance to ionizing radiation through inhibition of JNK1 activation.


Assuntos
Integrina beta1/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/radioterapia , Tolerância a Radiação/fisiologia , Animais , Apoptose/fisiologia , Apoptose/efeitos da radiação , Caspase 3/metabolismo , Linhagem Celular Tumoral , Humanos , Integrina beta1/genética , Masculino , Camundongos , Camundongos Knockout , Camundongos Nus , Camundongos Transgênicos , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Neoplasias da Próstata/patologia , RNA Interferente Pequeno/genética , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Transplante Heterólogo
3.
Cancer Discov ; 2(10): 906-21, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22777769

RESUMO

We show that the VEGF receptor neuropilin-2 (NRP2) is associated with high-grade, PTEN-null prostate cancer and that its expression in tumor cells is induced by PTEN loss as a consequence of c-Jun activation. VEGF/NRP2 signaling represses insulin-like growth factor-1 receptor (IGF-IR) expression and signaling, and the mechanism involves Bmi-1-mediated transcriptional repression of the IGF-IR. This mechanism has significant functional and therapeutic implications that were evaluated. IGF-IR expression positively correlates with PTEN and inversely correlates with NRP2 in prostate tumors. NRP2 is a robust biomarker for predicting response to IGF-IR therapy because prostate carcinomas that express NRP2 exhibit low levels of IGF-IR. Conversely, targeting NRP2 is only modestly effective because NRP2 inhibition induces compensatory IGF-IR signaling. Inhibition of both NRP2 and IGF-IR, however, completely blocks tumor growth in vivo.


Assuntos
Neuropilina-2/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Neoplasias da Próstata/metabolismo , Receptor IGF Tipo 1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Complexo Repressor Polycomb 1/genética , Neoplasias da Próstata/genética , Interferência de RNA , RNA Interferente Pequeno , Receptor IGF Tipo 1/genética , Transdução de Sinais/genética , Transcrição Gênica , Ativação Transcricional , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/genética
4.
Cancer Cell ; 17(4): 319-32, 2010 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-20385358

RESUMO

High Gleason grade prostate carcinomas are aggressive, poorly differentiated tumors that exhibit diminished estrogen receptor beta (ERbeta) expression. We report that a key function of ERbeta and its specific ligand 5alpha-androstane-3beta,17beta-diol (3beta-adiol) is to maintain an epithelial phenotype and repress mesenchymal characteristics in prostate carcinoma. Stimuli (TGF-beta and hypoxia) that induce an epithelial-mesenchymal transition (EMT) diminish ERbeta expression, and loss of ERbeta is sufficient to promote an EMT. The mechanism involves ERbeta-mediated destabilization of HIF-1alpha and transcriptional repression of VEGF-A. The VEGF-A receptor neuropilin-1 drives the EMT by promoting Snail1 nuclear localization. Importantly, this mechanism is manifested in high Gleason grade cancers, which exhibit significantly more HIF-1alpha and VEGF expression, and Snail1 nuclear localization compared to low Gleason grade cancers.


Assuntos
Receptor beta de Estrogênio/uso terapêutico , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Neoplasias da Próstata/prevenção & controle , Fatores de Transcrição/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Receptor beta de Estrogênio/fisiologia , Humanos , Masculino , Mesoderma/citologia , Mesoderma/fisiologia , Neoplasias da Próstata/patologia , Fatores de Transcrição da Família Snail , Fator de Crescimento Transformador beta/fisiologia
5.
Am J Pathol ; 176(1): 393-401, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19948822

RESUMO

Molecular chaperones of the heat shock protein-90 (Hsp90) family promote cell survival, but the molecular requirements of this pathway in tumor progression are not understood. Here, we show that a mitochondria-localized Hsp90 chaperone, tumor necrosis factor receptor-associated protein-1 (TRAP-1), is abundantly and ubiquitously expressed in human high-grade prostatic intraepithelial neoplasia, Gleason grades 3 through 5 prostatic adenocarcinomas, and metastatic prostate cancer, but largely undetectable in normal prostate or benign prostatic hyperplasia in vivo. Prostate lesions formed in genetic models of the disease, including the transgenic adenocarcinoma of the mouse prostate and mice carrying prostate-specific deletion of the phosphatase tensin homolog tumor suppressor (Pten(pc-/-)), also exhibit high levels of TRAP-1. Expression of TRAP-1 in nontransformed prostatic epithelial BPH-1 cells inhibited cell death, whereas silencing of TRAP-1 in androgen-independent PC3 or DU145 prostate cancer cells by small interfering RNA enhanced apoptosis. Targeting TRAP-1 with a novel class of mitochondria-directed Hsp90 inhibitors, ie, Gamitrinibs, caused rapid and complete killing of androgen-dependent or -independent prostate cancer, but not BPH-1 cells, whereas reintroduction of TRAP-1 in BPH-1 cells conferred sensitivity to Gamitrinib-induced cell death. These data identify TRAP-1 as a novel mitochondrial survival factor differentially expressed in localized and metastatic prostate cancer compared with normal prostate. Targeting this pathway with Gamitrinibs could be explored as novel molecular therapy in patients with advanced prostate cancer.


Assuntos
Citoproteção , Proteínas de Choque Térmico HSP90/metabolismo , Mitocôndrias/metabolismo , Chaperonas Moleculares/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Idoso , Animais , Morte Celular , Sobrevivência Celular , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais , Epitélio/metabolismo , Epitélio/patologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Metástase Neoplásica
6.
J Cell Biochem ; 107(5): 845-52, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19492418

RESUMO

Although the timing with which common epithelial malignancies arise and become established remains a matter of debate, it is clear that by the time they are detected these tumors harbor hundreds of deregulated, aberrantly expressed or mutated genes. This enormous complexity poses formidable challenges to identify gene pathways that are drivers of tumorigenesis, potentially suitable for therapeutic intervention. An alternative approach is to consider cancer pathways as interconnected networks, and search for potential nodal proteins capable of connecting multiple signaling networks of tumor maintenance. We have modeled this approach in advanced prostate cancer, a condition with current limited therapeutic options. We propose that the integration of three signaling networks, including chaperone-mediated mitochondrial homeostasis, integrin-dependent cell signaling, and Runx2-regulated gene expression in the metastatic bone microenvironment plays a critical role in prostate cancer maintenance, and offers novel options for molecular therapy.


Assuntos
Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Neoplasias da Próstata/genética , Descoberta de Drogas , Humanos , Integrina alfaV/metabolismo , Masculino , Mitocôndrias/metabolismo , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Transdução de Sinais
7.
Cancer Res ; 69(12): 4954-8, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19470765

RESUMO

Endogenous tumor suppression provides a barrier against oncogenesis, but the molecular requirements of this process are not well understood. Here, we show that the dual specificity phosphatase PTEN, a gene almost universally altered in human tumors, silences the expression of survivin, an essential regulator of cell division and apoptosis in cancer. This pathway is independent of p53, involves active repression of survivin gene transcription, and is mediated by direct occupancy of the survivin promoter by FOXO1 and FOXO3a factors. Conditional deletion of PTEN in the mouse prostate causes deregulated induction of survivin before full-blown transformation in vivo, whereas expression of survivin and PTEN is inversely correlated in cancer patients. Therefore, silencing the survivin gene is an essential requirement of endogenous PTEN tumor suppression.


Assuntos
Inativação Gênica , Proteínas Associadas aos Microtúbulos/genética , PTEN Fosfo-Hidrolase/fisiologia , Sequência de Bases , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Primers do DNA , Fatores de Transcrição Forkhead/fisiologia , Genes Supressores de Tumor , Humanos , Proteínas Inibidoras de Apoptose , PTEN Fosfo-Hidrolase/genética , Regiões Promotoras Genéticas , Survivina , Transcrição Gênica
8.
PLoS Genet ; 5(1): e1000334, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19148275

RESUMO

Alpha-methylacyl-coenzyme A racemase (AMACR) regulates peroxisomal beta-oxidation of phytol-derived, branched-chain fatty acids from red meat and dairy products -- suspected risk factors for colon carcinoma (CCa). AMACR was first found overexpressed in prostate cancer but not in benign glands and is now an established diagnostic marker for prostate cancer. Aberrant expression of AMACR was recently reported in Cca; however, little is known about how this gene is abnormally activated in cancer. By using a panel of immunostained-laser-capture-microdissected clinical samples comprising the entire colon adenoma-carcinoma sequence, we show that deregulation of AMACR during colon carcinogenesis involves two nonrandom events, resulting in the mutually exclusive existence of double-deletion at CG3 and CG10 and deletion of CG12-16 in a newly identified CpG island within the core promoter of AMACR. The double-deletion at CG3 and CG10 was found to be a somatic lesion. It existed in histologically normal colonic glands and tubular adenomas with low AMACR expression and was absent in villous adenomas and all CCas expressing variable levels of AMACR. In contrast, deletion of CG12-16 was shown to be a constitutional allele with a frequency of 43% in a general population. Its prevalence reached 89% in moderately differentiated CCas strongly expressing AMACR but only existed at 14% in poorly differentiated CCas expressing little or no AMACR. The DNA sequences housing these deletions were found to be putative cis-regulatory elements for Sp1 at CG3 and CG10, and ZNF202 at CG12-16. Chromatin immunoprecipitation, siRNA knockdown, gel shift assay, ectopic expression, and promoter analyses supported the regulation by Sp1 and ZNF202 of AMACR gene expression in an opposite manner. Our findings identified key in vivo events and novel transcription factors responsible for AMACR regulation in CCas and suggested these AMACR deletions may have diagnostic/prognostic value for colon carcinogenesis.


Assuntos
Colo/enzimologia , Neoplasias do Colo/genética , Ilhas de CpG/genética , Regulação Neoplásica da Expressão Gênica , Regiões Promotoras Genéticas , Racemases e Epimerases/genética , Adenoma Viloso/genética , Adenoma Viloso/metabolismo , Adenoma Viloso/patologia , Sequência de Bases , Sítios de Ligação , Diferenciação Celular , Linhagem Celular Tumoral , Colo/metabolismo , Colo/patologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Humanos , Dados de Sequência Molecular , Polimorfismo Genético , Racemases e Epimerases/metabolismo , Proteínas Repressoras/metabolismo , Deleção de Sequência/genética , Transcrição Gênica
9.
Cancer Res ; 68(6): 1625-30, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18339840

RESUMO

SOX9 is a transcription factor that plays a critical role in the development of multiple tissues. We previously reported that SOX9 in normal human adult prostate was restricted to basal epithelium. SOX9 was also expressed in a subset of prostate cancer (PCa) cells and was increased in relapsed hormone-refractory PCa. Moreover, SOX9 expression in PCa cell lines enhanced tumor cell proliferation and was beta-catenin regulated. Here we report additional in vivo results showing that SOX9 is highly expressed during fetal prostate development by epithelial cells expanding into the mesenchyme, suggesting it may contribute to invasive growth in PCa. Indeed, SOX9 overexpression in LNCaP PCa xenografts enhanced growth, angiogenesis, and invasion. Conversely, short hairpin RNA-mediated SOX9 suppression inhibited the growth of CWR22Rv1 PCa xenografts. These results support important functions of SOX9 in both the development and maintenance of normal prostate, and indicate that these functions contribute to PCa tumor growth and invasion.


Assuntos
Proteínas de Grupo de Alta Mobilidade/biossíntese , Próstata/embriologia , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Fatores de Transcrição/biossíntese , Animais , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Regulação para Baixo , Células Epiteliais/metabolismo , Proteínas de Grupo de Alta Mobilidade/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos SCID , Invasividade Neoplásica , Neovascularização Patológica/metabolismo , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Interferente Pequeno/genética , Fatores de Transcrição SOX9 , Fatores de Transcrição/genética
10.
J Cell Physiol ; 214(1): 56-64, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17541940

RESUMO

Bone morphogenetic proteins (BMPs) play pivotal roles in bone and cartilage growth and repair. Through phenotypes of short-ear (se) mice, which have BMP-5 mutations, a role for BMP-5 in some specific aspects of skeletogenesis and cartilage growth is known. This report examines BMP-5 expression in the growth plate and in differentiating cultures of primary chondrocytes, and the effects of addition of BMP-5 or its inhibition by anti-BMP-5 antibody in chondrocyte cultures. By laser capture microdissection and immunohistochemistry, we found that BMP-5 is expressed in proliferating zone (PZ) chondrocytes and that the expression increases sharply with hypertrophic differentiation. A similar pattern was observed in differentiating cultures of primary chondrocytes, with BMP-5 expression increasing as cells differentiated, in contrast to other BMPs. BMP-5 added to cultures increased cell proliferation early in the culture period and also stimulated cartilage matrix synthesis. Also, BMP-5 addition to the cultures activated phosphorylation of Smad 1/5/8 and p38 MAP kinase and caused increased nuclear accumulation of phospho-Smads. Anti-BMP-5 antibody inhibited the endogenous BMP-5, reducing cell proliferation and phospho-Smad nuclear accumulation. Together, the results demonstrate that BMP-5 is normally an important regulator of chondrocyte proliferation and differentiation. Whether other BMPs may compensate in BMP-5 loss-of-function mutations is discussed.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/fisiologia , Proliferação de Células , Condrócitos/metabolismo , Matriz Extracelular/metabolismo , Animais , Proteína Morfogenética Óssea 5 , Calcificação Fisiológica/fisiologia , Cartilagem Articular/citologia , Cartilagem Articular/enzimologia , Células Cultivadas , Condrócitos/citologia , Glicosaminoglicanos/análise , Imuno-Histoquímica , Técnicas In Vitro , Ratos , Costelas/citologia
11.
Am J Pathol ; 171(4): 1334-41, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17717140

RESUMO

Oxidative and nitrosative stress have been implicated in prostate carcinogenesis, but the cause(s) of redox imbalance in the gland remains poorly defined. We and others have reported that administration of testosterone plus 17beta-estradiol to Noble rats for 16 weeks induces dysplasia and stromal inflammation of the lateral prostate (LP) but not the ventral prostate. Here, using laser capture microdissected specimens, we found that the combined hormone regimen increased the expression of mRNA of specific members of NAD(P)H oxidase (NOX-1, NOX-2, and NOX4), nitric-oxide synthase [NOS; inducible NOS and endothelial NOS], and cyclooxygenase (COX-2) in the LP epithelium and/or its adjacent inflammatory stroma. Accompanying these changes was the accumulation of 8-hydroxy-2'-deoxyguanosine, 4-hydroxynonenal protein adducts, and nitrotyrosine, primarily in the LP epithelium, suggesting that NOX, NOS, and COX may mediate hormone-induced oxidative/nitrosative stress in epithelium. We concluded that the oxidative/nitrosative damage resulting from the testosterone-plus-17beta-estradiol treatment is not solely derived from stromal inflammatory lesions but likely also originates from the epithelium per se. In this context, the up-regulation of COX-2 from epithelium represents a potential mechanism by which the hormone-initiated epithelium might induce inflammatory responses. Thus, we link alterations in the hormonal milieu with oxidative/nitrosative/inflammatory damage to the prostate epithelium that promotes carcinogenesis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Estradiol/metabolismo , Estresse Oxidativo , Neoplasias da Próstata/etiologia , Testosterona/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Animais , Transformação Celular Neoplásica/induzido quimicamente , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análise , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Estradiol/administração & dosagem , Estradiol/toxicidade , Hormônios Esteroides Gonadais/administração & dosagem , Hormônios Esteroides Gonadais/metabolismo , Hormônios Esteroides Gonadais/toxicidade , Lasers , Masculino , Microdissecção , NADH NADPH Oxirredutases/genética , NADH NADPH Oxirredutases/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Oxirredução , Próstata , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos , Testosterona/administração & dosagem , Testosterona/toxicidade
12.
Cancer Res ; 66(16): 8076-82, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16912184

RESUMO

The Ini1 subunit of the SWI/SNF chromatin remodeling complex suppresses formation of malignant rhabdoid tumors in humans and mice. Transduction of Ini1 into Ini1-deficient tumor-derived cell lines has indicated that Ini1 arrests cell growth, controls chromosomal ploidy, and suppresses tumorigenesis by regulating components of the retinoblastoma (Rb) signaling pathway. Furthermore, conditional inactivation of Ini1 in mouse fibroblasts alters the expression of various Rb-E2F-regulated genes, indicating that endogenous Ini1 levels may control Rb signaling in cells. We have reported previously that loss of one allele of Ini1 in mouse fibroblasts results only in a 15% to 20% reduction in total Ini1 mRNA levels due to transcriptional compensation by the remaining Ini1 allele. Here, we examine the effects of Ini1 haploinsufficiency on cell growth and immortalization in mouse embryonic fibroblasts. In addition, we examine pituitary tumorigenesis in Rb-Ini1 compound heterozygous mice. Our results reveal that heterozygosity for Ini1 up-regulates cell growth and immortalization and that exogenous Ini1 down-regulates the growth of primary cells in a Rb-dependent manner. Furthermore, loss of Ini1 is redundant with loss of Rb function in the formation of pituitary tumors in Rb heterozygous mice and leads to the formation of large, atypical Rb(+/-) tumor cells lacking adrenocorticotropic hormone expression. These results confirm in vivo the relationship between Rb and Ini1 in tumor suppression and indicate that Ini1 plays a role in maintaining the morphologic and functional differentiation of corticotrophic cells.


Assuntos
Divisão Celular/fisiologia , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Neoplasias Hipofisárias/patologia , Proteína do Retinoblastoma/fisiologia , Fatores de Transcrição/genética , Animais , Proteínas Cromossômicas não Histona/deficiência , Primers do DNA , Proteínas de Ligação a DNA/deficiência , Embrião de Mamíferos , Fibroblastos/citologia , Fibroblastos/fisiologia , Genótipo , Humanos , Camundongos , Proteína do Retinoblastoma/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína SMARCB1 , Fatores de Transcrição/deficiência
13.
Am J Pathol ; 164(6): 2003-12, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15161636

RESUMO

Estrogen receptor (ER)-beta is thought to exert anti-proliferative effects in the normal prostate but supports prostate cancer (PCa) cell survival. We previously reported that the receptor's expression declined as PCa developed in the gland but reappeared in lymph node and bone metastases. To investigate whether hypermethylation was the underlying mechanism for these phenomena, we first identified two CpG islands (CGIs) encompassing 41 CpG dinucleotides, located separately in the untranslated exon 0N and the promoter region of ER-beta. Using immunostained, laser capture-microdissected samples from 56 clinical specimens, we demonstrated an inverse relationship exists between the extent of ER-beta CGI methylation and receptor expression in normal, hyperplastic, premalignant, and malignant foci of the prostate and in lymph node and bone metastases. Treatment of PCa cell lines (LNCaP and DU145), that express little ER-beta mRNA, with a demethylating agent increased levels of receptor expression thus corroborating our in vivo findings that methylation is involved in ER-beta silencing. Methylation centers in the promoter region and exon 0N were identified by hierarchical cluster analysis of bisulfite sequencing data obtained from 710 alleles. Methylation at these centers was insignificant in normal epithelium, reached 80 to 90% in grade 4/5 PCa, but declined to less than 20% in bone metastases. In addition, progressive methylation spreading from the exonic CGI to the promoter CGI, which correlated with loss of ER-beta expression, was detected in microdissected samples and in cell cultures. Using a new class of methylated oligonucleotides that mediate sequence-specific methylation in cellulo, we demonstrated that methylation of the promoter CGI, but not the exonic CGIs, led to transcriptional inactivation of ER-beta. Our results present the first evidence that epigenetic regulation of ER-beta is a reversible and tumor stage-specific process and that gene silencing via methylated oligonucleotides may have therapeutic potential in the treatment of advanced PCa.


Assuntos
Metilação de DNA , Neoplasias da Próstata/patologia , Receptores de Estrogênio/genética , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Fosfatos de Dinucleosídeos/análise , Receptor beta de Estrogênio , Humanos , Masculino , Dados de Sequência Molecular , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Estadiamento de Neoplasias , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , Neoplasias da Próstata/cirurgia , TATA Box
14.
Clin Cancer Res ; 9(12): 4475-82, 2003 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-14555521

RESUMO

PURPOSE: Estrogen exposure has been linked to a risk for the development of testicular germ cell cancers. The effects of estrogen are now known to be mediated by estrogen receptor (ER)-alpha and -beta receptor subtypes, but only ER-beta has been found in human normal testis. The goal of the present study was to compare the localization and expression levels of these ER subtypes in testicular germ cell cancers (seminomas and nonseminomatous germ cell tumors) with normal testis. For completeness, expression of androgen and progesterone receptors was also investigated. EXPERIMENTAL DESIGN: Immunohistochemistry was used to localize the expression of steroid receptors in 39 archival testicular germ cell cancers and 5 morphologically normal testes. Expression of the steroid receptors at the transcript level was semiquantified by reverse transcription-PCR in 5 paired fresh-frozen specimens of normal and neoplastic testes. RESULTS: ER-alpha was not expressed in the human normal testis. It was also absent in all of the testicular germ cell cancers studied. In contrast, ER-beta was strongly expressed in various germ cells of the normal testis. However, its expression was markedly diminished in seminomas, embryonal cell carcinomas, and in mixed germ cell tumors, at both transcriptional and translational levels. In contrast, ER-beta remained highly expressed in endodermal sinus tumors and teratomas. Progesterone receptor, an estrogen-regulated gene, was localized to spermatagonia of the normal testis, but its expression dramatically reduced in seminomas. With the exception of spermatagonia, androgen receptor was found in all of the germ cells of the normal testis, but, aside from trace staining in 3 of 5 endodermal sinus tumor cells, it was not detected immunohistochemically in any other germ cell cancer. CONCLUSIONS: We confirm expression of ER-beta, but not ER-alpha, in normal testicular cells, suggesting that only the former ER subtype mediates the action of estrogen in the human male gonad. Our results provide the first evidence that only ER-beta is expressed in testicular germ cell tumors. Its expression is down-regulated in seminomas and embryonal cell carcinomas but remains high in endodermal sinus tumors and in teratomas. The observed differences in ER-beta expression levels among different testicular germ cell tumors may reflect divergent pathways of differentiation/dedifferentiation of these neoplasms from a common precursor. Collectively, these findings provide a possible mechanistic link between estrogen exposure and testicular cancer risk.


Assuntos
Germinoma/metabolismo , Receptores de Estrogênio/metabolismo , Neoplasias Testiculares/metabolismo , Adulto , Carcinoma Embrionário/genética , Carcinoma Embrionário/metabolismo , Carcinoma Embrionário/patologia , Diferenciação Celular , Tumor do Seio Endodérmico/genética , Tumor do Seio Endodérmico/metabolismo , Tumor do Seio Endodérmico/patologia , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Germinoma/genética , Germinoma/patologia , Humanos , Técnicas Imunoenzimáticas , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/genética , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Túbulos Seminíferos/metabolismo , Seminoma/genética , Seminoma/metabolismo , Seminoma/patologia , Teratoma/genética , Teratoma/metabolismo , Teratoma/patologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia
15.
Cancer Epidemiol Biomarkers Prev ; 12(8): 775-83, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12917210

RESUMO

An enzyme previously identified as alpha-methylacyl-CoA racemase (AMACR) is overexpressed in high-grade prostatic intraepithelial neoplasia and in a majority (60-100%) of prostate cancers (CaPs) as compared with normal and benign hyperplastic lesions of the prostate, where it is minimally expressed. This enzyme is required for the beta-oxidation of branched-chain fatty acids, which include phytanic acid and its alpha-oxidation product, pristanic acid. Interestingly, there is an established correlation between CaP risk and the consumption of dairy and beef products, which also contain marked quantities of these two phytols. In this context, it has also been reported that sex steroids influence lipogenesis through the induction of fatty acid synthase in CaP-derived cell lines and CaP tissues. These findings indicate a potential role for AMACR and the possible influence of sex steroids in both the early development and subsequent progression of CaP. Despite the recent interest in AMACR as a histological marker for CaP, little is known about the regulation of this enzyme and its role in CaP development. To identify potential AMACR-regulating factors, we treated LNCaP cells (an androgen-responsive CaP-derived cell line) and NPrEC cells (a normal prostate basal epithelial cell line) with increasing concentrations of pristanic acid, phytanic acid, 5alpha-dihydrotestosterone, and 17beta-estradiol. Neither the biologically potent androgen 5alpha-dihydrotestosterone nor 17beta-estradiol had any apparent effect on AMACR expression at the protein or transcriptional levels in either cell line. Conversely, pristanic acid and, to a much lesser extent, phytanic acid markedly increased AMACR protein levels selectively in the LNCaP cell line, but not the NPrEC cell line. However, no change was measured at the transcriptional level in either cell line. AMACR is therefore significantly increased at the protein level in CaP cells, through what appears to be the stabilizing effect of the same fatty acids that are present at appreciable concentrations in beef and dairy products, which have been associated with CaP risk. Our findings therefore provide a link between the consumption of dietary fatty acids and the enhanced expression of AMACR, an enzyme that may play an important role in genesis and progression of CaP.


Assuntos
Laticínios/análise , Ácidos Graxos/farmacologia , Produtos da Carne/análise , Neoplasias da Próstata/enzimologia , Racemases e Epimerases/análise , 5-alfa-Di-Hidroprogesterona/farmacologia , Animais , Bovinos , Estradiol/farmacologia , Ácidos Graxos/química , Humanos , Masculino , Ácido Fitânico/farmacologia , Racemases e Epimerases/fisiologia , Células Tumorais Cultivadas
16.
Hum Pathol ; 34(3): 228-33, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12673556

RESUMO

Carcinomas of the transition zone (TZ) constitute approximately 20% of all prostate cancers. The TZ is the site of origin of grade 1 and grade 2 cancers, the most well-differentiated of the Gleason grade tumors, as well as for benign prostatic hyperplasia (BPH). In this regard, grade 1 carcinoma has architectural features that closely mimic gland-rich BPH nodules. Although a relationship between cancers arising in this zone and BPH has been suspected, such an association remains undefined. To gain insight into the origin, development, and progression of cancers arising in the TZ, we used a highly specific rabbit monoclonal antibody (P504S) directed against alpha-methylacyl-CoA racemase (AMACR) to study the expression of the enzyme in 25 cases of evolving and fully developed carcinomas of this zone. AMACR has been proposed as a new molecular marker for prostate cancer, because the enzyme is reportedly overexpressed in high-grade dysplasias, also termed prostatic intraepithelial neoplasia, a purported precursor of prostatic carcinoma, and in all grades of prostatic carcinoma of the peripheral zone. Using P504S, P63, or antikeratin 34beta E12 antibodies, we found it possible to define areas of transition from hyperplasia to carcinoma in 6 BPH nodules. In 3 other cancer-containing BPH nodules, staining for AMCAR was observed in benign hyperplastic glands that were juxtaposed to carcinoma. Enzyme expression was also evident in 5 additional cases in which BPH was found adjacent to cancer. In contrast; AMACR was not visualized in any other BPH nodules that we studied. Thus, using the enzyme as a marker, we document for the first time that some carcinomas of the TZ arise from an AMCAR-positive transition lesion within a subset of BPH nodules. Moreover, the finding of enhanced AMACR expression in benign glands within cancer-containing nodules as well as in BPH lesions adjacent to carcinoma suggests that in some cases, up-regulation of the enzyme may precede morphological evidence of neoplastic transformation. AMACR was lightly expressed in transition lesions and grade 1 carcinomas but more strongly expressed in higher-grade TZ cancers, suggesting that enzyme expression is enhanced with progression in this zone. Because AMACR is involved in the beta oxidation of branched fatty acids and their derivatives, enhanced expression of the enzyme in evolving carcinomas in BPH nodules, as well as its up-regulation in juxtaposed morphologically benign glands and grade 1 carcinomas, suggests that increased utilization of fatty acids may play an important role in carcinoma development and progression in the TZ.


Assuntos
Hiperplasia Prostática/enzimologia , Neoplasias da Próstata/enzimologia , Racemases e Epimerases/análise , Anticorpos Monoclonais , Biomarcadores Tumorais/análise , Humanos , Imuno-Histoquímica , Masculino , Hiperplasia Prostática/patologia , Neoplasia Prostática Intraepitelial/enzimologia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia
17.
Prostate ; 52(1): 69-81, 2002 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11992621

RESUMO

BACKGROUND: Although androgens have long been implicated in the development, regulation, and pathophysiology of the prostate, evidence suggests that estrogens may also affect these processes. Specifically, estrogens have been shown to influence the development of the fetal and neonatal rodent prostate and to induce a pathognomonic change, termed squamous metaplasia, in the developing and adult prostate. Studies have been inconclusive, however, as to whether estrogens enhance or restrain the growth of the gland. Although the fetal rodent prostate has been reported to contain both estrogen receptor alpha (ER-alpha) and beta (ER-beta), there have been no reports as to whether either of the ER subtypes is expressed in the developing human prostate. METHODS: In the present study, we used a novel antibody, directed against a unique sequence in the F domain of ER-beta, and laser capture microdissection/reverse transcriptase-polymerase chain reaction to study the expression of the receptor in the fetal, neonatal, and prepubertal human prostate. Results were compared with the expression of ER-alpha, androgen receptor (AR), prostatic acid phosphatase (PAP), prostate specific antigen (PSA), high molecular weight cytokeratin (HMCK), and the proliferative marker Ki67. RESULTS: For the first time, we report that ER-beta is the only estrogen receptor detected at the protein level in the morphologically normal developing human fetal prostate. By midgestation, strong immunostaining for ER-beta was detected in the nuclei of nearly 100% of epithelial and in the majority of stromal cells. This pattern of expression was evident in the fetal, neonatal, and early prepubertal prostate. However, by 11 years postnatal, staining for the receptor became restricted primarily to the basal epithelial and stromal compartments, a pattern analogous to that observed in the normal adult gland. ER-alpha mRNA was present in microdissected stroma of the fetal gland. Although ER-alpha was not immunodetected in any morphologically normal fetal epithelial or stromal cells, weak staining for the receptor, however, was found in some examples of squamous metaplasia, suggesting the role of alpha-subtype in this lesion. ER-alpha was clearly visualized immunohistochemically at 1 month of postnatal development where it was then localized exclusively in periacinar stromal nuclei, which suggests that it may exert paracrine influences on further prostatic glandular development. Interestingly, the expression of ER-beta early in prostatic development occurred coincident with both the increasing rate of epithelial cell proliferation, observed in the first half of gestation, and the reported high levels of estrogen in the gland from midgestation until term. Paradoxically, however, staining for the receptor remained intense, despite the dramatic decrease in Ki67 labeling observed in the second half of gestation. CONCLUSION: Our results indicate that the effects of estrogens on the growth of the human fetal prostate are mediated primarily by ER-beta but that ER-alpha contributes to postnatal glandular development. Furthermore, these results suggest that ER-beta, possibly in concert with androgens, may mediate diverse effects on prostate epithelial proliferation by first promoting cell expansion early in gestation, and then acting to limit growth later in prostatic development.


Assuntos
Expressão Gênica , Próstata/embriologia , Próstata/crescimento & desenvolvimento , Receptores de Estrogênio/genética , Fosfatase Ácida , Criança , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Doenças Fetais/metabolismo , Idade Gestacional , Humanos , Imuno-Histoquímica , Lactente , Recém-Nascido , Queratinas/análise , Antígeno Ki-67/análise , Masculino , Metaplasia/metabolismo , Peso Molecular , Próstata/química , Próstata/patologia , Antígeno Prostático Específico/análise , Proteínas Tirosina Fosfatases/análise , Receptores Androgênicos/análise , Receptores de Estrogênio/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Endocrinology ; 143(6): 2093-105, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12021174

RESUMO

We previously demonstrated that 1) treatment of Noble rats for 16 wk with testosterone (T) and estradiol-17 beta (E2) led to 100% incidence of dorsolateral prostate (DLP) dysplasia and hyperprolactinemia and 2) blockade of PRL release with bromocriptine cotreatment significantly lowered the incidence of DLP dysplasia. In the current study, we sought to determine whether E2 exerts direct effects, independent of PRL, in this model system. The pure antiestrogen ICI 182,780 (ICI), reported to have no effect on PRL release in female rats, was administered biweekly to T + E2-treated rats at 3 mg/kg body weight. ICI cotreatment completely prevented DLP dysplasia development but it also blocked hyperprolactinemia in the dual hormone-treated rats. Gene profiling with an 1185 gene rat cDNA array identified approximately 100 genes displaying > or = 3-fold changes in rat lateral prostates (LPs) following T + E2 treatment. Significantly more genes were up-regulated (77) than down-regulated (14), reflecting cellular/molecular changes associated with enhanced cell proliferation, DNA damage, heightened protein and RNA synthesis, increased energy metabolism, and activation of several proto-oncogenes and intracellular signaling pathways. Post hoc analyses, using quantitative real-time RT-PCR, corroborated differential expression of eight genes, exhibiting three different patterns of altered expression. Genes encoding the early growth response protein 1 and metalloendopeptidase meprin beta-subunit were similarly altered in T + E2- and T + E2 + ICI-treated animals when compared with untreated controls. In contrast, transcripts of fos-related antigen-2, growth arrest and DNA damage-inducible protein-45, and signal transducer and activator of transcription-3 were significantly increased in the LPs of T + E2-treated animals, but the increases were reversed by cotreatment with ICI. Differential expression of fos-related antigen-2 and growth arrest and DNA damage-inducible protein-45 were further confirmed at the protein level by immunohistochemistry. Lastly, levels of A-RAF, VIP-1 receptor, and calpastatin mRNA were distinctly lessen in rat LPs under T + E2 influence, but rebound with ICI cotreatment. In conclusion, our findings further implicated pituitary PRL in the induction of dysplasia in rat LP. Gene profiling provided clues that molecular events related to enhancement of cell proliferation, DNA damage, and activation of proto-oncogenes and transforming factors may be causally linked to the genesis of LP dysplasia in this rat model.


Assuntos
Proteínas de Arabidopsis , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Imediatamente Precoces , Hiperplasia Prostática/genética , Testosterona , Adenosina Trifosfatases , Animais , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteína 1 de Resposta de Crescimento Precoce , Fulvestranto , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Katanina , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Plantas/biossíntese , Proteínas de Plantas/genética , Hiperplasia Prostática/induzido quimicamente , Hiperplasia Prostática/prevenção & controle , Biossíntese de Proteínas , Proteínas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Radioimunoensaio , Ratos , Receptores de Peptídeo Intestinal Vasoativo/biossíntese , Receptores de Peptídeo Intestinal Vasoativo/genética , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3 , Transativadores/biossíntese , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...