Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Oncogene ; 25(4): 579-87, 2006 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-16186802

RESUMO

Although the transcription factor USF2 has been implicated in the regulation of cellular growth and proliferation, it is unknown whether alterations in USF2 contribute to tumorigenesis and tumor development. We examined the role of USF2 in prostate tumorigenesis. Western blot analysis revealed markedly decreased USF2 levels in three androgen-independent prostate cancer cell lines, PC-3, DU145, and M12, as compared to nontumorigenic prostate epithelial cells or the androgen-dependent cell line, LNCaP. Ectopic expression of USF2 in PC-3 cells did not affect the cell proliferation rate of PC-3 cells on plastic surfaces. However, it dramatically decreased anchorage-independent growth of PC-3 cells in soft agar (90-98% inhibition) and the invasion capability (80% inhibition) of PC-3 cells in matrix gel assay. Importantly, expression of USF2 in PC-3 cells inhibited the tumorigenicity of PC-3 cells in an in vivo nude mice xenograft model (80-90% inhibition). These results suggest that USF2 has tumor-suppression function. Consistent with its function in tumor suppression, we found that the USF2 protein is present in normal prostate epithelial cells but absent in 18 of 42 (43%) human prostate cancer tissues (P = 0.015). To further examine the functional role of USF2 in vivo, we generated mice with genetic deletion of USF2 gene. We found that USF2-null mice displayed marked prostate hyperplasia at a young age, suggesting that USF2 is involved in the normal growth and differentiation of prostate. Together, these studies demonstrate that USF2 has tumor-suppressor function and plays a role in prostate carcinogenesis.


Assuntos
Neoplasias da Próstata/prevenção & controle , Fatores Estimuladores Upstream/fisiologia , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Humanos , Masculino , Camundongos , Hiperplasia Prostática/etiologia , Neoplasias da Próstata/química , Fatores Estimuladores Upstream/análise , Fatores Estimuladores Upstream/genética
2.
Prostate ; 66(2): 218-25, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16173037

RESUMO

BACKGROUND: Selenium compounds have been shown to induce apoptosis in a variety of human prostate cancer cell lines. However, the effects of selenium have yet to be examined in normal and malignant cells derived from the same individual. Selenite metabolism consumes glutathione (GSH) and produces superoxide. The generation of reactive oxygen species is an important mechanism in selenite-induced apoptosis. METHODS: Three patient-matched pairs of primary prostatic epithelial cell cultures from normal and cancer were evaluated for their response to selenite. Apoptosis was measured and the differential response of normal and cancer cells was correlated with the expression of bcl-2, bax, GSH, and manganese superoxide dismutase (MnSOD). RESULTS: The cancer-derived cells were significantly more sensitive to selenite-induced apoptosis than the corresponding normal cells. Tumor-selective killing was not observed in cells treated with selenomethionine. The ratio of bcl-2:bax was decreased in the cancer-derived cells treated with selenite. Total GSH concentrations were similar in paired normal and cancer cells. Therefore, differences in GSH content do not appear to play a role in tumor-selective killing by selenite. Superoxide is a by-product of selenite metabolism and normal cells showed increased MnSOD expression and SOD activity compared to the cancer-derived cells. Prostate cancer cells treated with the MnSOD mimetic, MnTMPyP, were protected against the cytotoxic effects of selenite. CONCLUSIONS: Higher MnSOD expression in normal cells may play an important role in eliminating superoxide radicals produced as a result of selenite metabolism and contribute to the tumor-selective killing by selenite in prostate cancer.


Assuntos
Apoptose/efeitos dos fármacos , Próstata/citologia , Próstata/patologia , Neoplasias da Próstata/patologia , Selenito de Sódio/farmacologia , Western Blotting , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Epitélio/química , Epitélio/efeitos dos fármacos , Glutationa/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Próstata/química , Próstata/efeitos dos fármacos , Neoplasias da Próstata/química , Proteínas Proto-Oncogênicas c-bcl-2/análise , Sensibilidade e Especificidade , Selenito de Sódio/metabolismo , Superóxido Dismutase/metabolismo , Células Tumorais Cultivadas , Proteína X Associada a bcl-2/análise
3.
Endocr Relat Cancer ; 12(1): 19-47, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15788637

RESUMO

This review focuses on primary cultures of human prostatic epithelial cells and their applications as models of normal and malignant biological behavior. Current abilities to culture cells from normal tissues, from premalignant dysplastic lesions (prostatic intraepithelial neoplasia), from primary adenocarcinomas, and from metastases are described. Evidence for representation of the interrelated cells of the normal prostatic epithelium--stem cells, basal epithelial cells, secretory epithelial cells, transit amplifying cells and neuroendocrine cells--in primary cultures is presented. Comparisons between normal and cancer-derived primary cultures are made regarding biological activities relevant to carcinogenesis, such as proliferation, apoptosis, differentiation, senescence, adhesion, migration, invasion, steroid hormone metabolism, other metabolic pathways and angiogenesis. Analyses of tumor suppressor activity, differential gene expression and cytogenetics in primary cultures have revealed changes relevant to prostate cancer progression. Preclinical studies with primary cultures have provided information useful for designing new strategies for chemoprevention, chemotherapy, cytotoxin therapy, radiation therapy, gene therapy and imaging. While the behavior of normal primary cultures is often used as a basis for comparison with established, immortal prostate cancer cell lines, the most informative studies are performed with donor-matched pairs of normal and malignant primary cultures, grown under identical conditions. Challenges that remain to be addressed if the full potential of primary cultures as a model system is to be realized include isolation, culture and characterization of stem cells, improved methodology to induce or maintain a fully differentiated, androgen-responsive phenotype, and identification of cell surface antigens or other markers with which to purify pure populations of live cancer or premalignant cells apart from non-malignant epithelial cells prior to culture.


Assuntos
Adenocarcinoma/patologia , Modelos Biológicos , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/patologia , Células Epiteliais/citologia , Regulação da Expressão Gênica , Humanos , Masculino , Células-Tronco Neoplásicas/patologia , Próstata/citologia , Neoplasias da Próstata/metabolismo
4.
J Cell Physiol ; 202(2): 361-70, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15389580

RESUMO

Three mammalian isoforms of transforming growth factor-beta (TGFbeta) are known, TGFbeta1, 2, and 3, that have non-overlapping functions during development. However, their specific roles in cancers such as prostate cancer are less clear. Here we show that primary cultures of prostatic epithelial cells preferentially produce and activate the latent TGFbeta2 isoform. Paired cultures of normal and malignant prostate cells from prostate cancer patients produced predominantly the TGFbeta2 isoform, with 30- to 70-fold less TGFbeta1. By mono-Q ion exchange chromatography, three major peaks of latent TGFbeta2 activity were observed corresponding to the known small latent TGFbeta2 complex, the known large latent TGFbeta2 complex and a novel eluting peak of latent TGFbeta2. Although prostate cells are known to activate latent TGFbeta, the mechanism for activation is currently unclear. We investigated whether prostate specific antigen (PSA), a serine protease used as a clinical marker for prostate cancer, could play a role in the activation of latent TGFbeta. Unlike plasmin, a known activator of both latent TGFbeta1 and 2, PSA specifically activated the recombinant small latent form of TGFbeta2, but not TGFbeta1. Prostate epithelial cells, therefore, preferentially produce the TGFbeta2 isoform and PSA, a protease produced by the prostate, specifically targets the activation of this TGFbeta isoform. PSA-mediated activation of latent TGFbeta2 may be an important mechanism for autocrine TGFbeta regulation in the prostate and may potentially contribute to the formation of osteoblastic lesions in bone metastatic prostate cancer.


Assuntos
Antígeno Prostático Específico/farmacologia , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/efeitos dos fármacos , Idoso , Animais , Bovinos , Linhagem Celular Tumoral , Células Cultivadas , Células Epiteliais/metabolismo , Humanos , Masculino , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Fator de Crescimento Transformador beta2
5.
Mol Cell Endocrinol ; 225(1-2): 73-6, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15451570

RESUMO

Inhibin is a member of the TGF-beta superfamily of growth and differentiation factors and a tumor suppressor. Consistent with the tumor suppressive function of the inhibin alpha subunit in prostate cancer, we reported a loss of gene expression due to DNA hypermethylation and loss of heterozygosity (LOH) as well as down regulation of inhibin alpha subunit immunoreactivity. Paradoxically, an expanded study to evaluate the prognostic significance of inhibin alpha subunit expression in men with prostate cancer resulted in a contradictory outcome, whereby an up-regulation of subunit expression was recorded. In seeking a more comprehensive explanation for all data sets, we offer a unifying hypothesis. We propose that the tumor suppressor activities of the inhibin alpha subunit dominate in non-malignant tissue, but its oncogenic activities emerge during tumorigenesis. An explanation such as this, involving a switch in gene function from being tumor suppressive to pro-oncogenic, may account for the discrepant findings in other types of cancer.


Assuntos
Inibinas/fisiologia , Neoplasias da Próstata/etiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Inibinas/genética , Masculino , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia
6.
J Urol ; 171(1): 192-6, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14665874

RESUMO

PURPOSE: Less than 50% of men who undergo radical prostatectomy for prostate cancer are cured of disease. We evaluate tumor expression of inhibin alpha, a putative tumor suppressor, and the related protein, follistatin, to determine whether expression correlated with failure to be cured by surgery. MATERIALS AND METHODS: Tissues were selected from an archival collection of 379 prostatectomy specimens from men with followup of at least 5 years after surgery. Since previous studies showed that such men with only Gleason grade 3 cancer had a greater than 95% chance of no biochemical recurrence (increase in serum prostate specific antigen), our investigation was confined to 174 men with 2% or greater grade 4/5 cancer. These men had an intermediate rate of failure, providing an opportunity to analyze the potential contribution of inhibin alpha or follistatin to progression. Intensity of immunohistochemical labeling for inhibin alpha and follistatin in each cancer was compared with that in normal glands within the same tissue section. RESULTS: The majority of cases showed more intense expression of inhibin alpha in cancer than in normal glands. Those individuals whose cancers had the most elevated expression of inhibin alpha had a higher risk of recurrence, although this association was not statistically significant. Follistatin was expressed equivalently in normal and cancer cells in the majority of cases and did not correlate with recurrence. CONCLUSIONS: Our finding that inhibin alpha is frequently overexpressed in high grade prostate cancer suggests that the role of inhibin alpha as a tumor suppressor needs to be reevaluated. Furthermore, assessment of inhibin alpha as a serum marker of prostate cancer, as used to diagnose ovarian cancer, may be warranted.


Assuntos
Inibinas/biossíntese , Neoplasias da Próstata/metabolismo , Idoso , Folistatina/biossíntese , Humanos , Masculino , Prognóstico , Prostatectomia , Neoplasias da Próstata/cirurgia , Falha de Tratamento
7.
Endocr Relat Cancer ; 10(2): 131-40, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12790775

RESUMO

Prostate cancer is a leading cause of cancer-related deaths in many countries. Premalignant lesions and invasive cancer occur more frequently in the prostate than in any organ other than the skin. Yet, the incidence of clinically detected prostate cancer is much lower than the histopathological incidence. The slow growth of prostate cancer and the low incidence of clinically manifest disease in some geographical locations or racial/ethnic groups suggest that prostate cancer can be controlled, perhaps by dietary factors. Vitamin D and retinoids have emerged as leading candidates both to prevent and to treat prostate cancer. Many of the activities of these compounds, established from epidemiological studies, research with cell culture and animal models, and clinical trials, are consistent with tumor suppressor effects. However, retinoids may have additional tumor enhancer properties that balance or negate anti-cancer activity. This perhaps explains the overall lack of protective effects of vitamin A compounds against prostate cancer found in epidemiological studies, and the minimal efficacy of retinoids in clinical trials to treat prostate cancer. While current efforts focus on developing strategies to use vitamin D compounds to control prostate cancer, the possibility exists that prostate cancer cells may become resistant to tumor suppressor effects of vitamin D. Analyses of experimental model systems show that prostate cancer cells become less sensitive to vitamin D through loss of receptors or signaling molecules that mediate vitamin D's actions, or through changes in metabolic enzymes that synthesize or degrade vitamin D compounds. The potential promise of exploiting vitamin D to control prostate cancer is tempered by the possibility that prostate cancer, perhaps even at early stages, may develop mechanisms to escape tumor suppressor activities of vitamin D and/or retinoids.


Assuntos
Anticarcinógenos/uso terapêutico , Estado Nutricional , Neoplasias da Próstata/prevenção & controle , Retinoides/uso terapêutico , Vitamina D/uso terapêutico , Progressão da Doença , Humanos , Masculino
8.
Cell Death Differ ; 9(3): 252-63, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11859408

RESUMO

The primary objective of this study was to determine the sequence of biochemical signaling events that occur after modulation of the cellular redox state in the B cell lymphoma line, PW, with emphasis on the role of mitochondrial signaling. L-Buthionine sulphoximine (BSO), which inhibits gamma glutamyl cysteine synthetase (gammaGCS), was used to modulate the cellular redox status. The sequence and role of mitochondrial events and downstream apoptotic signals and mediators was studied. After BSO treatment, there was an early decline in cellular glutathione (GSH), followed by an increase in reactive oxygen species (ROS) production, which induced a variety of apoptotic signals (detectable at different time points) in the absence of any external apoptotic stimuli. The sequence of biochemical events accompanying apoptosis included a 95% decrease in total GSH and a partial (25%) preservation of mitochondrial GSH, without a significant increase in ROS production at 24h. Early activation and nuclear translocation of the nuclear factor kappa B subunit Rel A was observed at approximately 3h after BSO treatment. Cytochrome c release into the cytosol was also seen after 24h of BSO treatment. p53 protein expression was unchanged after redox modulation for up to 72 h, and p21waf1 independent loss of cellular proliferation was observed. Surprisingly, a truncated form of p53 was expressed in a time-dependent manner, beginning at 24h after BSO incubation. Irreversible commitment to apoptosis occurred between 48 and 72 h after BSO treatment when mitochondrial GSH was depleted, and there was an increase in ROS production. Procaspase 3 protein levels showed a time-dependent reduction following incubation with BSO, notably after 48 h, that corresponded with increasing ROS levels. At 96 h, caspase 3 cleavage products were detectable. The pan-caspase inhibitor zVADfmk, partially blocked the induction of apoptosis at 48 h, and was ineffective after 72 h. PW cells could be rescued from apoptosis by removing them from BSO after up to 48, but not 72 h incubation with BSO. Mitochondrial transmembrane potential (DeltaPsi(m)) remained intact in most of the cells during the 72 h observation period, indicating that DeltaPsi(m) dissipation is not an early signal for the induction of redox dependent apoptosis in PW cells. These data suggest that a decrease in GSH alone can act as a potent early activator of apoptotic signaling. Increased ROS production following mitochondrial GSH depletion, represents a crucial event, which irreversibly commits PW cells to apoptosis.


Assuntos
Apoptose/fisiologia , Butionina Sulfoximina/farmacologia , Glutationa/metabolismo , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Grupo dos Citocromos c/metabolismo , Citosol/metabolismo , Ditiotreitol/farmacologia , Glutationa/deficiência , Humanos , Linfoma de Células B/metabolismo , Linfoma de Células B/patologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mitocôndrias/fisiologia , NF-kappa B/metabolismo , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Transdução de Sinais/fisiologia , Fator de Transcrição RelA , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese
9.
Prostate ; 49(1): 9-18, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11550206

RESUMO

OBJECTIVES: Hensin induces terminal differentiation in rabbit kidney collecting tubule cells. Rabbit hensin and human DMBT1 result from alternative splicing of the same gene. The human DMBT1 gene is located on chromosome 10q25-26, a region often deleted in prostate cancer. In this study we examined the potential role of this gene in terminal differentiation of prostate, as well as its role in prostatic carcinogenesis. METHODS: We searched for deletions of this gene in prostatic cells cultured from cancer and benign tissues using PCR and cDNA cloning. The expression of hensin/DMBT1 in cultured cells and during prostate development was characterized by immunochemistry. RESULTS: No deletions of hensin/DMBT1 similar to those found in glioblastomas, lung and esophageal cancers were observed in prostate cancer or BPH cells. Hensin/DMBT1 protein was localized in intracellular vesicles of epithelial cells in neonatal and 6-week-old mouse prostates. By 6 weeks, hensin/DMBT1 began to localize in the basal lamina of the prostate and vas deferens. In matured 6-month-old prostates, there was extensive deposition of hensin/DMBT1 in the basal lamina. CONCLUSIONS: There is no evidence that hensin/DMBT1 is implicated in prostatic carcinogenesis. The localization of hensin/DMBT1 during maturation raises the possibility that hensin/DMBT1 is involved in terminal differentiation of the prostate and vas deferens.


Assuntos
Aglutininas , Diferenciação Celular , Transformação Celular Neoplásica , Proteínas de Membrana , Próstata/patologia , Receptores de Superfície Celular/biossíntese , Receptores Imunológicos/biossíntese , Receptores de Lipoproteínas , Ducto Deferente/patologia , Proteínas de Ligação ao Cálcio , DNA Complementar/genética , Proteínas de Ligação a DNA , Proteínas da Matriz Extracelular , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Reação em Cadeia da Polimerase , Próstata/citologia , Receptores Depuradores , Receptores Depuradores Classe B , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor , Ducto Deferente/citologia
10.
J Urol ; 166(4): 1560-4, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11547131

RESUMO

PURPOSE: We examined the association of androgen receptor gene cytosine-adenine-guanine (CAG) repeat length and the 2 single nucleotide polymorphisms A49T and V89L in the type II 5 alpha-reductase gene with prostate enlargement measured as the weight of the surgically removed prostate. MATERIALS AND METHODS: A total of 68 men with a prostate weighing 80 gm. or greater were compared with 197 controls with a prostate weighing less than 80 gm. These men had undergone radical prostatectomy between 1992 and 1996. DNA was extracted from archival prostate tissue uninvolved with cancer and genotyped for 3 polymorphic markers. The effects of genetic variants and clinicopathological variables on prostate enlargement risk were estimated by logistic regression. RESULTS: The age adjusted odds ratio estimate of prostate enlargement risk in men with 23 or greater versus 20 or fewer CAG repeats was 0.41 (95% confidence interval 0.19 to 0.89). This risk reduction was consistently found when an alternative prostate enlargement definition and subject restriction were used. No consistent association with prostate enlargement risk was observed for A49T or V89L polymorphisms. CONCLUSIONS: Our finding further supports the hypothesis that the shorter CAG repeat length of the androgen receptor gene is related to prostate enlargement.


Assuntos
Oxirredutases/genética , Polimorfismo Genético , Hiperplasia Prostática/genética , Receptores Androgênicos/genética , Adenina , Idoso , Colestenona 5 alfa-Redutase , Citosina , Guanina , Humanos , Masculino , Pessoa de Meia-Idade , Repetições de Trinucleotídeos/genética
11.
Cell Tissue Res ; 305(3): 389-97, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11572092

RESUMO

Cultured prostatic epithelial cells have been extensively studied as a model of prostate biology. What is the lineage relationship of the cultured cells to the epithelial cell types in tissue? How different are cultured cells derived from tumor tissue to those derived from benign tissue? Expression of cluster designation (CD) cell surface molecules has been shown to be useful in characterizing cells according to lineage. A CD profile was therefore generated for cultured human prostatic epithelial cells and compared with those previously established for basal and luminal epithelial cells in the prostate. Presence of CD44, CD49b, CD49f, and CD104 and absence of CD57 suggests that cultured cells were derived from basal cells of prostatic tissues. However, expression of certain CD antigens characteristic of luminal epithelial cells was also observed in subpopulations of cultured cells. The pattern of CD antigens in cultured cells reflects a phenotype similar to that of transit-amplifying cells that have been described in the prostate. Several CD antigens were found expressed by both cultured prostatic epithelial and stromal cells, and are probably associated with cell proliferation. The CD profiles of cultured epithelial cell strains derived from normal compared with malignant tissues were notably similar to each other and to that of the prostate cancer cell line PC-3. We conclude that cells in culture retain expression of certain lineage-characteristic CD antigens. Furthermore, CD antigens can define subpopulations of cells with differential gene expression.


Assuntos
Antígenos CD/análise , Antígenos CD/genética , Células Epiteliais/química , Neoplasias da Próstata , Idoso , Primers do DNA , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Receptores de Hialuronatos/análise , Receptores de Hialuronatos/genética , Integrina alfa2 , Integrina alfa6 , Integrina beta4 , Masculino , Fenótipo , Células Estromais/química , Células Estromais/citologia , Células Estromais/fisiologia , Células Tumorais Cultivadas
13.
Urology ; 58(2 Suppl 1): 123-6, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11502466

RESUMO

The high rate of progression of prostate cancer after androgen deprivation therapy mandates that new strategies be developed. Adjuvant therapy combined with androgen deprivation may slow or prevent progression. Ketoconazole plus calcitriol therapy is an example of 1 such a combination with a mechanistic basis for synergistic activity. Ketoconazole is commonly used as a second-line androgen deprivation therapy. This imidazole derivative is an inhibitor of P-450 enzymes, including those involved in steroidogenesis. Other P-450 enzymes that are inhibited by ketoconazole include 1alpha-hydroxylase and 24-hydroxylase, which metabolize vitamin D. Growth inhibition of prostate cancer cells by vitamin D depends on levels of the active metabolite, 1,25-dihydroxyvitamin D(3) (calcitriol). The enzyme 24-hydroxylase converts calcitriol to less active products. The inhibition of 24-hydroxylase by ketoconazole maintains the magnitude and duration of response to calcitriol. Combined ketoconazole/calcitriol therapy might therefore potentiate the antitumor activity of calcitriol. Because androgen-independent prostate cancer cells often remain responsive to growth inhibition by calcitriol, it is also possible that calcitriol would slow or prevent development of androgen-independent cancer growth. Another consideration is that ketoconazole blocks 1alpha-hydroxylase activity, which is the key enzyme that creates calcitriol in the body. Therefore, patients receiving ketoconazole therapy are likely to be deficient in vitamin D. The detrimental consequences of vitamin D deficiency in these patients would also be alleviated by the addition of calcitriol to the therapeutic regimen.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Cetoconazol/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Vitamina D/uso terapêutico , Antagonistas de Androgênios/administração & dosagem , Antagonistas de Androgênios/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Calcitriol/administração & dosagem , Calcitriol/farmacologia , Calcitriol/uso terapêutico , Sinergismo Farmacológico , Humanos , Cetoconazol/administração & dosagem , Cetoconazol/farmacologia , Masculino , Vitamina D/administração & dosagem
14.
Nat Cell Biol ; 3(5): 527-30, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11331884

RESUMO

Interactions between Eph receptor tyrosine kinases (RTKs) and membrane-anchored ephrin ligands critically regulate axon pathfinding and development of the cardiovascular system, as well as migration of neural cells. Similar to other RTKs, ligand-activated Eph kinases recruit multiple signalling and adaptor proteins, several of which are involved in growth regulation. However, in contrast to other RTKs, activation of Eph receptors fails to promote cell proliferation or to transform rodent fibroblasts, indicating that Eph kinases may initiate signalling pathways that are distinct from those transmitted by other RTKs. Here we show that stimulation of endogenous EphA kinases with ephrin-A1 potently inhibits the Ras/MAPK cascade in a range of cell types, and attenuates activation of mitogen-activated protein kinase (MAPK) by receptors for platelet-derived growth factor (PDGF), epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF). In prostatic epithelial cells and endothelial cells, but not fibroblasts, treatment with ephrin-A1 inhibits cell proliferation. Our results identify EphA kinases as negative regulators of the Ras/MAPK pathway that exert anti-mitogenic functions in a cell-type-specific manner.


Assuntos
Sistema de Sinalização das MAP Quinases , Proteínas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas ras/antagonistas & inibidores , Animais , Divisão Celular , Linhagem Celular , Fatores de Crescimento Endotelial/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Efrina-A1 , Fator de Crescimento Epidérmico/metabolismo , Fibroblastos/metabolismo , Humanos , Immunoblotting , Queratinócitos/metabolismo , Linfocinas/metabolismo , Masculino , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Fator de Crescimento Derivado de Plaquetas/metabolismo , Testes de Precipitina , Neoplasias da Próstata/metabolismo , Ratos , Receptor EphA1 , Receptor EphA2 , Transdução de Sinais , Fatores de Tempo , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Proteínas ras/metabolismo
15.
Cancer Res ; 61(7): 2852-6, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11306457

RESUMO

Evidence from epidemiological, molecular, and genetic studies suggests a role for vitamin D in the development and/or progression of prostate cancer. In experimental models and clinical trials, 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] was shown to exert antiproliferative, prodifferentiating, and antimetastatic/invasive effects on prostatic epithelial cells. Because the direct clinical application of 1,25(OH)2D3 is limited by the major side effect of hypercalcemia, we investigated the potential therapeutic utility of its less calcemic precursor, 25-hydroxyvitamin D3 [25(OH)D3], which is converted locally within the prostate to 1,25(OH)2D3 by 1alpha-hydroxylase. Quantification of 1alpha-hydroxylase activity in human prostatic epithelial cells by enzyme-substrate reaction analyses revealed a significantly decreased activity in cells derived from adenocarcinomas compared with cells derived from normal tissues or benign prostatic hyperplasia (BPH). In growth assays, we found that 25(OH)D3 inhibited growth of normal or BPH cells similarly to 1,25(OH)2D3. In contrast, in primary cultures of cancer cells and established cell lines, the antiproliferative action of 25(OH)D3 was significantly less pronounced than that of 1,25(OH)2D3. Our results indicate that growth inhibition by 25(OH)D3 depends on endogenous 1alpha-hydroxylase activity, and that this activity is deficient in prostate cancer cells. This finding has ramifications for both the prevention and therapy of prostate cancer with vitamin D compounds.


Assuntos
25-Hidroxivitamina D3 1-alfa-Hidroxilase/metabolismo , Calcifediol/farmacologia , Neoplasias da Próstata/enzimologia , Divisão Celular/efeitos dos fármacos , Células Epiteliais/enzimologia , Inibidores do Crescimento/farmacologia , Humanos , Masculino , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia
16.
Cancer Res ; 61(5): 2112-8, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11280774

RESUMO

The androgen-signaling pathway is important in the growth and progression of prostate cancer. Androgen ablation therapy, which may result in programmed cell death, is often used to treat advanced prostate cancer. The growth-promoting effects of androgen are mediated mostly through the androgen receptor (AR). Transforming growth factor beta (TGF-beta) plays critical roles in controlling prostate cell proliferation, differentiation, and apoptosis. Normal transcripts and proteins of TGF-beta receptors are frequently lost in prostate cancer cells, especially in advanced stages of the disease. However, the mechanisms by which TGF-beta inhibits proliferation and induces apoptosis in prostate cancer cells is not clear. We investigated the molecular mechanism by which TGF-beta inhibits transcriptional activation mediated by AR. Using transient transfection systems, we demonstrated that Smad3 specifically represses transcriptional activation mediated by AR on two natural androgen-responsive promoters. This repression is transmitted through TGF-beta signaling and can be regulated by other Smad proteins. A protein-protein interaction between AR and Smad3 was identified in vitro and in vivo, and the transcription activation domain of AR and the MH2 of Smad3 were identified as being responsible for binding. Additional functional experiments showed that the repression of AR by Smad3 is mediated solely through the MH2 domain. These results provide fresh insight for understanding the mechanism by which TGF-beta regulates the androgen-signaling pathway in prostate cancer cells.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Receptor Cross-Talk/fisiologia , Receptores Androgênicos/fisiologia , Transativadores/fisiologia , Transcrição Gênica/fisiologia , Antagonistas de Receptores de Andrógenos , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Haplorrinos , Humanos , Rim/citologia , Rim/fisiologia , Masculino , Próstata/citologia , Próstata/fisiologia , Estrutura Terciária de Proteína , Receptores Androgênicos/metabolismo , Transdução de Sinais/fisiologia , Proteína Smad3 , Transativadores/genética , Transativadores/metabolismo , Ativação Transcricional/fisiologia , Transfecção , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/fisiologia
17.
Rev Urol ; 3 Suppl 3: S15-22, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-16986004

RESUMO

New developments in molecular and cellular biology are rapidly increasing knowledge of the mechanisms of androgen action in the prostate. Molecular profiling of recurrent cancer after androgen deprivation therapy has revealed genetic changes related to cell survival and proliferation. It is now clear that loss of the androgen receptor (AR) does not explain androgen-independent growth of a majority of recurrent cancers. Rather, overexpression, mutation, and ligand-independent activation of the AR may confer enhanced sensitivity to androgen or other growth factors. Therefore, the term "androgen-independent" is often a misnomer. This emerging concept is the basis of novel strategies to prevent or slow recurrence.

18.
Prostate ; 45(2): 115-23, 2000 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11027410

RESUMO

BACKGROUND: Initial efforts to develop in vitro models to study prostatic biology focused on the culture and characterization of epithelial cells. Recently, attention has turned towards inclusion of stromal cells in experimental systems. METHODS: Improved methods to isolate and culture stromal cells have been developed. An array of markers are employed to characterize subtypes of stromal cells, with particular interest in smooth muscle differentiation. RESULTS: Defined, serum-free media are available for certain experimental applications. Conditions that promote smooth muscle differentiation have been identified. Investigators have characterized hormonal and peptide factors that regulate the growth of prostatic stromal cells, and have also described paracrine factors produced by stromal cells that influence epithelial biology. CONCLUSIONS: Prostatic stromal-cell cultures are now widely employed by a large number of investigators for a diverse array of experimental purposes. While further refinement is required to obtain model systems that fully mimic in vivo processes, the availability of stromal- and epithelial-cell cultures provides a valuable resource for studying normal prostatic biology as well as benign prostatic hyperplasia (BPH) and cancer.


Assuntos
Técnicas de Cultura de Células/métodos , Mesoderma/citologia , Adenocarcinoma , Agonistas Adrenérgicos/metabolismo , Androgênios/metabolismo , Animais , Biomarcadores , Linhagem Celular , Células Cultivadas , Células Epiteliais/metabolismo , Estrogênios/metabolismo , Substâncias de Crescimento/metabolismo , Humanos , Masculino , Mesoderma/fisiologia , Modelos Biológicos , Hiperplasia Prostática , Células Estromais/citologia , Células Estromais/metabolismo , Células Estromais/fisiologia , Sistema Urogenital/citologia
19.
Cancer Genet Cytogenet ; 120(2): 117-26, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10942801

RESUMO

Cytogenetics, fluorescence in situ hybridization (FISH), and comparative genomic hybridization (CGH) were used to identify genes that are involved in the development and progression of prostate cancer. For that purpose, we chose a cell line established in vitro from a prostatic adenocarcinoma which was nontumorigenic in nude mice and followed its progression to a tumorigenic cell line. Stepwise changes were observed in the cell line as it became tumorigenic. The composite karyotype at the nontumorigenic stage (CA-HPV-10) was 68 approximately 77,XXY,-(1, 9, 13, 14, 19, 22),+(4, 5, 11, 18, 20, 21),+(del(1) (q23q31)=M1 (two copies), +der(9)t(1;9)(q24 approximately q31;p23)=M5(two copies), der(14)t(14;?)(q10;?)=M17 in the majority of metaphases. These two derivative chromosomes were also observed a previous study. Our CGH analysis clearly showed that this deleted region in M1 is, in fact, translocated with derivative M5 and, in reality, is amplified. The cell line established from nodule (SCID 5019 p11), showed a number of new changes, as described; however, the most significant change was amplification of the 8q23 approximately qter region, harboring c-myc. This region was translocated with chromosomes 2, 4, and 16 as der(2)t(2;8)(q33;q23)=M12, der(4)t(4;8)(q34;q23)=M11, and der(16)t(8;16)(q24;q21)=M9. We deduce from our study that amplification of c-myc and other genes in the 8q23 approximately qter region were important in progression but did not lead to tumorigenicity. The population that became tumorigenic (SCID 5019 II) showed almost all of the same changes in the karyotype as observed in the nodular cell line; the only significant change was the appearance of der(11)t(4;11)(q32;q22)=M7 and the addition of another copy of t(3q;7p)=M2. These new changes lead to loss of chromosomes 3p, 4pter approximately q34, 6, 7q21 approximately qter, 11q22 approximately qter, and 18q, and gain of 3q, 7p, 8q23 approximately qter, and 11pter approximately q22, before the cell line became tumorigenic. The clonal selection of the population is proven by the presence of a number of the same derivative chromosomes in both the nodular and tumorigenic cell line. As it progressed to tumorigenicity, some of the same changes observed in the original study re-appear at different stages of malignancy, although it was absent in the nontumorigenic cell line. These are: der(16)t(8;16)(q24;q21)=M9 in the nodular cell line and der(11)t(4;11)(q32;q22)=M7 in the tumorigenic cell line. In our system, amplification of c-myc and other genes in der(2)t(2;8)(q33;q23)=M12,der(4) t(4;8)(q34;q23)=M11 together with the presence of der(16)t(8;16)(q24;q21)=M9 and der(11)t(4;11)(q32;q22)=M5 makes the cell line tumorigenic. It is either nontumorigenic, with the presence of a marker equivalent to der(16)=M9 and der(11)=M7 observed in the original study, and only nodular (SCID 5019 p11, present study), with the presence of number of markers with c-myc amplification (M9, M11, and M12). There is accumulation of all the above-mentioned changes in the same cell before it becomes tumorigenic.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Viral , Papillomaviridae/crescimento & desenvolvimento , Neoplasias da Próstata/genética , Aneuploidia , Animais , Linhagem Celular Transformada , Deleção Cromossômica , Análise Citogenética , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Hibridização de Ácido Nucleico , Fenótipo , Neoplasias da Próstata/patologia , Translocação Genética , Transplante Heterólogo , Células Tumorais Cultivadas
20.
J Urol ; 164(3 Pt 1): 836-41, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10953164

RESUMO

PURPOSE: The objective of this study was to investigate growth-inhibitory and apoptotic activity of the experimental antitumor drug, brefeldin A (BFA), on primary cultures of human epithelial cells derived from prostatic adenocarcinomas. MATERIALS AND METHODS: Clonal assays were performed to evaluate the effects of BFA on growth of prostatic cancer cell strains. Loss of cell viability in response to BFA was assessed by trypan blue exclusion. Induction of apoptosis by BFA was evaluated by morphologic criteria, electrophoretic assay of DNA fragmentation, and a cell death ELISA. Immunoblots were used to monitor p53 and pRB expression in response to BFA. RESULTS: BFA was growth-inhibitory at a half-maximal concentration of 5 ng./ml. (18 nM). Morphological manifestations of apoptosis were evident by 24 hours of treatment. Cell viability declined and the cell death ELISA indicated an 18-fold increase in apoptosis in BFA-treated versus untreated cells at 48 hours. DNA fragmentation was also seen at 48 hours. Levels of p53 were not altered by BFA, but pRB was maintained in a hypophosphorylated state by BFA treatment. CONCLUSIONS: BFA is a potent inducer of apoptosis in prostatic cancer cells via a p53-independent mechanism. Cells derived from low-grade as well as high-grade cancers responded similarly to BFA. Since p53-mediated pathways of apoptosis may frequently be abrogated in prostatic cancer cells, agents such as BFA that induce p53-independent cell death may be promising candidates for chemotherapeutic agents.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Brefeldina A/uso terapêutico , Genes p53/genética , Neoplasias da Próstata/tratamento farmacológico , Inibidores da Síntese de Proteínas/uso terapêutico , Adenocarcinoma/genética , Adenocarcinoma/patologia , Apoptose/genética , Morte Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Clonais/efeitos dos fármacos , Corantes , Fragmentação do DNA , Eletroforese em Gel de Ágar , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/efeitos dos fármacos , Humanos , Immunoblotting , Masculino , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteína do Retinoblastoma/efeitos dos fármacos , Azul Tripano , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...