Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Urol Oncol ; 35(3): 119, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28159492

RESUMO

PURPOSE: Percutaneous biopsy obtained from a single location is prone to sampling error in large heterogeneous renal masses, leading to nondiagnostic results or failure to detect poor prognostic features. We evaluated the accuracy of percutaneous biopsy for large renal masses using a modified multi-quadrant technique vs. a standard biopsy technique. MATERIALS AND METHODS: Clinical and pathological data for all patients with cT2 or greater renal masses who underwent percutaneous biopsy from 2009 to 2014 were reviewed. The multi-quadrant technique was defined as multiple core biopsies from at least 4 separate solid enhancing areas in the tumor. The incidence of nondiagnostic findings, sarcomatoid features and procedural complications was recorded, and concordance between biopsy specimens and nephrectomy pathology was compared. RESULTS: A total of 122 biopsies were performed for 117 tumors in 116 patients (46 using the standard biopsy technique and 76 using the multi-quadrant technique). Median tumor size was 10cm (IQR: 8-12). Biopsy was nondiagnostic in 5 of 46 (10.9%) standard and 0 of 76 (0%) multi-quadrant biopsies (P = 0.007). Renal cell carcinoma was identified in 96 of 115 (82.0%) tumors and nonrenal cell carcinoma tumors were identified in 21 (18.0%). One complication occurred using the standard biopsy technique and no complications were reported using the multi-quadrant technique. Sarcomatoid features were present in 23 of 96 (23.9%) large renal cell carcinomas studied. Sensitivity for identifying sarcomatoid features was higher using the multi-quadrant technique compared to the standard biopsy technique at 13 of 15 (86.7%) vs. 2 of 8 (25.0%) (P = 0.0062). CONCLUSIONS: The multi-quadrant percutaneous biopsy technique increases the ability to identify aggressive pathological features in large renal tumors and decreases nondiagnostic biopsy rates.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Biópsia , Humanos , Rim , Estudos Retrospectivos
2.
World J Urol ; 35(5): 729-735, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27631325

RESUMO

INTRODUCTION: A significant proportion of patients that fail active surveillance (AS) for prostate cancer management do so because of cancer upgrading. A previously validated upgrading nomogram generates a score that predicts risk of biopsy Gleason 6 upgrading following radical prostatectomy in lower-risk populations that are candidates for Active Surveillance (Cancer, 2013). OBJECTIVES: We hypothesize that the upgrading risk (UR) score generated by this nomogram at diagnosis improves the ability to predict patients that will subsequently fail AS. METHODS: To evaluate the nomogram, retrospective data from several institutional cohorts of patients who met AS criteria, group 1 (n = 75) and group 2 (n = 1230), were independently examined. A UR score was generated using the coefficients from the nomogram consisting of PSA density (PSAD), BMI, maximum % core involvement (MCI), and number of positive cores. AS failure was defined as Gleason score (GS) >6, >50 % maximum core involvement, or >2 positive cores on biopsy. Univariate and multivariate Cox proportional-hazards regression models, upgrading risk score, and other clinicopathologic features were each assessed for their ability to predict AS failure. RESULTS: Clinicopathologic parameters were similar in both groups with the exception of mean PSAD (0.13 vs. 0.11, p < 0.01) and follow-up (2.1 vs. 3.2 years, p = 0.2). Most common cause of AS failure was GS > 6 (group 1) compared to >2 positive cores (group 2). On univariate analysis in both populations, features at diagnosis including PSAD and the UR score were significant in predicting AS failure by upgrading (Gleason > 6) and any failure. Multivariate analysis revealed the UR score predicts AS failure by GS upgrading (HR 1.8, 95 % CI 1.12-2.93; p = 0.01) and any failure criteria (HR 1.7, 95 % CI 1.06-2.65); p = 0.02) for group 1. Likewise, the UR score in group 2 predicts AS failure with GS upgrading (HR 1.3, 95 % CI 1.15-1.42; p < 0.0001) and any failure criteria (HR 1.18, 95 % CI 1.18-1.38; p < 0.0001). An ROC generated an AUC of 0.66. Decision curve analysis demonstrated a high net benefit for the UR score across a range of threshold probabilities. Based on these outcomes, at 3 years, patients in the lowest risk quartile have a 15 % risk of AS failure versus a 46 % risk in the highest quartile (p < 0.0001). CONCLUSIONS: The UR score was predictive of pathologic AS failure on multivariate analysis in several AS cohorts. It outperformed single clinicopathologic criteria and may provide a useful adjunct using clinicopathologic data to stratify patients considering AS.


Assuntos
Algoritmos , Neoplasias da Próstata/patologia , Conduta Expectante , Fatores Etários , Idoso , Biópsia com Agulha de Grande Calibre , Índice de Massa Corporal , Estudos de Coortes , Gerenciamento Clínico , Humanos , Calicreínas/sangue , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Gradação de Tumores , Prognóstico , Modelos de Riscos Proporcionais , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/sangue , Neoplasias da Próstata/terapia , Estudos Retrospectivos , Medição de Risco
3.
Prostate ; 73(16): 1721-30, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23999928

RESUMO

BACKGROUND: Increasing age is a significant risk factor for prostate cancer. The prostate is exposed to environmental and endogenous stress that may underlie this remarkable incidence. DNA methylation, genomic imprinting, and histone modifications are examples of epigenetic factors known to undergo change in the aging and cancerous prostate. In this review we examine the data linking epigenetic alterations in the prostate with aging to cancer development. METHODS: An online search of current and past peer reviewed literature on epigenetic changes with cancer and aging was performed. Relevant articles were analyzed. RESULTS: Epigenetic changes are responsible for modifying expression of oncogenes and tumor suppressors. Several of these changes may represent a field defect that predisposes to cancer development. Focal hypermethylation occurs at CpG islands in the promoters of certain genes including GSTP1, RARß2, and RASSF1A with both age and cancer, while global hypomethylation is seen in prostate cancer and known to occur in the colon and other organs. A loss of genomic imprinting is responsible for biallelic expression of the well-known Insulin-like Growth Factor 2 (IGF2) gene. Loss of imprinting (LOI) at IGF2 has been documented in cancer and is also known to occur in benign aging prostate tissue marking the presence of cancer. Histone modifications have the ability to dictate chromatin structure and direct gene expression. CONCLUSIONS: Epigenetic changes with aging represent molecular mechanisms to explain the increased susceptibly of the prostate to develop cancer in older men. These changes may provide an opportunity for diagnostic and chemopreventive strategies given the epigenome can be modified.


Assuntos
Envelhecimento/genética , Epigênese Genética/genética , Predisposição Genética para Doença/genética , Neoplasias da Próstata/genética , Ilhas de CpG/genética , Metilação de DNA/genética , Impressão Genômica/genética , Histonas/genética , Humanos , Fator de Crescimento Insulin-Like II/genética , Masculino , Neoplasias da Próstata/epidemiologia , Fatores de Risco
4.
Br J Cancer ; 107(1): 100-7, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22596233

RESUMO

BACKGROUND: DNA methylation is an important epigenetic mechanism in prostate cancer (PCa) progression. Given the role of even-skipped homeobox 1 (EVX1) in the regulation of multiple genes during embryogenesis, we postulated that EVX1 methylation is altered in PCa progression. METHODS: Bisulphite sequencing and quantitative MethyLight were used to assess methylation in human prostate epithelial cells, four PCa cell lines, liver, lung, spleen, kidney, 35 paired tumour and tumour-associated benign tissues, and 11 normal prostate tissues. Prostate cancer cell lines were treated with 5-azacytidine (AzaC) or trichostatin A (TSA), and expression of EVX1 transcript and variants was assessed by qPCR. Hypermethylation was compared with clinicopathological features in a validation set of 58 patients using microarray. RESULTS: Even-skipped homeobox 1 hypermethylation was observed in all four PCa cell lines and 57% of tumours. High-grade tumours exhibited increased methylation compared with intermediate-grade tumours. Even-skipped homeobox 1 expression was induced in PCa cell lines after treatment with AzaC or TSA. In the validation set, 83% of tumours were hypermethylated and hypermethylation was associated with worse recurrence-free survival. CONCLUSION: In this first evaluation of EVX1 methylation in human cancer, EVX1 is one of the most commonly hypermethylated genes observed in PCa and predicted treatment failure in moderate risk patients.


Assuntos
Metilação de DNA , Proteínas de Homeodomínio/genética , Neoplasias da Próstata/genética , Azacitidina/farmacologia , Linhagem Celular Tumoral , Intervalo Livre de Doença , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Masculino , Prognóstico , Próstata/metabolismo , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Recidiva
5.
Minerva Urol Nefrol ; 60(1): 31-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18427433

RESUMO

Robotic assisted laparoscopic prostatectomy (RALP) is rapidly gaining acceptance in the urologic community as an effective treatment modality for the treatment of localized prostate cancer. For the open surgeon, this technology assists in the transition to laparoscopic-based surgery. With early stage cancer, the absence of tactile feedback appears to be offset by the superior visualization and instrumentation that the daVinci robot (Intuitive Surgical, Sunnyvale, CA, USA) provides. Economical considerations also appear to be outweighed by patient demand and benefits. Short-term perioperative, functional, and oncological outcomes all appear to be equivalent to the gold standard, open radical retropubic prostatectomy. In conclusion, RALP is an excellent option for patients considering prostate cancer treatment and should be part of the urologic surgeon's armamentarium.


Assuntos
Laparoscopia , Prostatectomia/instrumentação , Neoplasias da Próstata/cirurgia , Robótica , Humanos , Masculino , Prostatectomia/economia , Prostatectomia/métodos , Robótica/economia , Robótica/métodos , Resultado do Tratamento
6.
Urology ; 58(4): 517-20, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11597529

RESUMO

OBJECTIVES: Hand-assisted laparoscopic surgery is easier to learn than standard laparoscopy and simplifies intact specimen removal. We present our experience performing hand-assisted laparoscopic radical nephrectomy (HALRN) and compare it with contemporary open radical nephrectomy performed at our institution. METHODS: We performed 18 HALRNs for renal tumors ranging in size from 2 to 11 cm (average 4.5). Patients ranged in age from 40 to 83 years (average 62.9). All patients underwent HALRN with intact removal through a 7 to 8-cm vertical midline incision through an impermeable wound protector. Two or three working ports were used. We retrospectively compared our results with the results of 18 open radical nephrectomies performed during the same period, with the patients matched for age, body mass index, and American Society of Anesthesiologists' score. RESULTS: In the HALRN group, the average operating room time was 220.5 minutes, average length of stay 3.9 days, average time to return to normal activity 15.8 days, and average time to return to work 26.8 days. The median time to return to 100% normal was 28.0 days. No conversions or re-explorations were necessary in the HALRN series. The final pathologic examination revealed renal cell carcinoma in 15, oncocytoma in 1, angiomyolipoma in 1, and a complex cyst in 1. At a maximum of 48 months of follow-up (average 12.2), no recurrences were identified. Three deaths occurred in the series; 2 patients died with no evidence of disease and 1 patient died of metastatic disease (the nephrectomy was palliative). In the open group, the average operating room time was 117.8 minutes, average length of stay 5.1 days, average time to return to normal activity 23.5 days, and average time to return to work 52.2 days. The median time to return to 100% normal was 150 days, with 3 patients never returning to 100% normal. CONCLUSIONS: Our series demonstrated that HALRN is a safe, effective, minimally invasive option for treating renal cell carcinoma and provides a shorter hospital stay (P = 0.02), earlier return to work (P = 0.04), and earlier return to 100% normal (P = 0.0002) than open radical nephrectomy.


Assuntos
Neoplasias Renais/cirurgia , Laparoscopia/métodos , Nefrectomia/métodos , Adenoma Oxífilo/cirurgia , Adulto , Idoso , Idoso de 80 Anos ou mais , Angiomiolipoma/cirurgia , Carcinoma de Células Renais/cirurgia , Feminino , Seguimentos , Humanos , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Cuidados Paliativos , Estudos Retrospectivos , Resultado do Tratamento
8.
Oncogene ; 20(57): 8184-92, 2001 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-11781834

RESUMO

Cellular senescence has been proposed to be an in vitro and in vivo block that cells must overcome in order to immortalize and become tumorigenic. To characterize these pathways, we focused on changes in the cyclin-dependent kinase inhibitors and their binding partners that underlie the cell cycle arrest at senescence. As a model, we utilized normal human prostate epithelial cell (HPEC) and human uroepithelial cell (HUC) cultures. After 30-40 population doublings cells became growth-arrested in G0/1 with a threefold decrease in Cdk2-associated activity, a point defined as pre-senescence. Temporally following this growth arrest, the cells develop a senescence morphology and express senescence-associated beta-galactosidase (SA-beta-gal). Levels of p16(INK4a) and p57(KIP2) rise in HUCs during progressive passages, whereas only p16 increases in HPEC cultures. The induced expression of p57, similar to p16, produces a senescent-like phenotype. pRB, cyclin D, p19(INK4d) and p27(KIP1) decrease in both cell types. We find that p53, p21(CIP1) and p15(INK4b) are transiently elevated in HPECs and HUCs at the pre-senescent growth arrest, then return to low proliferating levels at terminal senescence. Analysis of p53, p21(CIP1), p15(INK4b), p16(INK4a), and p57(KIP2) reveals altered expression in immortalized, non-tumorigenic HPV16 E6 and E7 prostate lines and in tumorigenic prostate cancer cells. These results indicate: (i) the existence of a subset of growth inhibiting genes elevated at the onset of the senescence, (ii) a distinct class of genes involved in the maintenance of senescence, and (iii) the frequent inactivation of these pathways during immortalization.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular/fisiologia , Senescência Celular , Quinases Ciclina-Dependentes/antagonistas & inibidores , Próstata/metabolismo , Sistema Urinário/metabolismo , Adulto , Western Blotting , Ciclo Celular , Divisão Celular , Linhagem Celular Transformada , Transformação Celular Viral , Células Cultivadas , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p57 , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Próstata/citologia , Próstata/enzimologia , Neoplasias da Próstata/etiologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Transfecção , Células Tumorais Cultivadas , Sistema Urinário/citologia , Sistema Urinário/enzimologia , beta-Galactosidase/metabolismo
9.
Cancer Res ; 60(14): 3862-71, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10919661

RESUMO

Inactivation of both the pRb (pRb-cyclin D1/cyclin-dependent kinase 4/6-p16) and p53 (p53-p21(WAF1)-p14(ARF)) pathways is thought to be essential for immortalization in vitro and malignant transformation in vivo. We identified different combinations of pRb and p53 pathway alterations in 12 invasive transitional cell carcinomas (TCCs) and addressed the functional significance of the different combinations observed. Results showed four combinations of alterations including -pRb/-p53 (ie., pRb inactivated in the pRb pathway and p53 inactivated in the p53 pathway; four TCCs), -p16/-p53 (four TCCs), -p16/-p21(WAF1) (one TCC), and -p16/ -p14(ARF) (two TCCs). These groups include two new combinations (ie., -p16/-p53 and -p16/-p21(WAF1)) not reported previously for TCCs. An alteration in the key components of the p53 pathway was not detected in one invasive TCC that had inactivated p16. Note that all four TCCs with inactivated pRb had mutant p53; thus, the combinations of -pRb/ -p21(WAF1) and -pRb/-p14(ARF) were not observed. Only two of eight TCCs with altered p16 had concomitant p14(ARF) loss, demonstrating that simultaneous inactivation of these two 9p21INK4a tumor suppressor genes is not obligatory. To determine the biological phenotypes of TCCs with different combinations of pRb and p53 pathway alterations, their downstream responses to gamma radiation were studied in vitro. As expected, none of eight TCCs with mutant p53 responded to gamma radiation by elevation of p53, p21(WAF1), or mdm2 or by cell cycle arrest. Only two of four TCCs with wild-type p53 and wild-type pRb (the combination of -p16/-p14(ARF)) showed normal downstream responses to gamma radiation and underwent cell cycle arrest. Two TCCs with wild-type pRb and wild-type p53 (the combination of -pl6/-p21(WAF1) and one TCC with -p16) failed to show cell cycle arrest in response to radiation. This was attributed to the absence of p21(WAF1) in one TCC. In summary, these data support a model of invasive bladder cancer pathogenesis in which both the pRb and p53 pathways are usually inactivated and the biology of the tumor is impacted by the mechanism of their inactivations.


Assuntos
Genes p53/genética , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Neoplasias da Bexiga Urinária/genética , Northern Blotting , Southern Blotting , Carcinoma de Células de Transição/genética , Carcinoma de Células de Transição/metabolismo , Proteínas de Transporte/metabolismo , Cromossomos Humanos Par 9/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Invasividade Neoplásica/genética , Fenótipo , Proteínas/metabolismo , Análise de Sequência de DNA , Proteína Supressora de Tumor p14ARF , Neoplasias da Bexiga Urinária/metabolismo
10.
Cancer Res ; 60(13): 3623-30, 2000 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10910077

RESUMO

Advanced hormone-independent prostate cancer is characterized by a significant loss of androgen receptor (AR) expression in 20-30% of the tumors. The transcriptional block underlying this phenomenon is not known, but we have proposed that methylation of CpG sites in the AR promoter may reversibly inactivate transcription of the AR (D. F. Jarrard et al, Cancer Res., 58: 5310-5314, 1998). In this study, detailed methylation analysis using bisulfite sequencing was performed on a series of AR expression-positive and -negative prostate cancer cells. We found that methylation of several consensus sequences in the AR promoter (from -131 to -121 and +44 to +54) are tightly linked to the loss of AR expression in metastatic hormone-independent prostate cancer cell lines. These consensus sites of methylation correlate with the minimal promoter region critical for AR transcription. In human tissues, no methylation was demonstrated in normal or primary prostate cancers that express the AR. Four of 15 tumors obtained from men who had died from hormone-independent prostate cancer demonstrated a significant loss of AR expression immunohistochemically and two (50%) of these AR-negative tumors contained AR methylation. We conclude that the AR promoter contains specific CpG methylation hot spots that are markers for gene silencing. Furthermore, AR methylation may represent a phenotype important in the development of hormone independence in a subset of advanced prostate cancer in which AR expression is lost. The finding of AR methylation also represents the first report of aberrant methylation on an X-linked gene associated with a somatic male cancer.


Assuntos
Inativação Gênica , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , Receptores Androgênicos/genética , Transcrição Gênica , Regiões 3' não Traduzidas/genética , Sequência de Bases , Sequência Consenso , Metilação de DNA , Primers do DNA , DNA de Neoplasias/química , Fosfatos de Dinucleosídeos/análise , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Células Tumorais Cultivadas
12.
Anal Biochem ; 278(2): 165-9, 2000 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10660458

RESUMO

DNA methylation is an important epigenetic modification that alters transcription in those genes containing CpG islands. In this report we demonstrate that the familiar technique single-strand conformation polymorphism (SSCP) can be easily applied to bisulfite-treated DNA to detect methylation differences over large ( approximately 250 bp) CG-enriched regions. In the methylation-sensitive SSCP (Ms-SSCP) technique, sodium bisulfite modification of DNA converts unmethylated cytosine to uracil under conditions in which 5-methylcytosine remains unaltered. Amplification of these regions to be tested is then performed using primers specific for bisulfite-treated DNA. The resulting products are then subjected to nondenaturing gel analysis. Based on differential band mobility, Ms-SSCP is able to sensitively detect alterations in methylation density and determine if unmethylated alleles are present in a sample. Ms-SSCP is a rapid and simple technique for screening multiple samples for methylation changes. It has several advantages over existing techniques, including the utilization of nanogram amounts of DNA, the avoidance of difficulties in sequencing CG-enriched regions, and the screening of multiple sites for methylation across large regions of DNA.


Assuntos
Metilação de DNA , DNA/análise , Conformação de Ácido Nucleico , Polimorfismo Genético , DNA/química , Humanos , Masculino , Células Tumorais Cultivadas
14.
Cancer Res ; 59(12): 2957-64, 1999 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10383161

RESUMO

The cell cycle regulatory genes p16/CDKN2 and RB are frequently deleted in prostate cancers. In this study, we examined the role of alterations in p16 and pRb during growth, senescence, and immortalization in vitro of human prostate epithelial cells (HPECs). HPECs are established from normal prostate tissues and cultured on collagen-coated dishes. Our results show that p16 is reproducibly elevated at senescence in HPECs. HPECs are immortalized using human papilloma virus 16 E6 and/or E7 as molecular tools to inactivate p53 and/or pRb, respectively. Immortalization occurs infrequently in this system and only after a latent period during which additional genetic/epigenetic changes are thought to occur. Notably, all of the E6-immortalized HPEC lines but none of the E7 lines show inactivation of p16/CDKN2 (by deletion, methylation, or mutation) in association with immortalization. In contrast, E7 lines, in which pRb function is abrogated by E7 binding, retain the high levels of p16 observed at senescence. Thus, all lines show either a p16 or pRb inactivation. Analysis of six independent lines from metastatic prostate cancers reveals a similar loss of either p16 or pRb. Comparative genomic hybridization of HPECs shows that gains of chromosomes 5q, 8q, and 20 are nonrandomly associated with bypassing senescence (probability = 0.95). These results suggest that high levels of the cyclin-dependent kinase inhibitor p16 mediate senescence G1 arrest in HPECs and that bypassing this block by a p16/pRb pathway alteration is required for immortalization in vitro and possibly tumorigenesis in vivo. Our results further indicate that inactivation of the p16/pRb pathway alone is not sufficient to immortalize HPECs and that additional genetic alterations are required for this process.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Células Epiteliais/metabolismo , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Repressoras , Proteína do Retinoblastoma/metabolismo , Idoso , Senescência Celular , Aberrações Cromossômicas , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Metilação de DNA , Células Epiteliais/patologia , Deleção de Genes , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Metástase Neoplásica , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/metabolismo , Proteínas E7 de Papillomavirus , Próstata/patologia , Neoplasias da Próstata/patologia , Proteína Supressora de Tumor p53/metabolismo
15.
Cancer Res ; 58(23): 5310-4, 1998 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9850055

RESUMO

Androgen-independent metastatic prostate cancer is characterized by a heterogeneous loss of androgen receptor (AR) expression among tumor cells. In this study, we evaluate DNA hypermethylation as a potential transcriptional regulatory mechanism in AR-negative prostate cancer cell lines. Nucleotide sequence analysis demonstrates an approximately 15-kb CpG island in the AR gene that encompasses the transcription start site and exon 1. Using Southern blotting with methylation-sensitive restriction enzymes and methylation-specific PCR, we find aberrant methylation in the AR expression-negative cell lines Du145, DuPro, TSU-PR1, and PPC1. Incomplete methylation in the AR CpG island is also seen in normal female breast and ovarian tissues consistent with the inactivation of one X chromosome by hypermethylation. In contrast, prostate cancer cell lines LNCaP and PC3 express AR and are unmethylated. Normal prostate epithelial cell strains demonstrate no methylation. Exposure of AR-negative prostate cancer cell lines to 5-aza-2' deoxycytidine, a demethylating agent, induces the reexpression of AR RNA in DuPro and TSU-PR1. This reexpression is associated with a demethylation of this region. Prostate-specific antigen, an androgen-responsive gene, is also specifically induced in these lines after AR reexpression. Therefore, in vitro DNA methylation of the 5' CpG AR island may be associated with the loss of AR expression. Furthermore, our results demonstrate that treatment with demethylating agents may engender the reexpression and function of the androgen receptor in AR-negative cell lines.


Assuntos
Ilhas de CpG/fisiologia , Metilação de DNA , Regiões Promotoras Genéticas/fisiologia , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/biossíntese , Humanos , Masculino , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/ultraestrutura , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas
16.
Genes Dev ; 12(2): 163-74, 1998 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9436977

RESUMO

Elevation of p16, the CDKN2/p16 tumor suppressor gene (TSG) product, occurs at senescence in normal human uroepithelial cells (HUC). Immortal HUCs and bladder cancer cell lines show either alteration of p16 or pRb, the product of the retinoblastoma (RB) TSG. In addition, many human cancers show p16 or pRb alteration along with other genetic alterations that we associated with immortalization, including +20q and -3p. These observations led us to hypothesize that p16 elevation plays a critical role in senescence cell cycle arrest and that overcoming this block is an important step in tumorigenesis in vivo, as well as immortalization in vitro. Using a novel approach, we tested these hypotheses in the present study by examining p16 and pRb status in primary culture (P0) and after passage in vitro of transitional cell carcinoma (TCC) biopsies that represented both superficial bladder tumors and invasive bladder cancers. We demonstrated that all superficial TCCs showed elevated p16 after limited passage in vitro and then senesced, like normal HUCs. In contrast, all muscle invasive TCCs contained either a p16 or a pRb alteration at P0 and all spontaneously bypassed senescence (P = 0.001). Comparative genomic hybridization (CGH) was used to identify regions of chromosome loss or gain in all TCC samples. The application of a statistical model to the CGH data showed a high probability of elevated alteration rates of +20q11-q12 (0.99) and +8p22-pter (0.94) in the immortal muscle invasive TCCs, and of -9q (0.99) in the superficial TCCs. Three myoinvasive TCCs lost 3p13-p14. In this study, four of six myoinvasive TCCs also showed TP53 mutation that associated well with genome instability (P = 0.001), as previously hypothesized. Notably, TP53 mutation, which has been used as a marker of tumor progression in many human cancers, was less significant in associating with progression in this study (P = 0.04) than was p16 or pRb alteration (P = 0.001). Thus, these data support a new model in which overcoming senescence plays a critical role in human cancer pathogenesis and requires at least two genetic changes that occur in several combinations that can include either p16 or pRb loss and at least one additional alteration, such as +20q11-q12, -3p13-p14, or -8p21-pter.


Assuntos
Carcinoma de Células de Transição/metabolismo , Senescência Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Invasividade Neoplásica/genética , Retinoblastoma/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Southern Blotting , Western Blotting , Carcinoma de Células de Transição/genética , Aberrações Cromossômicas , Transtornos Cromossômicos , Inibidor p16 de Quinase Dependente de Ciclina/genética , Epitélio/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Retinoblastoma/genética , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Neoplasias da Bexiga Urinária/genética
17.
Genes Chromosomes Cancer ; 19(2): 90-6, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9171999

RESUMO

The tumor suppressor gene CDKN2 (p16/MTS1) resides on chromosome 9p21 and encodes a 16 kDa inhibitor of the cyclin-dependent kinases. Inactivation of CDKN2 by homozygous deletion, point mutation, and recently described aberrant methylation in the 5' promoter region may increase progression through the cell cycle in tumors. In this study, we examine the CDKN2 gene for the presence of inactivating alterations in human prostate cancer. Sequence analysis of cell lines revealed no mutation in LNCaP, PC3, and TSU-PR1 and a missense mutation, GAC-->TAC (asp to tyr), in exon 2 of the DU145 cell line at codon 76. No mutations were identified in three primary prostate cancers or in seven lymph node metastases. Loss of heterozygosity (LOH) was analyzed by analysis of microsatellite markers in the vicinity of the CDKN2 gene. LOH was detected in 12 (20%) of 60 primary tumors at one or more loci and in 13 (46%) of 28 metastases. Methylation analysis of the CpG-rich promoter region revealed a dense methylation of CDKN2 in cell lines PC3, PPC1, and TSU-PR1, and this was found to correlate with a lack of mRNA expression by reverse transcription-polymerase chain reaction. A demethylating agent, 5-aza-2'-deoxycytidine, induced reexpression when cells were exposed in vitro. DU145 and LNCaP expressed the CDKN2 transcript and were unmethylated in the promoter region. Three of twenty-four (13%) primary prostate cancers and 1 of 12 metastatic tumors demonstrated promoter methylation. No normal prostate tissues were methylated at the CDKN2 gene promoter. One tumor was found to contain concomitant LOH and promoter methylation indicative of biallelic inactivation. A comprehensive analysis of CDKN2 in prostate cancer reveals that point mutations are infrequent, but gene deletion and methylation combine to inactivate CDKN2 in a subset of tumors. Moreover, alterations in this gene may represent a late event in prostate cancer progression.


Assuntos
Proteínas de Transporte/genética , Deleção Cromossômica , Metilação de DNA , Análise Mutacional de DNA , DNA de Neoplasias/análise , Neoplasias da Próstata/genética , Idoso , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Inibidor p16 de Quinase Dependente de Ciclina , Metilases de Modificação do DNA , Decitabina , Expressão Gênica , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Reação em Cadeia da Polimerase , Neoplasias da Próstata/patologia , Análise de Sequência de DNA , Células Tumorais Cultivadas
18.
Br J Urol ; 79 Suppl 1: 37-43, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9088271

RESUMO

Cadherins are a family of calcium-dependent cell-cell adhesion molecules involved in cell-cell aggregation and morphoregulatory cell function. Dysfunction of the cadherin pathway is involved in tumour invasiveness and disease progression for a variety of carcinomas. E-cadherin is a prognostic marker in prostatic cancer, based on the correlation of the grade of E-cadherin expression and tumour grade, stage, metastasis and survival, as well as recurrence after radical prostatectomy. P-cadherin was shown to be lost in all prostatic cancers, although this most likely reflects loss of the basal cell population rather than a transcriptional down-regulation, suggesting that loss of P-cadherin expression is an early event in the tumorigenesis of prostatic carcinomas. Catenins, particularly alpha-catenin, also play an important role in the dysfunction of the cell adhesion complex. Mechanisms of inactivation of the cadherin-catenin pathway include LOH, gene deletions and gene promoter hypermethylation. Therapeutic strategies have been investigated in tumour models, i.e. the use of demethylating agents for the hypermethylated promoter region of E-cadherin or gene transfer in PC-3 cells with homozygous deletion of the alpha-catenin gene. The complexity of neoplastic changes cannot be explained by alterations of cell adhesion molecules alone; but as demonstrated, cadherins and catenins play an important role in this process.


Assuntos
Caderinas/fisiologia , Neoplasias da Próstata/patologia , Transativadores , Caderinas/metabolismo , Adesão Celular/fisiologia , Proteínas do Citoesqueleto , Metilação de DNA , Progressão da Doença , Humanos , Masculino , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , beta Catenina
19.
Cancer Res ; 57(1): 35-7, 1997 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-8988036

RESUMO

Production of the potent vasoconstrictor endothelin-1 (ET-1) by human prostate cancer cells accompanies prostate cancer progression in vivo. The predominant endothelin receptor expressed by normal prostate epithelium, ETB, is not expressed by any of the established human prostate cancer cell lines, and ETB binding is decreased on prostate cancer tissues. ETB, which may mediate ET-1 clearance and may inhibit ET-1 secretion, is encoded by a gene that contains a 5' CpG island encompassing the transcriptional regulatory region. We examined this regulatory region of the ETB receptor gene (EDNRB) to determine whether hypermethylation of cytidine nucleotides accompanies decreased ETB expression in human prostate cancer. We found somatic methylation of CpG island sequences in EDNRB in 5 of 5 human prostate cancer cell lines, 15 of 21 primary prostate cancer tissues, and 8 of 14 prostate cancer metastases (70% of samples overall). Normal tissues contained only unmethylated EDNRB. Treatment of human prostatic carcinoma cell line cultures with 5-azacytidine induced ETB mRNA expression, suggesting that CpG island methylation changes might accompany the apparent transcriptional silencing of EDNRB in vivo.


Assuntos
Fosfatos de Dinucleosídeos/metabolismo , Neoplasias da Próstata/genética , Receptores de Endotelina/genética , Sequências Reguladoras de Ácido Nucleico , Fosfatos de Dinucleosídeos/genética , Humanos , Masculino , Metilação , Neoplasias da Próstata/metabolismo , Receptor de Endotelina B , Receptores de Endotelina/metabolismo , Células Tumorais Cultivadas
20.
Clin Cancer Res ; 3(11): 2121-8, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9815605

RESUMO

P-Cadherin is a member of the cadherin family of cell surface glycoproteins that mediate Ca2+-dependent cell-cell adhesion and is expressed in a differential fashion in normal epithelial tissues. The expression of P-cadherin in human prostate cancer development has not been investigated previously. By immunohistochemistry, we show that P-cadherin expression is restricted to the cell-cell border of basal epithelial cells in 30 normal prostate samples. This staining is down-regulated in prostatic intraepithelial neoplasia and is absent in all 25 of the well to poorly differentiated prostate cancer specimens analyzed. To examine potential P-cadherin-regulatory elements, we sequenced the 5'-flanking region of this gene. Similar to the mouse gene, the human P-cadherin promoter is TATA-less, contains an Sp-1 binding site and, analogous to the human E-cadherin sequence, demonstrates a GC-rich region characteristic of a CpG island. Cytosine methylation of this region occurs in P-cadherin-negative prostate cancer cell lines but not in cell lines expressing this gene. In vivo, a lack of expression in 12 clinical prostate cancer specimens is not associated with methylation of the P-cadherin promoter. These results demonstrate that the expression of the basal cell marker P-cadherin is lost in prostate cancer development and that in vivo mechanisms other than cytosine methylation regulate this consistent loss of expression.


Assuntos
Caderinas/análise , Próstata/citologia , Neoplasias da Próstata/patologia , Animais , Composição de Bases , Sítios de Ligação , Biomarcadores/análise , Caderinas/genética , Fosfatos de Dinucleosídeos/análise , Células Epiteliais/citologia , Células Epiteliais/patologia , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Regiões Promotoras Genéticas , Próstata/patologia , Mapeamento por Restrição , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Sp1/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...