Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Nat Commun ; 11(1): 5070, 2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-33033260

RESUMO

The evolutionary progression from primary to metastatic prostate cancer is largely uncharted, and the implications for liquid biopsy are unexplored. We infer detailed reconstructions of tumor phylogenies in ten prostate cancer patients with fatal disease, and investigate them in conjunction with histopathology and tumor DNA extracted from blood and cerebrospinal fluid. Substantial evolution occurs within the prostate, resulting in branching into multiple spatially intermixed lineages. One dominant lineage emerges that initiates and drives systemic metastasis, where polyclonal seeding between sites is common. Routes to metastasis differ between patients, and likely genetic drivers of metastasis distinguish the metastatic lineage from the lineage that remains confined to the prostate within each patient. Body fluids capture features of the dominant lineage, and subclonal expansions that occur in the metastatic phase are non-uniformly represented. Cerebrospinal fluid analysis reveals lineages not detected in blood-borne DNA, suggesting possible clinical utility.


Assuntos
Linhagem da Célula , Biópsia Líquida , Neoplasias da Próstata/patologia , Líquidos Corporais/metabolismo , Cromossomos Humanos Par 8/genética , Células Clonais , Variações do Número de Cópias de DNA/genética , DNA de Neoplasias/genética , Loci Gênicos , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Filogenia
2.
Oncogene ; 31(35): 3939-48, 2012 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-22139082

RESUMO

A clearer definition of the molecular determinants that drive the development and progression of prostate cancer (PCa) is urgently needed. Efforts to map recurrent somatic deletions in the tumor genome, especially homozygous deletions (HODs), have provided important positional information in the search for cancer-causing genes. Analyzing HODs in the tumors of 244 patients from two independent cohorts and 22 PCa xenografts using high-resolution single-nucleotide polymorphism arrays, herein we report the identification of CHD1, a chromatin remodeler, as one of the most frequently homozygously deleted genes in PCa, second only to PTEN in this regard. The HODs observed in CHD1, including deletions affecting only internal exons of CHD1, were found to completely extinguish the expression of mRNA of this gene in PCa xenografts. Loss of this chromatin remodeler in clinical specimens is significantly associated with an increased number of additional chromosomal deletions, both hemi- and homozygous, especially on 2q, 5q and 6q. Together with the deletions observed in HEK293 cells stably transfected with CHD1 small hairpin RNA, these data suggest a causal relationship. Downregulation of Chd1 in mouse prostate epithelial cells caused dramatic morphological changes indicative of increased invasiveness, but did not result in transformation. Indicating a new role of CHD1, these findings collectively suggest that distinct CHD1-associated alterations of genomic structure evolve during and are required for the development of PCa.


Assuntos
Montagem e Desmontagem da Cromatina , DNA Helicases/genética , DNA Helicases/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Deleção de Genes , Neoplasias da Próstata/genética , Animais , Linhagem Celular , Regulação para Baixo , Células HEK293 , Homozigoto , Humanos , Masculino , Camundongos , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , PTEN Fosfo-Hidrolase/genética , Polimorfismo de Nucleotídeo Único , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Transplante Heterólogo
3.
Am J Pathol ; 159(5): 1815-26, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11696442

RESUMO

GSTP1 CpG island hypermethylation is the most common somatic genome alteration described for human prostate cancer (PCA); lack of GSTP1 expression is characteristic of human PCA cells in vivo. We report here that loss of GSTP1 function may have been selected during the pathogenesis of human PCA. Using a variety of techniques to detect GSTP1 CpG island DNA hypermethylation in PCA DNA, we found only hypermethylated GSTP1 alleles in each PCA cell in all but two PCA cases studied. In these two cases, CpG island hypermethylation was present at only one of two GSTP1 alleles in PCA DNA. In one of the cases, DNA hypermethylation at one GSTP1 allele and deletion of the other GSTP1 allele were evident. In the other case, an unmethylated GSTP1 allele was detected, accompanied by abundant GSTP1 expression. GSTP1 CpG island DNA hypermethylation was responsible for lack of GSTP1 expression by LNCaP PCA cells: treatment of the cells with 5-azacytidine (5-aza-C), an inhibitor of DNA methyltransferases, reversed the GSTP1 promoter DNA hypermethylation, activated GSTP1 transcription, and restored GSTP1 expression. GSTP1 promoter activity, assessed via transfection of GSTP1 promoter-CAT reporter constructs in LNCaP cells, was inhibited by SssI-catalyzed CpG dinucleotide methylation. Remarkably, although selection for loss of GSTP1 function may be inferred for human PCA, GSTP1 did not act like a tumor suppressor gene, as LNCaP cells expressing GSTP1, either after 5-aza-C treatment or as a consequence of transfection with GSTP1 cDNA, grew well in vitro and in vivo. Perhaps, GSTP1 inactivation may render prostatic cells susceptible to additional genome alterations, caused by electrophilic or oxidant carcinogens, that provide a selective growth advantage.


Assuntos
Ilhas de CpG/fisiologia , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Neoplasias da Próstata/metabolismo , Alelos , Sequência de Bases/genética , Southern Blotting , Testes de Carcinogenicidade , Divisão Celular/fisiologia , Ilhas de CpG/genética , DNA de Neoplasias/genética , Glutationa S-Transferase pi , Glutationa Transferase/antagonistas & inibidores , Glutationa Transferase/deficiência , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/deficiência , Masculino , Metilação , Neoplasias da Próstata/patologia , Valores de Referência , Células Tumorais Cultivadas
4.
Prostate ; 48(4): 248-53, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11536304

RESUMO

BACKGROUND: We have previously identified 12p12-13 as a region of frequent genetic loss in prostate carcinoma. A candidate tumor suppressor gene at this locus is the cyclin dependent kinase inhibitor p27(kip1), which has been implicated as a marker of aggressive prostate carcinoma. Herein, we examine metastatic prostate tumors, xenografts, and cell lines for gene inactivation via mutational inactivation or promoter hypermethylation. METHODS: Mutation analysis was performed on metastatic prostate tumors of 18 patients, eight prostate carcinoma cell lines, and 18 xenografts by PCR amplification of the entire open reading frame of p27(kip1). PCR products were sequenced directly using internal primers. Methylation analysis was performed on four cell lines and nine xenografts using direct sequencing of cloned PCR products of bisulfite treated DNA. Presence of a CpG was consistent with methylation of that cytosine in the original sample. RESULTS: With the exception of the previously reported homozygous deletion, no additional mutations were identified. Methylated CpG residues were identified in three xenografts (LuCAP23, LuCAP35, and PC82) and the methylated residues clustered at six sites; the cytosines 69, 149, 191, 286, 349, and 487 base pairs 5' of the ATG start codon. However, no sample demonstrated promotor methylation in all sequenced clones and the number of methylated base pairs ranged from seven to three, not the level usually associated with gene silencing. CONCLUSIONS: Mutational inactivation of p27(kip1) is a rare event in metastatic prostate carcinoma. While CpG methylation does occur, it is an infrequent event and does not appear to be the mechanism of p27(kip1) down regulation in prostate carcinoma.


Assuntos
Proteínas de Ciclo Celular/genética , Metilação de DNA , Neoplasias da Próstata/genética , Proteínas Supressoras de Tumor , Inibidor de Quinase Dependente de Ciclina p27 , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Genes cdc , Humanos , Masculino , Mutação , Neoplasias da Próstata/metabolismo , Análise de Sequência de DNA , Células Tumorais Cultivadas
5.
Cancer Res ; 61(17): 6372-6, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11522628

RESUMO

Osteoblastic metastases are common in lethal prostate cancer. Effective therapy for bone metastases is lacking. Thus, developing an appropriate in vitro screening system is critical to prioritize which of the newly developed agents should undergo additional expensive and time-consuming in vivo evaluation in bone metastases animal models. In the past, such in vitro screening evaluated the response of prostate cancer cells to chemotherapeutic agents in monoculture without the presence of osteoblasts. In such monoculture, prostate cancer cells have a high (i.e., >90%) proliferative growth fraction. In contrast, the growth fraction (i.e., mean: 7.1 +/- 0.8%; median: 3.1%) in 117 metastatic sites of prostate cancer obtained from 11 androgen ablation failing patients at "warm" autopsy was found to be >10-fold lower. To better mimic the lower growth fraction observed clinically, LNCaP human prostate cancer cells were cocultured with membrane-separated hFOB human osteoblasts. Such coculturing significantly lowered the growth fraction of the LNCaP cells (i.e., from >90 to <30%) without enhancing their low rate (i.e., <5%) of apoptosis. This lowering of the growth fraction was documented using flow cytometry, Ki-67 immunohistochemistry, and 5-bromo-2-deoxyuridine incorporation. Using RNase protection assays, it was documented that coculture with osteoblasts causes enhanced p53, p27, and p21 expression leading to a decrease in the number of LNCaP cells entering the cell cycle (i.e., enhanced number of LNCaP cells in G(0)-G(1) and a decrease in S and G(2)-M and thus the growth fraction). This osteoblast-induced enhanced G(0)-G(1) checkpoint control affected the chemosensitivity of LNCaP cells. This was documented by coculturing LNCaP cells with hFOB cells to condition the medium for 3 days to lower the growth fraction to <30% before exposing the LNCaP cells for 48 h to various concentrations of Taxol, doxorubicin, or thapsigargin (TG). In standard high (i.e., >90%) growth fraction cultures (i.e., cultures in the absence of osteoblast-conditioned medium), there was a dose-dependent and significant (P < 0.05) increase in apoptosis of LNCaP cells exposed to Taxol or doxorubicin. In contrast, even the highest dose of Taxol (1 microM) did not enhance apoptosis of lower growth fraction LNCaP cells cultured in osteoblast-conditioned medium. Similarly, only the highest concentration of doxorubicin (1 microM) enhanced apoptosis in lower growth fraction cells. In contrast, 100 nM TG induced high levels of apoptosis in both lower and high-growth fraction LNCaP cultures. These results demonstrate that the osteoblast/LNCaP coculture system is a better in vitro screen than monoculture to identify proliferation-independent agents for the treatment of prostate cancer bone metastases, and TG is such an agent.


Assuntos
Comunicação Celular/fisiologia , Fase G1/fisiologia , Osteoblastos/citologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Fase de Repouso do Ciclo Celular/fisiologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Técnicas de Cocultura , Meios de Cultivo Condicionados , Doxorrubicina/farmacologia , Fase G1/efeitos dos fármacos , Humanos , Masculino , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/patologia , Paclitaxel/farmacologia , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Tapsigargina/farmacologia , Células Tumorais Cultivadas
6.
Hum Pathol ; 32(4): 417-27, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11331959

RESUMO

Tissue microarray technology promises to enhance tissue-based molecular research by allowing improved conservation of tissue resources and experimental reagents, improved internal experimental control, and increased sample numbers per experiment. Organized, well-validated collection and analysis of the voluminous image data produced by tissue microarray technology is critical to maximize its value. Web-based technology for visual analysis and searchable storage of microarray image data could provide optimal flexibility for research groups in meeting this goal, but this approach has not been examined scientifically. Toward this goal, a prostate tissue microarray block containing 432 tissue cores (0.6 mm diameter) was constructed. Moderately compressed (200 kb).jpg images of each tissue spot were acquired and were saved using a naming convention developed by the SPORE Prostate Tissue Microarray Collaborative Group. Four hundred three tissue array spot images were uploaded into a database developed for this study and were converted to.fpx format to decrease Internet transmission times for high-resolution image data. In phase I of the image analysis portion of the study, testing and preliminary analysis of the Web technology was performed by 2 pathologists (M.A.R. and G.S.B.). In phase II, 2 pathologists (J.I.E. and T.M.W.) with no previous exposure to this technology and no knowledge of the structure of the study were presented a set of 130 sequential tissue spot images via the Web on their office computers. In phase III, the same pathologists were presented a set of 193 images, including all 130 from phase II and 63 others, with image presentation order randomized. With each zoomable tissue spot image, each pathologist was presented with a nested set of questions regarding overall interpretability of the image, presence or absence of cancer, and predominant and second most frequent Gleason grade. In phases II and III of the study, 319 of 323 (99%) image presentations using this Web technology were rated interpretable. Comparing the 2 pathologists' readings in phases II and III, Gleason grade determinations by each pathologist were identical in 179 of 221 (81%) determinations and were within 1 point of each other in 221 of 221 (100%) determinations, a performance rate similar to if not better than that previously reported for direct microscopic Gleason grading. Interobserver comparison of Gleason score determinations and intraobserver comparisons for Gleason grade and score also showed a pattern of uniformity similar to those reported in direct microscope-based Gleason grading studies. Interobserver (7.5%) and intraobserver (5% and 3%) variability in determining whether diagnosable cancer was present point out the existence of a "threshold effect" that has rarely been studied but may provide a basis for identification of features that are most amenable to improved diagnostic standardization. In summary, storage and analysis of tissue microarray spot images using Web-based technology is feasible and practical, and the quality of images obtained using the techniques described here appears adequate for most tissue-based pathology research applications. HUM PATHOL 32:417-427.


Assuntos
Bases de Dados Factuais , Processamento de Imagem Assistida por Computador , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Humanos , Masculino , Neoplasias da Próstata
7.
Prostate ; 47(2): 102-10, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11340632

RESUMO

BACKGROUND: Fatty acid synthase (FAS) performs the anabolic conversion of dietary carbohydrate or protein to fat. FAS expression is low in most normal tissues, but is elevated in many human cancers, including androgen-sensitive and androgen-independent prostate cancer. METHODS: Immunohistochemical evaluation of FAS expression was performed in human prostate cancer specimens under various states of androgen ablation. In vitro and in vivo prostate cancer models were evaluated for FAS expression and activity under androgenic and androgen-depleted conditions, and were tested for sensitivity to antimetabolite drugs that target fatty acid synthesis. RESULTS: While FAS expression in the prostate was androgen responsive, it persisted or was reactivated in human prostate carcinoma after androgen ablation, and was high in 82% of lethal tumors examined at autopsy. Similar patterns of FAS expression and fatty acid synthesis were seen in cell culture and xenograft models of human prostate cancer. Pharmacologic inhibition of FAS resulted in a dose-dependent reduction of tumor growth in these models, including fourfold inhibition of an androgen-independent human prostate cancer xenograft with little associated toxicity. CONCLUSIONS: The data suggest that FAS expression/FA synthesis provides an important functional aspect of the malignant phenotype in prostate cancer, perhaps supporting cell growth or survival. FAS expression may be upregulated by alternate signaling pathways important for prostate cancer growth under androgen withdrawal. The re-emergence of FAS expression and activity during the development of androgen independence demonstrate that FAS may serve as a novel target for antimetabolite therapy in prostate cancer.


Assuntos
4-Butirolactona/análogos & derivados , Androgênios/fisiologia , Ácido Graxo Sintases/biossíntese , Neoplasias da Próstata/enzimologia , 4-Butirolactona/farmacologia , Animais , Anticorpos Monoclonais , Antineoplásicos/farmacologia , Western Blotting , Cromatografia em Camada Fina , Inibidores Enzimáticos/farmacologia , Ácido Graxo Sintases/antagonistas & inibidores , Humanos , Imuno-Histoquímica , Lipídeos/análise , Masculino , Camundongos , Camundongos Nus , Neoplasias Hormônio-Dependentes/enzimologia , Distribuição Aleatória , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Prostate ; 45(4): 323-34, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11102958

RESUMO

BACKGROUND: E-cadherin and alpha-catenin are components of adherens junctions which mediate calcium-dependent, cell-cell adhesion in a homotypic manner. Both these molecules have been defined as useful tumor markers as their altered expression correlates with increased tumor aggressiveness and dedifferentiation. More recently, alterations of a third component of adherens junctions, beta-catenin, have been observed to play a role in several human cancers. Dysregulation of beta-catenin, either by direct mutation or by defects in interacting pathways/regulators, can result in its cytoplasmic accumulation and nuclear translocation. In the nucleus, beta-catenin forms a transcriptional complex capable of upregulating target genes, many of which encode proliferative factors. Given its oncogenic activity and connection to human cancer, we examined the beta-catenin gene and its expression in prostate cancer. METHODS: By single-stranded conformational polymorphism (SSCP) and DNA sequencing analyses, we screened exon 3 of beta-catenin from a panel of 81 primary tumors obtained at radical prostatectomy, 22 lymph node metastases from untreated patients, and a unique set of 61 metastatic tissues from 19 patients who died of hormone-refractory disease. RESULTS: We found putative activating mutations (missense and deletion) at a rate of 5% (7/138). One patient had the same 72 base pair deletion in each of nine separate metastases examined, indicating that this change was associated with a clonal population of metastatic cells. CONCLUSIONS: Immunohistological staining of mutation-positive tumors demonstrated beta-catenin accumulation and nuclear localization in a heterogeneous fashion. Consistent with this in vivo finding, our in vitro analyses demonstrate that certain mutations can result in increased beta-catenin nuclear activity in prostate cancer cell lines. These data implicate the beta-catenin signaling pathway in the development of a subset of prostate cancers.


Assuntos
Proteínas do Citoesqueleto/genética , Mutação de Sentido Incorreto , Neoplasias da Próstata/genética , Deleção de Sequência , Transativadores , Animais , Proteínas do Citoesqueleto/biossíntese , Análise Mutacional de DNA , DNA de Neoplasias/genética , Éxons , Humanos , Imuno-Histoquímica , Masculino , Microscopia de Fluorescência , Metástase Neoplásica , Estadiamento de Neoplasias , Polimorfismo Conformacional de Fita Simples , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transplante Heterólogo , Células Tumorais Cultivadas , beta Catenina
9.
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
10.
J Urol ; 164(1): 192-6, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10840458

RESUMO

PURPOSE: Our laboratory has recently identified a 1 to 2 Mb homozygous deletion at 12p12-13 in a prostate cancer specimen and determined that the p27/kip1 gene lies within the deletion. While immunohistochemical analysis has implicated p27/kip1 in prostate carcinoma, no previous studies had identified genetic abnormalities at this locus. Here, we examined primary and metastatic prostate tumors to determine if allelic loss occurs at this locus in localized disease and if it increases the risk of metastatic, high stage or high-grade disease. MATERIALS AND METHODS: DNA was extracted from prostate tumors and normal tissue of 99 patients. 60 tumors were primary, 20 were metastatic pelvic lymph nodes, and 19 were distant metastases. Multiple metastases were analyzed from 11 of 19 patients with metastatic disease. Polymorphic markers spanning our region of interest were PCR amplified from tumor and normal DNA. PCR products were then scored for allelic loss. RESULTS: Loss of heterozygosity (LOH) was identified in 14/60 (23%) primary tumors, 6/20 (30%) lymph node metastasis, and 9/19 (47%) distant metastases. The difference between primary and distant metastatic disease was statistically significant (p = 0.045, Fisher's exact test). The pattern of LOH was identical in all metastatic sites obtained from individual patients, indicating that genetic loss occurred prior to metastasis. Subset analysis of the 60 primary tumors demonstrated no association between LOH and adverse pathological feature [nodal involvement, seminal vesicle invasion, margin positivity, high Gleason score (7-10)]. CONCLUSIONS: Demonstrating that 12p12-13 LOH is a prominent feature of primary prostate tumors and that multiple metastatic foci have an identical LOH pattern, provides evidence that gene inactivation in this region occurs prior to metastasis. In addition, the strong association between LOH and distant metastasis raises the possibility that mutational inactivation of a gene at 12p12-13, possibly p27/kip1, plays a pivotal role in the development of metastatic disease.


Assuntos
Adenocarcinoma/genética , Perda de Heterozigosidade , Neoplasias da Próstata/genética , Adenocarcinoma/secundário , Cromossomos Humanos Par 12/genética , Humanos , Masculino , Neoplasias da Próstata/patologia
11.
Genes Chromosomes Cancer ; 25(3): 270-6, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10379873

RESUMO

The identification of homozygous deletions in malignant tissue is a powerful tool for the localization of tumor suppressor genes. Representational difference analysis (RDA) uses selective hybridization and the polymerase chain reaction (PCR) to isolate regions of chromosomal loss and has facilitated the identification of tumor suppressor genes, such as BRCA2 and PTEN. We have recently identified a 1-5-cM homozygous deletion on 12p12-13 in a prostate cancer xenograft and found that 47% of patients who died of prostate carcinoma demonstrate focal loss of heterozygosity (LOH) in this region in metastatic deposits. We have now characterized the region of interest by assembling a yeast artificial chromosome (YAC) contig spanning the homozygous deletion and identifying which known genes and expressed sequence tags (EST) lie within the homozygous deletion. A rib metastasis was harvested at autopsy and placed subcutaneously in a male SCID mouse. Genomic DNA from this xenograft and from the patient's normal renal tissue was extracted. Multiplex PCR, with the xenograft and normal DNA used as template, was performed using primers for loci on the Whitehead contig 12.1 believed to be near our region of interest. We found that our deletion lay in a 1-2-Mb interval between WI-664 and D12S358. We then used the same primers to construct a YAC contig across the homozygous deletion. PCR amplification of YAC DNA, using primers for the genomic sequences of known genes and ESTs reported to lie on 12p12-13, was used to identify candidate genes that lay within the deletion. Duplex PCR, with control primers known not to be deleted in the xenograft, was used to confirm that both the CDKN1B and ETV6 genes were homozygously deleted in the xenograft. Mutations in either or both of these genes may play an important role in metastatic prostate carcinoma.


Assuntos
Deleção Cromossômica , Mapeamento Cromossômico/métodos , Cromossomos Humanos Par 12/genética , Neoplasias da Próstata/genética , Animais , Neoplasias Ósseas/patologia , Carcinoma/genética , Carcinoma/patologia , Mapeamento de Sequências Contíguas , Humanos , Masculino , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Costelas/patologia
12.
Am J Pathol ; 154(6): 1835-40, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10362809

RESUMO

Peutz-Jeghers syndrome (PJS) is an autosomal-dominant disorder characterized by hamartomatous polyps in the gastrointestinal tract and by pigmented macules of the lips, buccal mucosa, and digits. Less appreciated is the fact that PJS also predisposes patients to an increased risk of gastrointestinal cancer, and pancreatic cancer has been reported in many PJS patients. It was recently shown that germline mutations of the STK11/LKB1 gene are responsible for PJS. We investigated the role of STK11/LKB1 in the development of pancreatic and biliary cancer in patients with and without the PJS. In a PJS patient having a germline splice site mutation in the STK11/LKB1 gene, sequencing analysis of an intestinal polyp and pancreatic cancer from this patient revealed loss of the wild-type allele of the STK11/LKB1 gene in the cancer. Inactivation of STK11/LKB1, by homozygous deletions or somatic sequence mutations coupled with loss of heterozygosity, was also demonstrated in 4-6% of 127 sporadic pancreatic and biliary adenocarcinomas. Our results demonstrate that germline and somatic genetic alterations of the STK11/LKB1 gene may play a causal role in carcinogenesis and that the same gene contributes to the development of both sporadic and familial forms of cancer.


Assuntos
Adenocarcinoma/genética , Neoplasias do Ducto Colédoco/genética , Mutação , Neoplasias Pancreáticas/genética , Síndrome de Peutz-Jeghers/genética , Proteínas Serina-Treonina Quinases/genética , Quinases Proteína-Quinases Ativadas por AMP , Adenocarcinoma/complicações , Adulto , Southern Blotting , Linhagem Celular , Análise Mutacional de DNA , Feminino , Marcadores Genéticos , Mutação em Linhagem Germinativa , Humanos , Perda de Heterozigosidade , Neoplasias Pancreáticas/complicações , Síndrome de Peutz-Jeghers/complicações , Reação em Cadeia da Polimerase , Deleção de Sequência
13.
Cancer Res ; 58(24): 5652-5, 1998 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-9865716

RESUMO

The identification of homozygous deletions in malignant tissue has been a powerful tool for the localization of tumor suppressor genes. Representational difference analysis (RDA) uses selective hybridization and the PCR to isolate regions of chromosomal loss and has facilitated the identification of tumor suppressor genes such as BRCA2 and PTEN. Twenty RDA clones were generated by comparing genomic DNA from a prostate cancer xenograft to the same patient's normal kidney DNA. Southern blot analysis of the tester and driver and of normal and xenograft DNA, using the differential products as probes, showed the homozygous deletion in 16 of 20 RDA clones. The sequence of one of the differential products overlapped HSU59962, a genomic GenBank sequence on chromosome 12p12-13. Multiplex PCR of the xenograft DNA using polymorphic repeats mapped the deletion to a 1-5-cM region on 12p. Genomic DNA isolated from a panel of cryostat microdissected metastatic prostate adenocarcinomas/normal pairs was screened for loss of heterozygosity using the same polymorphic repeats. Loss of heterozygosity was demonstrated in 9 (47%) of 19 patients. This region may contain, or lie in close proximity to, tumor suppressor genes important in the progression and/or initiation of prostate cancer.


Assuntos
Cromossomos Humanos Par 12 , Neoplasias da Próstata/genética , Deleção de Sequência , Adenocarcinoma/genética , Animais , Southern Blotting , Neoplasias Ósseas/secundário , Mapeamento Cromossômico , Humanos , Perda de Heterozigosidade , Masculino , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Transplante de Neoplasias , Reação em Cadeia da Polimerase , Neoplasias da Próstata/patologia
14.
Nat Genet ; 20(2): 175-9, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9771711

RESUMO

Over 200,000 new prostate cancer cases are diagnosed in the United States each year, accounting for more than 35% of all cancer cases affecting men, and resulting in 40,000 deaths annually. Attempts to characterize genes predisposing to prostate cancer have been hampered by a high phenocopy rate, the late age of onset of the disease and, in the absence of distinguishing clinical features, the inability to stratify patients into subgroups relative to suspected genetic locus heterogeneity. We previously performed a genome-wide search for hereditary prostate cancer (HPC) genes, finding evidence of a prostate cancer susceptibility locus on chromosome 1 (termed HPC1; ref. 2). Here we present evidence for the location of a second prostate cancer susceptibility gene, which by heterogeneity estimates accounts for approximately 16% of HPC cases. This HPC locus resides on the X chromosome (Xq27-28), a finding consistent with results of previous population-based studies suggesting an X-linked mode of HPC inheritance. Linkage to Xq27-28 was observed in a combined study population of 360 prostate cancer families collected at four independent sites in North America, Finland and Sweden. A maximum two-point lod score of 4.60 was observed at DXS1113, theta=0.26, in the combined data set. Parametric multipoint and non-parametric analyses provided results consistent with the two-point analysis. Significant evidence for genetic locus heterogeneity was observed, with similar estimates of the proportion of linked families in each separate family collection. Genetic mapping of the locus represents an important initial step in the identification of an X-linked gene implicated in the aetiology of HPC.


Assuntos
Predisposição Genética para Doença/genética , Neoplasias da Próstata/genética , Cromossomo X , Adulto , Idoso , Idoso de 80 Anos ou mais , Mapeamento Cromossômico , Marcadores Genéticos , Genótipo , Humanos , Escore Lod , Masculino , Pessoa de Meia-Idade , Linhagem , Receptores Androgênicos/genética
15.
J Urol ; 160(4): 1405-9, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9751364

RESUMO

PURPOSE: Reports in the osteoporosis literature demonstrating the increased activity of specific alleles of the vitamin D receptor and epidemiological data linking vitamin D levels with prostate cancer have stimulated research into possible associations between vitamin D receptor genotype and the development of prostate cancer. Recent studies showed that patients homozygous for a less active vitamin D receptor have a 4 to 5 times increased risk of localized prostate cancer. In 1 study this association was strongest in patients with advanced disease. To understand better the relationship between advanced disease and the vitamin D receptor we compared the vitamin D receptor genotype of 41 patients who died of prostate cancer to 41 controls with no clinical evidence of prostate cancer. MATERIALS AND METHODS: Noncancerous deoxyribonucleic acid was isolated from lethal prostate cancer and control cohorts. To determine the TaqI restriction fragment length polymorphism a 740 base pairs (bp) segment of the vitamin D receptor was amplified by PCR, digested with TaqI endonuclease and resolved on an agarose gel. Depending on the presence or absence of a TaqI restriction site at codon 352 in each allele, products were digested into 2 fragments of 495 and 245 bp (T allele) or 3 fragments of 290, 245 and 205 bp (t allele). Individuals were classified as TT, Tt or tt. To determine the size of the poly-A microsatellite repeat an approximately 410 bp fragment was amplified by polymerase chain reaction using [gamma-32P] labeled primers, resolved by gel electrophoresis and sized by autoradiography. Fragments 410 bps or greater corresponded to repeats 18 bps or greater (L allele) and fragments less than 410 bps corresponded to repeats less than 18 bps (S allele). Individuals were classified as LL, LS or SS. RESULTS: The TaqI restriction fragment length polymorphism and poly-A microsatellite repeat were in strong linkage disequilibrium, indicating that both polymorphic markers identified the same vitamin D receptor genotype in the majority of cases. Using the TaqI restriction fragment length polymorphism 33 of 41 controls (80.5%) and 35 of 41 cases (85.3%) were heterozygous (Tt) or homozygous (TT) for the less active vitamin D receptor allele, while 8 of 41 controls (19.5%) and 6 of 41 cases (14.6%) were homozygous (tt) for the more active vitamin D receptor allele. Using the poly-A microsatellite repeat 32 of 40 controls (80%) and 32 of 38 cases (84.2%) were heterozygous (LS) or homozygous (LL) for the less active vitamin D receptor allele, while 8 of 40 controls (20.0%) and 6 of 38 cases (15.8%) were homozygous (SS) for the more active vitamin D receptor allele. There was no statistically significant difference in the distribution of either marker between cases and controls. CONCLUSIONS: We failed to demonstrate an association between vitamin D receptor genotype and lethal prostate cancer. Our data do not support the hypothesis that specific vitamin D receptor genotypes are associated with an aggressive prostate cancer phenotype. Further studies that take into account cofactors important in vitamin D activity and/or a better definition of prostate cancer phenotype may explain the discrepancy between our findings and those of a previous study.


Assuntos
Polimorfismo Genético/genética , Neoplasias da Próstata/genética , Receptores de Calcitriol/genética , Idoso , Humanos , Masculino , Pessoa de Meia-Idade
16.
J Urol ; 160(3 Pt 1): 660-3, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9720516

RESUMO

PURPOSE: We answer the question of whether, following radical prostatectomy, prostate cancer in patients with a family history of prostate cancer relapses biochemically at the same rate as prostate cancer in appropriately matched patients with no family history. MATERIALS AND METHODS: The study was performed in 2 parts. In both parts prostate specific antigen (PSA) progression was defined as a postoperative elevation in serum PSA greater than 0.2 ng./ml. Part 1 included 656 patients who underwent radical prostatectomy by the same surgeon. Men with a family history of prostate cancer in a father or brother (94) were compared to those with no history of prostate cancer in a father or brother (562). Part 2 comprised 52 men with a family history of prostate cancer consistent with hereditary prostate cancer (HPC). HPC is defined as a family with 3 generations affected, 3 first-degree relatives affected or 2 relatives affected before age 55 years. Each member of this HPC group was matched by postoperative Gleason score and postoperative pathological stage with a patient who also underwent radical prostatectomy in the same time frame by the same surgeon but who reported no family history of prostate cancer by telephone interview and questionnaire. RESULTS: In part 1, 94 probands (14%) reported a history of prostate cancer in the father or in at least 1 brother. The remaining 562 probands (85%) reported no known history of prostate cancer in the father or brother(s). There was no statistically significant difference in the probability of maintaining an undetectable PSA between these 2 groups. In part 2, 45 of 52 pairs (87%) were matched identically for all matching criteria. Mean follow time for the sporadic and hereditary groups was 5.4 and 5.1 years, respectively. There was no statistically significant difference in the probability of maintaining an undetectable PSA between the 2 groups. CONCLUSIONS: Men with an affected father or brother, or those with a family history consistent with HPC have the same outcome following radical prostatectomy as men with no family history of the disease. Combined with our previous studies, these findings suggest that there is no biologically important difference between hereditary and sporadic prostate cancers.


Assuntos
Recidiva Local de Neoplasia/genética , Prostatectomia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Progressão da Doença , Humanos , Masculino , Pessoa de Meia-Idade , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/sangue , Neoplasias da Próstata/cirurgia , Fatores de Tempo
17.
Prostate ; 36(3): 172-80, 1998 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-9687989

RESUMO

BACKGROUND: TrkA, the high affinity, tyrosine kinase receptor for nerve growth factor (NGF) has been implicated as an oncogene in several neoplasms. In prostate cancer, inhibitors of the NGF/TrkA signal pathway results in tumor growth inhibition. In contrast, inhibition of this trk pathway in the normal prostate produces no effect. One explanation for this difference between normal and malignant prostate is that TrkA is mutated in prostate cancer, changing its function. To test this possibility human primary prostate cancers were screened for evidence of mutations in the TrkA gene to identify how this gene might be activated in prostate cancer. METHODS: Single-strand conformation polymorphism was used to screen genomic DNA, isolated from 42 human primary prostate cancers. In samples in which an aberrant banding pattern was identified, the screen was repeated using both the tumor DNA and DNA isolated from normal tissue of the same patients. Genetic changes were confirmed by direct sequencing of the aberrantly migrating bands. RESULTS: Although somatic mutations were not identified in any of the exons screened, four polymorphisms were detected in three different exons. Some of these polymorphisms occurred in the majority of the patients screened, but their frequencies were similar when compared with DNA isolated from a control group. CONCLUSIONS: Genetic mutations of TrkA do not seem to play a significant role in activation of this pathway in prostate cancer. However, the absence of mutations in otherwise genetically unstable prostate tumor DNA suggests that intact NGF/TrkA pathways may be important in prostate cancer development.


Assuntos
Mutação , Fatores de Crescimento Neural/genética , Polimorfismo Conformacional de Fita Simples , Neoplasias da Próstata/genética , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Receptores de Fator de Crescimento Neural/genética , Sequência de Aminoácidos , Análise Mutacional de DNA , Primers do DNA , Humanos , Masculino , Dados de Sequência Molecular , Neoplasias da Próstata/etiologia , Receptor trkA
18.
Cancer Epidemiol Biomarkers Prev ; 7(6): 531-6, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9641498

RESUMO

Prostate intraepithelial neoplasia (PIN) is a purported prostate cancer precursor lesion and a candidate biomarker for efficacy assessment in prostate cancer chemoprevention trials. Loss of expression of the pi-class glutathione S-transferase enzyme GSTP1, which is associated with the hypermethylation of deoxycytidine residues in the 5'-regulatory CG island region of the GSTP1 gene, is a near-universal finding in human prostate cancer. GSTP1 expression was assessed by immunohistochemistry in 60 high-grade PIN samples adjacent to and distant from prostate adenocarcinoma. Whereas abundant enzyme polypeptide expression was evident in all normal prostatic tissues, all samples of high-grade PIN and adenocarcinoma were completely devoid of GSTP1. DNA from 10 high-grade PIN lesions was analyzed for GSTP1 CG island methylation changes using a PCR technique targeting a polymorphic (ATAAA)n repeat sequence in the promoter region of the GSTP1 gene. Somatic GSTP1 CG island methylation changes were detected in DNA from 7 of the 10 PIN lesions. Allele discrimination was possible for 5 of the 10 DNA samples: 2 of the 5 samples exhibited DNA methylation changes at both alleles; whereas 3 samples displayed no DNA methylation changes at either allele. GSTP1 CG island methylation changes were present in each of the five homozygous samples. Hypermethylation of the 5'-regulatory region of the GSTP1 gene may serve as an important molecular genetic biomarker for both prostate cancer and PIN. The finding of frequent GSTP1 methylation changes in PIN and prostate cancer supports a role for PIN lesions as a prostate cancer precursor and may provide insight to the molecular pathogenesis of prostate cancer.


Assuntos
Adenocarcinoma/enzimologia , Biomarcadores Tumorais/genética , Glutationa Transferase/genética , Neoplasia Prostática Intraepitelial/enzimologia , Neoplasias da Próstata/enzimologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Metilação de DNA , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Reação em Cadeia da Polimerase , Neoplasia Prostática Intraepitelial/genética , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia
19.
Cancer Res ; 58(2): 204-9, 1998 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9443392

RESUMO

The long arm of chromosome 10 is frequently affected by allelic loss in prostate cancer. PTEN/MMAC1, a candidate tumor suppressor gene located at 10q23.3, a region commonly deleted in prostate cancer, was recently identified and found to be deleted or mutated in cancer cell lines derived from a variety of human tissues including prostate. To examine the role of PTEN/MMAC1 in the progression of prostate cancer, we screened a unique set of 50 metastatic prostate cancer tissues from 19 cancer-death patients for alterations in the PTEN/MMAC1 gene, using single-strand conformational polymorphism analysis and direct sequencing to identify sequence changes and microsatellite analysis to examine allelic loss in the vicinity of PTEN/MMAC1. Overall, gene alterations (deletions or point mutations) were observed in at least 1 metastatic site in 12 of the 19 patients studied. Two cases had homozygous deletions that were confirmed by fluorescence in situ hybridization analysis. Four patients harbored point mutations, with one mutation being found in all four tumors (a primary lesion and three different metastases) from the same patient. The remaining three mutations were detected in only one of multiple metastases. Loss of heterozygosity was found in 10 of 18 informative cases, with 1 case showing a unique pattern of microsatellite instability in each of six different metastases examined. Loss of the same allele was found in all metastases in a given patient in 9 of 10 cases. These results indicate that PTEN/MMAC1 gene alterations occur frequently in lethal prostate cancer, although a substantial amount of mutational heterogeneity is found among different metastatic sites within the same patient. These latter findings emphasize the potentially complex genetic relationship that can exist between various clonal lineages of prostate cancer cells as they evolve during the metastatic process and suggest a molecular basis for phenotypic heterogeneity of different prostate cancer foci in patients with disseminated disease.


Assuntos
Deleção de Genes , Genes Supressores de Tumor/genética , Monoéster Fosfórico Hidrolases , Mutação Puntual/genética , Neoplasias da Próstata/genética , Proteínas Tirosina Fosfatases/genética , Proteínas Supressoras de Tumor , Idoso , Idoso de 80 Anos ou mais , Primers do DNA/química , DNA de Neoplasias/análise , Humanos , Hibridização in Situ Fluorescente , Perda de Heterozigosidade/genética , Masculino , Repetições de Microssatélites/genética , Pessoa de Meia-Idade , PTEN Fosfo-Hidrolase , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas
20.
Cancer Res ; 57(21): 4707-9, 1997 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9354426

RESUMO

In a recent study of 91 families having at least three first degree relatives with prostate cancer, we reported the localization of a major susceptibility locus for prostate cancer (HPC1) to chromosome 1 [band q24; J. R. Smith et al., Science (Washington DC), 274: 1371-1373, 1996]. There was significant evidence for locus heterogeneity, with an estimate of 34% of the families being linked to this locus. In this report, we investigate the importance of age at diagnosis of prostate cancer and number of affected individuals within a family as variables in the linkage analysis of an expanded set of markers on 1q24. Under two different models for the prostate cancer locus, we find that the evidence for linkage to HPC1 is provided primarily by large (five or more members affected) families with an early average age at diagnosis. Specifically, for 40 North American families with an average age at diagnosis <65 years, the multipoint lod score is 3.96, whereas for 39 families with an older average age at diagnosis, this value is -0.84. Assuming heterogeneity, the proportion of families linked is 66% for the 14 families with the earliest average ages at diagnoses, but it decreases to 7% for the families with the latest ages at diagnoses. A similar age effect is observed in 12 Swedish pedigrees analyzed. To test the hypotheses generated by these analyses, we examined an additional group of 13 newly identified prostate cancer families. Overall, these families provided additional evidence for linkage to this region (nonparametric linkage Z = 1.91; P = 0.04 at marker D1S1660), contributed primarily by the families in this group with early age at diagnosis [nonparametric linkage Z = 2.50 (P = 0.01) at D1S422]. These results are consistent with the existence of a locus in this region that predisposes men to develop early-onset prostate cancer.


Assuntos
Cromossomos Humanos Par 1/genética , Família , Ligação Genética , Modelos Genéticos , Neoplasias da Próstata/genética , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Suscetibilidade a Doenças , Marcadores Genéticos , Humanos , Masculino , Pessoa de Meia-Idade , Suécia , Estados Unidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...