Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Vet Pathol ; 60(2): 185-189, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36541627

RESUMO

Sebaceous carcinomas of the human ocular adnexa commonly exhibit pagetoid spread, mutations in tumor-suppressor genes, and protooncogene copy number gain. Sebaceous carcinomas are rarely reported in other species, and while the Meibomian gland (MG) represents the most common ocular adnexal structure of the canine eyelid to develop neoplasia, most are clinically and histologically benign. The objective of this study was to compare molecular features of canine MG carcinomas and adenomas. Two retrospectively identified MG carcinomas were subject to immunohistochemistry and qPCR. When compared with normal glands, MYC was upregulated in benign and malignant MG neoplasms. Aberrant p53 expression was restricted to the nuclei of intraepithelial neoplastic cells in MG carcinomas. Adipophilin expression was diminished in MG neoplasms compared with the normal MG. Our findings, if confirmed in a larger cohort of cases, could suggest that MG oncogenesis in a dog may exhibit similar molecular features as their human counterparts.


Assuntos
Adenoma , Carcinoma Basocelular , Doenças do Cão , Neoplasias das Glândulas Sebáceas , Neoplasias Cutâneas , Humanos , Cães , Animais , Glândulas Tarsais/metabolismo , Glândulas Tarsais/patologia , Proteína Supressora de Tumor p53 , Estudos Retrospectivos , Neoplasias das Glândulas Sebáceas/química , Neoplasias das Glândulas Sebáceas/metabolismo , Neoplasias das Glândulas Sebáceas/patologia , Neoplasias das Glândulas Sebáceas/veterinária , Neoplasias Cutâneas/veterinária , Carcinoma Basocelular/veterinária , Transformação Celular Neoplásica , Adenoma/patologia , Adenoma/veterinária , Mutação
2.
J Comp Pathol ; 171: 30-37, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31540623

RESUMO

The terminology applied to canine prostatic epithelial lesions, especially carcinomas, is currently not standardized and this hampers the ability of pathologists to study the biological and clinical significance of these lesions. The aim of this review is to present the essential histomorphological diagnostic attributes of a wide spectrum of prostatic epithelial lesions in dogs. In addition to the traditionally recognized prostatic hyperplasia, hormonal atrophy, prostatitis, squamous metaplasia, adenocarcinoma and transitional cell (urothelial) carcinoma, new entities are described and discussed in order to provide veterinary pathologists with a basic atlas of common histological lesions of the canine prostate that is comprehensive and easy to use.


Assuntos
Doenças do Cão/patologia , Próstata/patologia , Hiperplasia Prostática/veterinária , Neoplasias da Próstata/veterinária , Terminologia como Assunto , Animais , Cães , Masculino , Hiperplasia Prostática/patologia , Neoplasias da Próstata/patologia
3.
J Comp Pathol ; 158: 1-5, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29422309

RESUMO

Proliferative inflammatory atrophy (PIA), which is comprised of highly proliferative but atrophic prostate epithelial cells in association with chronic inflammation, is considered a risk lesion for prostate cancer in men, while its role in canine prostate carcinogenesis is still unknown. We evaluated the value of immunohistochemical labelling for the basal cell marker cytokeratin-5 (CK5) in identifying PIA lesions in 87 samples of formalin-fixed and paraffin wax-embedded canine prostate. Canine PIA showed cytological features identical to the human counterpart and in most cases was associated with chronic lymphoplasmacytic inflammation. PIA lesions were identified in a higher number of CK5-labelled slides (43 out of 87) compared with slides stained by haematoxylin and eosin (HE) (24 out of 87). This lesion was frequently present in normal, hyperplastic and neoplastic canine prostates, although it was underestimated on evaluation of HE-stained slides. Therefore, CK5 can be considered a useful basal cell marker with high sensitivity and specificity for PIA.


Assuntos
Biomarcadores/análise , Doenças do Cão/diagnóstico , Queratina-5/análise , Lesões Pré-Cancerosas/veterinária , Próstata/patologia , Animais , Atrofia , Cães , Masculino
4.
Virchows Arch ; 468(5): 607-17, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26861919

RESUMO

The prognostic value of phosphatase and tensin homolog (PTEN) loss in prostate cancer has primarily been evaluated by either fluorescence in situ hybridization (FISH) or immunohistochemistry (IHC). Previously, we found that PTEN loss by IHC was associated with increased risk of upgrading from biopsy (Gleason 3 + 3) to prostatectomy (Gleason 7+). Now, using an evaluable subset of 111 patients with adjacent biopsy sections, we analyzed the association between PTEN deletion in cancer and the odds of upgrading by a highly sensitive and specific four-color FISH assay. We also compared the concordance of PTEN loss by IHC and PTEN deletion by FISH. PTEN deletion was found in 27 % (12/45) of upgraded cases compared with 11 % (7/66) of controls (P = 0.03). Cancers with PTEN deletions were more likely to be upgraded than those without deletions (adjusting for age odds ratio = 3.40, 95 % confidence interval 1.14-10.11). With respect to concordance, of 93 biopsies with PTEN protein detected by IHC, 89 (96 %) had no PTEN deletion by FISH, and of 18 biopsies without PTEN protein by IHC, 15 had homozygous or hemizygous PTEN deletion by FISH. Only 4 biopsies of the 93 (4 %) with PTEN protein intact had PTEN deletion by FISH. When the regions of uncertainty in these biopsies were systematically studied by FISH, intra-tumoral variation of PTEN deletion was found, which could account for variation in immunoreactivity. Thus, FISH provides a different approach to determining PTEN loss when IHC is uncertain. Both FISH and IHC are concordant, showing consistent positive associations between PTEN loss and upgrading.


Assuntos
Biomarcadores Tumorais/análise , Hibridização in Situ Fluorescente , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias da Próstata/química , Neoplasias da Próstata/patologia , Idoso , Biópsia por Agulha , Humanos , Imuno-Histoquímica/métodos , Hibridização in Situ Fluorescente/métodos , Masculino , Pessoa de Meia-Idade , Prostatectomia/métodos
5.
Prostate Cancer Prostatic Dis ; 18(3): 264-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25939516

RESUMO

BACKGROUND: Biopsies performed for elevated serum PSA often show inflammatory infiltrates. However, the influence of intraprostatic inflammation on serum PSA in men without biopsy indication and negative for prostate cancer has not been described in detail. METHODS: We studied 224 men in the placebo arm of the Prostate Cancer Prevention Trial (PCPT) who underwent end-of-study biopsy per trial protocol, had PSA <4 ng ml(-1), normal digital rectal examination and a biopsy negative for cancer. We analyzed data from hematoxylin and eosin-stained slides containing a mean of three biopsy cores. Inflammation measures included the extent (percentage of tissue area with inflammation) and intensity (product of scores for extent and grade) of total, acute and chronic inflammation in the entire tissue area examined, and by tissue compartment. We calculated median measures of inflammation by prebiopsy serum PSA tertile (>0 to ≤0.8, >0.8 to ≤1.5 and >1.5 to <4.0 ng ml(-1)). We estimated the association between percentage of tissue area with inflammation and natural logarithm of PSA using linear regression adjusting for age at biopsy. RESULTS: Median percentage of tissue area with inflammation increased from 2 to 5 to 9.5% across PSA tertiles (P-trend <0.0001). For every 5% increase in tissue area with inflammation, log PSA increased by 0.061 ng ml(-1) (P=0.0002). Median extent and intensity scores increased across PSA tertiles in luminal and intraepithelial compartments for acute inflammation and in stromal and intraepithelial compartments for chronic inflammation (all P-trend ≤0.05). CONCLUSIONS: In men without clinical suspicion of prostate cancer, greater overall inflammation, luminal and intraepithelial acute inflammation and stromal and intraepithelial chronic inflammation were associated with higher serum PSA.


Assuntos
Inflamação/patologia , Antígeno Prostático Específico/sangue , Próstata/patologia , Neoplasias da Próstata/sangue , Neoplasias da Próstata/patologia , Idoso , Idoso de 80 Anos ou mais , Biópsia , Estudos de Casos e Controles , Estudos Transversais , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Risco
6.
Prostate Cancer Prostatic Dis ; 16(2): 209-15, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23545904

RESUMO

BACKGROUND: ERG rearrangements and PTEN (phosphatase and tensin homolog deleted on chromosome 10) loss are two of the most common genetic alterations in prostate cancer. However, there is still significant controversy regarding the order of events of these two changes during the carcinogenic process. We used immunohistochemistry (IHC) to determine ERG and PTEN status, and calculated the fraction of cases with homogeneous/heterogeneous ERG and PTEN staining in a given tumor. METHODS: Using a single standard tissue section from the index tumor from radical prostatectomies (N=77), enriched for relatively high grade and stage tumors, we examined ERG and PTEN status by IHC. We determined whether ERG or PTEN staining was homogeneous (all tumor cells staining positive) or heterogeneous (focal tumor cell staining) in a given tumor focus. RESULTS: Fifty-seven percent (N=44/77) of tumor foci showed ERG positivity, with 93% of these (N=41/44) cases showing homogeneous ERG staining in which all tumor cells stained positively. Fifty-three percent (N=41/77) of tumor foci showed PTEN loss, and of these 66% (N=27/41) showed heterogeneous PTEN loss. In ERG homogeneously positive cases, any PTEN loss occurred in 56% (N=23/41) of cases, and of these 65% (N=15/23) showed heterogeneous loss. In ERG-negative tumors, 51.5% (N=17/33) showed PTEN loss, and of these 64.7% (N=11/17) showed heterogeneous PTEN loss. In a subset of cases, genomic deletions of PTEN were verified by fluorescence in situ hybridization in regions with PTEN protein loss as compared with regions with intact PTEN protein, which did not show PTEN genomic loss. CONCLUSIONS: These results support the concept that PTEN loss tends to occur as a subclonal event within a given established prostatic carcinoma clone after ERG gene fusion. The combination of ERG and PTEN IHC staining can be used as a simple test to ascertain PTEN and ERG gene rearrangement status within a given prostate cancer in either a research or clinical setting.


Assuntos
Adenocarcinoma/metabolismo , PTEN Fosfo-Hidrolase/genética , Neoplasias da Próstata/metabolismo , Transativadores/genética , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adulto , Idoso , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Progressão da Doença , Deleção de Genes , Homozigoto , Humanos , Imuno-Histoquímica , Perda de Heterozigosidade , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Fusão Oncogênica , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Transativadores/metabolismo , Regulador Transcricional ERG
7.
Oncogene ; 31(10): 1254-63, 2012 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-21822302

RESUMO

Alterations in nucleoli, including increased numbers, increased size, altered architecture and increased function are hallmarks of prostate cancer cells. The mechanisms that result in increased nucleolar size, number and function in prostate cancer have not been fully elucidated. The nucleolus is formed around repeats of a transcriptional unit encoding a 45S ribosomal RNA (rRNA) precursor that is then processed to yield the mature 18S, 5.8S and 28S RNA species. Although it has been generally accepted that tumor cells overexpress rRNA species, this has not been examined in clinical prostate cancer. We find that indeed levels of the 45S rRNA, 28S, 18S and 5.8S are overexpressed in the majority of human primary prostate cancer specimens as compared with matched benign tissues. One mechanism that can alter nucleolar function and structure in cancer cells is hypomethylation of CpG dinucleotides of the upstream rDNA promoter region. However, this mechanism has not been examined in prostate cancer. To determine whether rRNA overexpression could be explained by hypomethylation of these CpG sites, we also evaluated the DNA methylation status of the rDNA promoter in prostate cancer cell lines and the clinical specimens. Bisulfite sequencing of genomic DNA revealed two roughly equal populations of loci in cell lines consisting of those that contained densely methylated deoxycytidine residues within CpGs and those that were largely unmethylated. All clinical specimens also contained two populations with no marked changes in methylation of this region in cancer as compared with normal. We recently reported that MYC can regulate rRNA levels in human prostate cancer; here we show that MYC mRNA levels are correlated with 45S, 18S and 5.8S rRNA levels. Further, as a surrogate for nucleolar size and number, we examined the expression of fibrillarin, which did not correlate with rRNA levels. We conclude that rRNA levels are increased in human prostate cancer, but that hypomethylation of the rDNA promoter does not explain this increase, nor does hypomethylation explain alterations in nucleolar number and structure in prostate cancer cells. Rather, rRNA levels and nucleolar size and number relate more closely to MYC overexpression.


Assuntos
Metilação de DNA , DNA Ribossômico/genética , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , RNA Ribossômico/fisiologia , Adulto , Idoso , Linhagem Celular Tumoral , Ilhas de CpG , Regulação Neoplásica da Expressão Gênica , Genes myc , Humanos , Masculino , Pessoa de Meia-Idade
8.
Br J Cancer ; 105(5): 602-5, 2011 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-21792196

RESUMO

BACKGROUND: We investigated prostate involvement during sexually transmitted infections by measuring serum prostate-specific antigen (PSA) as a marker of prostate infection, inflammation, and/or cell damage in young, male US military members. METHODS: We measured PSA before and during infection for 299 chlamydia, 112 gonorrhoea, and 59 non-chlamydial, non-gonococcal urethritis (NCNGU) cases, and 256 controls. RESULTS: Chlamydia and gonorrhoea, but not NCNGU, cases were more likely to have a large rise (40%) in PSA than controls (33.6%, 19.1%, and 8.2% vs 8.8%, P<0.0001, 0.021, and 0.92, respectively). CONCLUSION: Chlamydia and gonorrhoea may infect the prostate of some infected men.


Assuntos
Antígeno Prostático Específico/sangue , Próstata/fisiologia , Infecções Sexualmente Transmissíveis/etiologia , Adulto , Estudos de Casos e Controles , Infecções por Chlamydia/sangue , Infecções por Chlamydia/epidemiologia , Gonorreia/sangue , Gonorreia/epidemiologia , Humanos , Masculino , Militares/estatística & dados numéricos , Concentração Osmolar , Próstata/microbiologia , Próstata/patologia , Antígeno Prostático Específico/análise , Infecções Sexualmente Transmissíveis/sangue , Infecções Sexualmente Transmissíveis/epidemiologia , Infecções Sexualmente Transmissíveis/transmissão
9.
Oncogene ; 25(40): 5561-9, 2006 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-16619041

RESUMO

Transforming growth factor-beta type 1 (TGF-beta) has been implicated as both a tumor suppressor and a tumor promoter in many solid epithelial cancers. We have previously demonstrated that the cyclin dependent kinase (CDK) inhibitor p21 acts as a molecular switch in determining a growth inhibitory versus growth proliferative response to TGF-beta in the spontaneously immortalized human mammary epithelial cell line MCF-10A. We now demonstrate that this proliferative effect of TGF-beta is mediated through the proinflammatory cytokine, interleukin-1alpha (IL-1alpha). Using gene expression array analysis, we identified IL-1alpha as a cytokine specifically upregulated only in cells lacking p21 and only upon TGF-beta stimulation. Cell proliferation assays verified that recombinant IL-1alpha was capable of inducing a growth proliferative response in p21 null MCF-10A cells, while neutralizing antibodies against IL-1alpha prevented the growth proliferative effects of TGF-beta. Mechanistically, both the CDK and proliferating cell nuclear antigen (PCNA) inhibitory functions of p21 were responsible for preventing TGF-beta induced cell proliferation, but only PCNA inhibition by p21 regulated IL-1alpha gene expression. These studies demonstrate a novel role for IL-1alpha in mediating a proliferative response to TGF-beta signaling, and suggest that therapies directed against IL-1alpha could abate the growth proliferative effects of TGF-beta without compromising its tumor suppressive function.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/genética , Interleucina-1/metabolismo , Sistemas do Segundo Mensageiro , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dactinomicina/farmacologia , Humanos , Interleucina-1/genética , Inibidores da Síntese de Ácido Nucleico/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Regulação para Cima
10.
Urologe A ; 43(5): 573-9, 2004 May.
Artigo em Alemão | MEDLINE | ID: mdl-15029477

RESUMO

BACKGROUND: Prostate cancer is the most commonly diagnosed cancer in men in Europe and North America. Despite its high prevalence, the molecular mechanism of its underlying development and progression is poorly understood. Many studies have revealed multiple molecular alterations during prostate cancer carcinogenesis. GSTP1 CpG island hypermethylation is one of the molecular changes that occur during carcinogenesis. METHODS: We evaluated the role of GSTP1 CpG island hypermethylation in prostatic cancers and discussed its possible role as a molecular biomarker of prostate cancer. RESULTS: Studies haven shown that GSTP1 CpG island hypermethylation is present in about 90% of prostatic carcinomas. The DNA alteration was also detectable in body fluids such as blood, urine, ejaculate, or prostatic secretions. One study showed hypermethylation in histologically unsuspicious lymph nodes in surgical specimens in patients with biochemical PSA (prostate-specific antigen) recurrence. Additionally, it is possible to distinguish between normal prostatic tissue, benign prostatic hyperplasia, and prostate cancer. CONCLUSIONS: The detection of GSTP1 CpG island hypermethylation serves as a molecular marker in prostate cancer screening, detection, and diagnosis. It may even provide information on prostate cancer prognosis. However, prospective trials to evaluate its predictive value are necessary.


Assuntos
Aciltransferases/genética , Biomarcadores Tumorais/genética , Ilhas de CpG/genética , Testes Genéticos/métodos , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/genética , Metilação de DNA , Regulação Neoplásica da Expressão Gênica/genética , Predisposição Genética para Doença/genética , Humanos , Masculino
11.
Cancer Res ; 61(24): 8611-6, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11751372

RESUMO

Among the many somatic genome alterations present in cancer cells, changes in DNA methylation may represent reversible "epigenetic" lesions, rather than irreversible "genetic" alterations. Cancer cell DNA is typically characterized by increases in the methylation of CpG dinucleotides clustered into CpG islands, near the transcriptional regulatory regions of critical genes, and by an overall reduction in CpG dinucleotide methylation. The transcriptional "silencing" of gene expression associated with such CpG island DNA hypermethylation presents an attractive therapeutic target: restoration of "silenced" gene expression may be possible via therapeutic reversal of CpG island hypermethylation. 5-Aza-cytidine (5-aza-C) and 5-aza-deoxycytidine (5-aza-dC), nucleoside analogue inhibitors of DNA methyltransferases, have been widely used in attempts to reverse abnormal DNA hypermethylation in cancer cells and restore "silenced" gene expression. However, clinical utility of the nucleoside analogue DNA methyltransferase inhibitors has been limited somewhat by myelosuppression and other side effects. Many of these side effects are characteristic of nucleoside analogues that are not DNA methyltransferase inhibitors, offering the possibility that nonnucleoside analogue DNA methyltransferase inhibitors might not possess such side effects. Human prostate cancer (PCA) cells characteristically contain hypermethylated CpG island sequences encompassing the transcriptional regulatory region of GSTP1, the gene encoding the pi-class glutathione S-transferase (GSTP1), and fail to express GSTP1 as a consequence of transcriptional "silencing." Inactivation of GSTP1 by CpG island hypermethylation, the most common somatic genome alteration yet reported for human PCAs, occurs early during human prostatic carcinogenesis and results in a loss of GSTP1 "caretaker" function, leaving prostate cells with inadequate defenses against oxidant and electrophile carcinogens. We report here that the drug procainamide, a nonnucleoside inhibitor of DNA methyltransferases, reversed GSTP1 CpG island hypermethylation and restored GSTP1 expression in LNCaP human PCA cells propagated in vitro or in vivo as xenograft tumors in athymic nude mice.


Assuntos
Metilação de DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Glutationa Transferase/genética , Isoenzimas/genética , Procainamida/farmacologia , Neoplasias da Próstata/genética , Animais , Ilhas de CpG/fisiologia , Metilases de Modificação do DNA/antagonistas & inibidores , Ativação Enzimática/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Glutationa S-Transferase pi , Glutationa Transferase/biossíntese , Humanos , Isoenzimas/biossíntese , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/enzimologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Cancer Res ; 61(24): 8617-23, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11751373

RESUMO

Cyclooxygenase-2 (COX-2) is the inducible isoform of the rate-limiting enzymes that convert arachidonic acid to proinflammatory prostaglandins as well as a primary target for nonsteroidal anti-inflammatory drugs. Accumulating evidence suggests that up-regulation of COX-2 is associated with carcinogenesis in multiple organ systems including the large bowel, lung, breast, and prostate. In this report, we examine the expression of COX-2 protein and mRNA in prostate tissue containing various lesions and in prostate cancer cell lines. In the cell lines, LNCaP, DU145, PC-3, and TSU, COX-2 protein expression was undetectable under basal conditions but could be induced transiently by phorbol ester treatment in PC-3 and TSU cells, but not in DU145 and LNCaP cells. Immunohistochemical analysis of 144 human prostate cancer cases suggested that, in contrast to several previous reports, there was no consistent overexpression of COX-2 in established prostate cancer or high-grade prostatic intraepithelial neoplasia, as compared with adjacent normal prostate tissue. Positive staining was seen only in scattered cells (<1%) in both tumor and normal tissue regions but was much more consistently observed in areas of proliferative inflammatory atrophy, lesions that have been implicated in prostatic carcinogenesis. Staining was also seen at times in macrophages. Western blotting and quantitative RT-PCR analyses confirmed these patterns of expression. These results suggest that if nonsteroidal anti-inflammatory drugs are indeed chemopreventive and/or chemotherapeutic for prostate cancer, their effects are likely to be mediated by modulating COX-2 activity in non-PCa cells (either inflammatory cells or atrophic epithelial cells) or by affecting a COX-2-independent pathway.


Assuntos
Isoenzimas/biossíntese , Prostaglandina-Endoperóxido Sintases/biossíntese , Próstata/patologia , Neoplasias da Próstata/enzimologia , Atrofia/enzimologia , Western Blotting , Ciclo-Oxigenase 2 , Progressão da Doença , Epitélio/enzimologia , Epitélio/patologia , Humanos , Imuno-Histoquímica , Isoenzimas/genética , Masculino , Proteínas de Membrana , Prostaglandina-Endoperóxido Sintases/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/enzimologia , Células Estromais/patologia , Células Tumorais Cultivadas , Regulação para Cima
13.
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
14.
Urology ; 58(4): 619-24, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11597556

RESUMO

OBJECTIVES: To examine the expression of the p63 protein in normal, preneoplastic, and neoplastic human prostatic tissue. The p63 gene, a member of the p53 gene family, is expressed in the basal epithelial cells of multiple organs. Irregularities in p63 expression have been associated with epithelial carcinogenesis. METHODS: We performed immunohistochemistry with an anti-p63 antibody on specimens from radical prostatectomies, prostate needle biopsies, and metastatic prostate adenocarcinoma. We analyzed p63 expression in regions of normal prostate, benign prostatic hyperplasia, proliferative inflammatory atrophy (PIA), high-grade intraepithelial neoplasia, and adenocarcinoma. RESULTS: Basal epithelial cells in normal, benign prostatic hyperplasia, and high-grade intraepithelial neoplasia tissue stained intensely for the p63 polypeptide, but the vast majority of adenocarcinoma specimens from 233 patients-66 (94%) of 70 radical prostatectomies, 132 (89%) of 148 prostate needle biopsies, and 14 (93%) of 15 metastases-did not. In tumors in which the adenocarcinoma cells were positive, the staining intensity was weak and occurred in less than 1% of the cells. Tumors that stained positive for p63 were more likely to be high grade than those that did not (P <0.0001). Basal cells in PIA expressed p63, but these cells were sparsely distributed relative to the basal cells in the normal glands. Luminal cells in PIA were, in general, negative for p63. CONCLUSIONS: In contrast to normal and preneoplastic prostatic tissue, the vast majority of prostate adenocarcinomas do not express p63. Therefore, p63 immunohistochemistry represents a potential novel adjuvant method for facilitating the pathologic diagnosis of prostate cancer in prostate needle biopsies. The selective expression of p63 in normal basal cells, coupled with the finding that p63 null mice fail to develop prostates, provides strong evidence that the basal cells represent prostatic epithelial stem cells. In addition, these findings suggest that p63 may protect prostatic epithelial cells against neoplastic transformation and support the hypothesis that intermediately differentiated cells in the luminal epithelium of PIA are the targets of neoplastic transformation in the prostate.


Assuntos
Adenocarcinoma/química , Proteínas de Membrana , Fosfoproteínas/análise , Neoplasias da Próstata/química , Transativadores/análise , Adenocarcinoma/diagnóstico , Adenocarcinoma/secundário , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia por Agulha , Neoplasias Ósseas/secundário , Diferenciação Celular , Proteínas de Ligação a DNA , Epitélio/química , Genes Supressores de Tumor , Humanos , Imuno-Histoquímica , Queratinas/análise , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Lesões Pré-Cancerosas/diagnóstico , Próstata/química , Hiperplasia Prostática/patologia , Neoplasias da Próstata/diagnóstico , Células-Tronco/patologia , Fatores de Transcrição , Proteínas Supressoras de Tumor
15.
Am J Pathol ; 159(3): 837-43, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11549576

RESUMO

With the completion of the Human Genome Project and high-throughput screening methods using cDNA array and tissue microarray (TMA) technology, there is a pressing need to manage the voluminous data sets generated from these types of investigations. Herein is described a database model to handle 1) clinical and pathology data, 2) TMA location information, and 3) web-based histology results. The model is useful for managing clinical, pathology, and molecular data on >1300 prostate cancer patients dating back to 1995 from the University of Michigan Specialized Program of Research Excellence for prostate cancer. The key components in this multidatabase model are 1) the TMA database, 2) the TMA-image database (TMA-I DB), and 3) the prostate pathology and clinical information databases. All databases were created in Microsoft Access (Microsoft, Redmond, WA). Desired patient, tissue, block, diagnosis, array location, and respective clinical and pathology information is obtained by linking the unique identifier fields among database tables. The TMA database is comprised of interrelated data from 336 prostate cancer patients transferred into 19 TMA blocks with 5451 TMA biopsy cores. Tissue samples include 1695 normal prostate, 3171 prostate cancer, 464 prostatic intraepithelial neoplasia, and 121 atrophy. All 19 TMA blocks have been analyzed over the Internet for several immunohistochemical biomarkers including E-cadherin, prostate-specific antigen, p27(Kip1), and Ki-67 labeling index. This system facilitates the statistical analysis of high-density TMA data with clinical and pathology information in an efficient and cost-effective manner. Because the review is performed over the Internet, this system is ideal for collaborative multi-institutional studies.


Assuntos
Bases de Dados como Assunto , Análise de Sequência com Séries de Oligonucleotídeos , Patologia/métodos , Próstata/patologia , Neoplasias da Próstata/patologia , Humanos , Masculino
16.
Prostate ; 49(1): 30-7, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11550208

RESUMO

BACKGROUND: Glutathione S-transferases (GSTs), inducible enzymes that catalyze the detoxification of reactive electrophiles and oxidants, protect against neoplastic transformation. Prostatic adenocarcinoma and high-grade prostatic intraepithelial neoplasia (HGPIN) fail to express GSTP1, a major class of GST. This failure of expression is associated with methlyation of the GSTP1 promoter, a somatic alteration proposed to be a critical step in prostatic carcinogenesis. However, simple atrophy and post-atrophic hyperplasia-proliferative lesions associated with chronic inflammation, which we have termed "proliferative inflammatory atrophy" (PIA)-express elevated levels of GSTP1. We postulated that this increase in GSTP1 expression in PIA occurs in response to increased oxidative stress. We examined the expression of another major class of GST, GSTA1, in the human prostate. METHODS: We performed immunohistochemistry against GSTA1 on formalin-fixed radical prostatectomies (n = 45). A stereological grid point counting method was used to estimate the percent of cells staining positive for GSTA1 in normal prostate, PIA, HGPIN, and adenocarcinoma. RESULTS: In contrast to GSTP1, normal peripheral zone epithelium was virtually devoid of GSTA1. Strikingly, though, epithelial cells in PIA demonstrated strong staining for GSTA1 (median of percent of cells staining positive = 44) as compared to those in normal peripheral zone (median = 3.0, P <.00001), HGPIN (median = 2.9, P <.00001), and adenocarcinoma (median = 3.8, P <.00001). Variations in GSTA1 were also detected between normal anatomic zones: the central zone showed an increase in the percentage of cells staining positive (median = 20.9) as compared to the transition (median = 0.47, P <.0002) and the peripheral (P <.0001) zones. CONCLUSIONS: Expression of GSTA1 is increased in PIA, supporting the concept that cells within these lesions are subject to localized increases in oxidative stress. Low levels of GSTA1 and GSTP1 in HGPIN and adenocarcinoma suggest a broad lack of detoxification activity in these cells, which may be associated with carcinogenesis in the prostate.


Assuntos
Adenocarcinoma/patologia , Regulação Neoplásica da Expressão Gênica , Glutationa Transferase/biossíntese , Estresse Oxidativo/fisiologia , Próstata/fisiologia , Neoplasias da Próstata/patologia , Adenocarcinoma/cirurgia , Glutationa S-Transferase pi , Glutationa Transferase/genética , Humanos , Imuno-Histoquímica , Isoenzimas/biossíntese , Isoenzimas/genética , Masculino , Lesões Pré-Cancerosas/patologia , Prostatectomia , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/cirurgia
17.
Hematol Oncol Clin North Am ; 15(3): 407-21, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11525288

RESUMO

The field of prostate cancer research is poised for dramatic improvements in our ability to better diagnose men at risk of prostate cancer and to better predict prognosis and response to treatment. Histopathologic and molecular analyses lie at the heart of these issues. Improvements in our understanding of the mechanisms of prostate carcinogenesis and in determining why the prostate seems to be so highly targeted for cancer development will lead to rational strategies of disease prevention.


Assuntos
Adenocarcinoma/patologia , Próstata/patologia , Neoplasias da Próstata/patologia , Adenocarcinoma/química , Adenocarcinoma/diagnóstico , Adenocarcinoma/epidemiologia , Adenocarcinoma/terapia , Idade de Início , Idoso , Atrofia , Biomarcadores Tumorais/análise , Biópsia por Agulha/métodos , Humanos , Hiperplasia , Incidência , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Proteínas de Neoplasias/análise , Prognóstico , Próstata/química , Antígeno Prostático Específico/sangue , Neoplasia Prostática Intraepitelial/química , Neoplasia Prostática Intraepitelial/diagnóstico , Neoplasia Prostática Intraepitelial/epidemiologia , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/química , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/epidemiologia , Neoplasias da Próstata/terapia , Prostatite/sangue , Prostatite/patologia , Coloração e Rotulagem/métodos
18.
Prostate ; 48(3): 128-30, 2001 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-11494327
19.
Urology ; 57(4 Suppl 1): 39-45, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11295593

RESUMO

Prostate cancer has become 1 of the most commonly diagnosed cancers in the United States and 1 of the leading causes of cancer death in North America and Western Europe. Survey studies of prostate tissues obtained at autopsy indicate that the development of life-threatening prostate cancer in the US likely occurs over decades. Insights from epidemiologic studies implicate environmental factors, principally dietary components, as major risk factors for prostate cancer development. An accumulating body of basic research data suggests that normal and neoplastic prostate cells may be subjected to a relentless barrage of genome-damaging stresses, and that dietary components and male sex steroids might modulate the level of genome threatening insults. Finally, over the past 5 years, analyses of somatic genome alterations in prostatic carcinoma cells have revealed that somatic inactivation of GSTP1, encoding the carcinogen-detoxification enzyme glutathione S-transferase pi, may serve as an initiating genome lesion for prostatic carcinogenesis. These diverse observations can be integrated into a transcendent mechanistic hypothesis for the pathogenesis of prostate cancer: normal prostate cells acquiring somatic GSTP1 defects may suffer chronic genome damage, influenced by dietary practices, that promote neoplastic transformation, while prostatic carcinoma cells, which characteristically contain defective GSTP1 alleles, remain susceptible to further genome-damaging stresses that promote malignant cancer progression. This hypothesized critical role for GSTP1 inactivation in the earliest steps of prostatic carcinogenesis provides several attractive opportunities for prostate cancer prevention strategies, including (1) restoration of GSTP1 function, (2) compensation for inadequate GSTP1 activity (via use of therapeutic inducers of other glutathione S-transferases (GST), and (3) abrogation or attenuation of genome-damaging stresses.


Assuntos
Glutationa Transferase/genética , Isoenzimas/genética , Neoplasias da Próstata/etiologia , Atrofia , Transformação Celular Neoplásica , Dano ao DNA , Metilação de DNA , Dieta/efeitos adversos , Inativação Gênica , Glutationa S-Transferase pi , Humanos , Masculino , Pessoa de Meia-Idade , Próstata/patologia , Neoplasia Prostática Intraepitelial/genética , Neoplasias da Próstata/epidemiologia , Neoplasias da Próstata/prevenção & controle , Fatores de Risco , Fatores de Tempo , Estados Unidos/epidemiologia
20.
Urology ; 57(4 Suppl 1): 103-14, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11295606

RESUMO

The development of drugs to prevent prostate cancer is underway, yet monitoring the potential efficacy of these agents during clinical trials relies on measuring intermediate endpoints. In this review, various candidate markers are presented that are under different stages of evaluation as intermediate endpoint biomarkers. In addition, the near future will bring an unprecedented wave of new potential biomarkers. For instance, through genomics-based methods many new genes are being discovered whose altered expression may be involved in different phases of prostate cancer development and progression. In the development of rational approaches for selecting which of these untested biomarkers may be useful to measure systematically, there must be an improved understanding of the mechanisms of prostatic carcinogenesis. We submit that this improved understanding will come through new knowledge of the biology of normal prostate epithelial cells, the determination of the precise target cells of transformation, and how their growth regulation is genetically and epigenetically perturbed during the phases of initiation and progression. In this review, therefore, we also present our recent immune-mediated oxidant injury and regeneration hypothesis of why and how the prostate is targeted for carcinogenesis.


Assuntos
Biomarcadores Tumorais , Mutação em Linhagem Germinativa , Neoplasia Prostática Intraepitelial/etiologia , Neoplasias da Próstata/etiologia , Apoptose , Atrofia , Biomarcadores Tumorais/análise , Previsões , Humanos , Masculino , Lesões Pré-Cancerosas/etiologia , Lesões Pré-Cancerosas/patologia , Próstata/patologia , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/química , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...