Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Cancer Gene Ther ; 29(1): 49-61, 2022 01.
Article in English | MEDLINE | ID: mdl-33414516

ABSTRACT

The outcome of prostate cancer (PCa) patients is highly variable and depends on whether or not distant metastases occur. Multiple chromosomal deletions have been linked to early tumor marker PSA recurrence (biochemical relapse, BCR) after radical prostatectomy (RP), but their potential role for distant metastasis formation is largely unknown. Here, we specifically analyzed whether deletion of the tumor suppressor CHD1 (5q21) influences the post-surgical risk of distant metastasis and whether CHD1 loss directly contributes to metastasis formation in vivo. By considering >6800 patients we found that the CHD1 deletion negatively influences metastasis-free survival in R0 patients (HR: 2.32; 95% CI: 1.61, 3.33; p < 0.001) independent of preoperative PSA, pT stage, pN status, Gleason Score, and BCR. Moreover, CHD1 deletion predicts shortened BCR-free survival in pT2 patients and cancer-specific survival in all patients. In vivo, CHD1 loss increases spontaneous pulmonary metastasis formation in two distinct PCa models coupled with a higher number of multicellular colonies as compared to single-cell metastases. Transcriptome analyses revealed down-regulation of the PCa-specific metastasis suppressor and TGFß signaling regulator PMEPA1 after CHD1 depletion in both tested PCa models. CHD1 loss increases the risk of postoperative metastasis in R0-resected PCa patients and promotes spontaneous metastasis formation in vivo.


Subject(s)
DNA Helicases , DNA-Binding Proteins , Prostate-Specific Antigen , Prostatic Neoplasms , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Disease-Free Survival , Genes, Tumor Suppressor , Humans , Male , Membrane Proteins , Neoplasm Grading , Neoplasm Recurrence, Local , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery
3.
EMBO Rep ; 17(11): 1609-1623, 2016 11.
Article in English | MEDLINE | ID: mdl-27596623

ABSTRACT

The CHD1 gene, encoding the chromo-domain helicase DNA-binding protein-1, is one of the most frequently deleted genes in prostate cancer. Here, we examined the role of CHD1 in DNA double-strand break (DSB) repair in prostate cancer cells. We show that CHD1 is required for the recruitment of CtIP to chromatin and subsequent end resection during DNA DSB repair. Our data support a role for CHD1 in opening the chromatin around the DSB to facilitate the recruitment of homologous recombination (HR) proteins. Consequently, depletion of CHD1 specifically affects HR-mediated DNA repair but not non-homologous end joining. Together, we provide evidence for a previously unknown role of CHD1 in DNA DSB repair via HR and show that CHD1 depletion sensitizes cells to PARP inhibitors, which has potential therapeutic relevance. Our findings suggest that CHD1 deletion, like BRCA1/2 mutation in ovarian cancer, may serve as a marker for prostate cancer patient stratification and the utilization of targeted therapies such as PARP inhibitors, which specifically target tumors with HR defects.


Subject(s)
DNA Helicases/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Biomarkers , Carrier Proteins/genetics , Cell Line, Tumor , Chromatin , DNA Breaks, Double-Stranded , DNA Helicases/deficiency , DNA Helicases/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Endodeoxyribonucleases , Humans , Male , Nuclear Proteins/genetics , Poly(ADP-ribose) Polymerase Inhibitors/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Prostatic Neoplasms/genetics , Recombinational DNA Repair
4.
Int J Oncol ; 46(4): 1637-42, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25625310

ABSTRACT

Prostate cancer is characterized by structural rearrangements, most frequently including translocations between androgen-dependent genes and members of the ETS family of transcription factor like TMPRSS2:ERG. In a recent whole genome sequencing study we identified 140 gene fusions that were unrelated to ETS genes in 11 prostate cancers. The aim of the present study was to estimate the prevalence of non-ETS gene fusions. We randomly selected 27 of these rearrangements and analyzed them by fluorescence in situ hybridization (FISH) in a tissue microarray format containing 500 prostate cancers. Using break-apart FISH probes for one fusion partner each, we found rearrangements of 13 (48%) of the 27 analyzed genes in 300-400 analyzable cancers per gene. Recurrent breakage, often accompanied by partial deletion of the genes, was found for NCKAP5, SH3BGR and TTC3 in 3 (0.8%) tumors each, as well as for ARNTL2 and ENOX1 in 2 (0.5%) cancers each. One rearranged tumor sample was observed for each of VCL, ZNF578, IMMP2L, SLC16A12, PANK1, GPHN, LRP1 and ZHX2. Balanced rearrangements, indicating possible gene fusion, were found for ZNF578, SH3BGR, LPR12 and ZHX2 in individual cancers only. The results of the present study confirm that rearrangements involving non-ETS genes occur in prostate cancer, but demonstrate that they are highly individual and typically non-recurrent.


Subject(s)
Chromosome Aberrations , Chromosomes, Human/genetics , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Adult , Aged , Humans , In Situ Hybridization, Fluorescence/methods , Male , Middle Aged , Proto-Oncogene Proteins c-ets/genetics , Tissue Array Analysis/methods
5.
PLoS One ; 9(5): e96533, 2014.
Article in English | MEDLINE | ID: mdl-24801208

ABSTRACT

BACKGROUND & AIMS: HMG-CoA-reductase-inhibitors (statins) have been shown to interfere with HCV replication in vitro. We investigated the mechanism, requirements and contribution of heme oxygenase-1(HO-1)-induction by statins to interference with HCV replication. METHODS: HO-1-induction by fluva-, simva-, rosuva-, atorva- or pravastatin was correlated to HCV replication, using non-infectious replicon systems as well as the infectious cell culture system. The mechanism of HO-1-induction by statins as well as its relevance for interference with HCV replication was investigated using transient or permanent knockdown cell lines. Polyacrylamide(PAA) gels of different density degrees or the Rho-kinase-inhibitor Hydroxyfasudil were used in order to mimic matrix conditions corresponding to normal versus fibrotic liver tissue. RESULTS: All statins used, except pravastatin, decreased HCV replication and induced HO-1 expression, as well as interferon response in vitro. HO-1-induction was mediated by reduction of Bach1 expression and induction of the Nuclear factor (erythroid-derived 2)-like 2 (NRF2) cofactor Krueppel-like factor 2 (KLF2). Knockdown of KLF2 or HO-1 abrogated effects of statins on HCV replication. HO-1-induction and anti-viral effects of statins were more pronounced under cell culture conditions mimicking advanced stages of liver disease. CONCLUSIONS: Statin-mediated effects on HCV replication seem to require HO-1-induction, which is more pronounced in a microenvironment resembling fibrotic liver tissue. This implicates that certain statins might be especially useful to support HCV therapy of patients at advanced stages of liver disease.


Subject(s)
DNA Replication/drug effects , Fatty Acids, Monounsaturated/pharmacology , Heme Oxygenase-1/metabolism , Hepacivirus/drug effects , Indoles/pharmacology , Kruppel-Like Transcription Factors/metabolism , Virus Replication/drug effects , Antiviral Agents/pharmacology , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Line , Fanconi Anemia Complementation Group Proteins/metabolism , Fluvastatin , Humans , NF-E2-Related Factor 2/metabolism , Replicon/drug effects
6.
Eur J Cancer ; 50(4): 852-61, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24380696

ABSTRACT

BACKGROUND: The RNA-binding motif protein 3 (RBM3) has recently been suspected as a prognostic biomarker in several cancers. METHODS: RBM3 expression was analysed by immunohistochemistry on a tissue microarray containing 11,152 prostate cancers. RESULTS: RBM3 expression was more often detectable in malignant compared to benign prostate. RBM3 immunostaining was found in 64% of the interpretable prostate cancers and was considered strong in 25.6%. High RBM3 expression was linked to advanced tumour stage, high Gleason score, positive nodal involvement and positive surgical margin status (p<0.0001 each). There was a remarkable accumulation of strong RBM3 expression in v-ets avian erythroblastosis virus E26 oncogene homolog (ERG) positive prostate cancers and tumours harbouring PTEN deletions (p<0.0001 each). Moreover, RBM3 staining was tightly related to early biochemical recurrence if all tumours or subgroups of ERG negative and ERG positive cancers were analysed (p<0.0001 each). In multivariate analysis, including RBM3 staining, Gleason grade, pT stage, prostate-specific antigen (PSA), surgical margin status, and nodal status, the prognostic impact of RBM3 staining retained statistically significance (p=0.0084). CONCLUSION: Our observations indicate that high RBM3 expression is an independent prognostic marker in prostate cancer. The tight link to ERG activation and PTEN deletions suggest interaction with key molecular pathways in prostate cancer.


Subject(s)
Biomarkers, Tumor/genetics , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/surgery , RNA-Binding Proteins/genetics , Trans-Activators/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Prognosis , Prostatectomy , Prostatic Neoplasms/genetics , RNA-Binding Proteins/metabolism , Transcriptional Activation , Transcriptional Regulator ERG
7.
Am J Pathol ; 184(3): 609-17, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24378408

ABSTRACT

Evidence suggests that class III ß-tubulin (ßIII-tubulin) may represent a prognostic and predictive molecular marker in prostate cancer. ßIII-Tubulin expression was determined by IHC in 8179 prostate cancer specimens in a TMA format. Results were compared with tumor phenotype, biochemical recurrence, v-ets avian erythroblastosis virus E26 oncogene homolog (ERG) status, and deletions on PTEN, 3p13, 5q21, and 6q15. ßIII-Tubulin expression was detectable in 25.6% of 8179 interpretable cancers. High ßIII-tubulin expression was strongly associated with both TMPRSS2:ERG rearrangement and ERG expression (P < 0.0001 each). High ßIII-tubulin expression was tightly linked to high Gleason grade, advanced pT stage, and early prostate-specific antigen (PSA) recurrence in all cancers (P < 0.0001 each), but also in the subgroups of ERG-negative and ERG-positive cancers. When all tumors were analyzed, the prognostic role of ßIII-tubulin expression was independent of Gleason grade, pT stage, pN stage, surgical margin status, and preoperative PSA. Independent prognostic value became even more evident if the analysis was limited to preoperatively available features, such as biopsy specimen Gleason grade, preoperative PSA, cT stage, and ßIII-tubulin expression (P < 0.0001 each). ßIII-Tubulin expression was associated with PTEN (P < 0.0001) when all tumors were analyzed, but also in the subgroups of ERG-negative and ERG-positive cancers. ßIII-Tubulin expression is an independent prognostic parameter. The significant associations with key genomic alterations of prostate cancer, such as TMPRSS2:ERG fusions and PTEN deletions, suggest interactions with several pivotal pathways involved in prostate cancer.


Subject(s)
Biomarkers, Tumor/genetics , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/genetics , Trans-Activators/metabolism , Tubulin/genetics , Aged , Biomarkers, Tumor/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , PTEN Phosphohydrolase/genetics , Phenotype , Prognosis , Prostate-Specific Antigen/metabolism , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Sequence Deletion , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Tissue Array Analysis , Trans-Activators/genetics , Transcriptional Regulator ERG , Tubulin/metabolism
8.
Mod Pathol ; 27(1): 96-106, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23887301

ABSTRACT

Increased levels of karyopherin α2 (KPNA2) expression have been described to be linked to poor prognosis in a variety of malignancies. This study was undertaken to evaluate the clinical impact of KPNA2 expression and its association with key genomic alterations in prostate cancers. A tissue microarray containing samples from 11 152 prostate cancers was analyzed for KPNA2 expression by immunohistochemistry. Results were compared with oncological follow-up data and genomic alterations such as TMPRSS2-ERG fusions and deletions of PTEN, 5q21, 6q15 or 3p13. KPNA2 expression was absent or weak in benign prostatic glands and was found to be in weak, moderate or strong intensities in 68.4% of 7964 interpretable prostate cancers. KPNA2 positivity was significantly linked to the presence of ERG rearrangement (P<0.0001). In ERG-negative and -positive prostate cancers, KPNA2 immunostaining was significantly associated with advanced pathological tumor stage (pT3b/pT4), high Gleason grade and early biochemical recurrence (P<0.0001 each). Multivariate analysis including all established prognostic criteria available after surgery revealed that the prognostic role of KPNA2 (P=0.001) was independent of high Gleason grade, advanced pathological tumor stage, high preoperative prostate-specific antigen level and positive surgical margin status (P<0.0001 each). The comparison of KPNA2 expression with deletions of PTEN, 5q21, 6q15 and 3p13 in ERG-positive and -negative cancers revealed a strong link to PTEN deletions in both subgroups (P<0.0001). In conclusion, the strong independent prognostic impact of KPNA2 expression raises the possibility that measurement of KPNA2 expression alone or in combination with other molecular parameters might possibly result in clinically useful information. The data also emphasize a critical role of the functionality of the nuclear import machinery for prostate cancer biology.


Subject(s)
Biomarkers, Tumor/analysis , Cell Nucleus/chemistry , Kallikreins/analysis , Prostate-Specific Antigen/analysis , Prostatectomy/methods , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/surgery , alpha Karyopherins/analysis , Aged , Biomarkers, Tumor/genetics , Chromosome Deletion , Gene Deletion , Gene Rearrangement , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neoplasm Grading , Neoplasm Staging , PTEN Phosphohydrolase/genetics , Proportional Hazards Models , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Time Factors , Tissue Array Analysis , Trans-Activators/genetics , Transcriptional Regulator ERG , Treatment Outcome
9.
Oncol Lett ; 6(5): 1245-1252, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24179503

ABSTRACT

The cyclin-dependent kinase inhibitor p27Kip1 has been suggested as a prognostic marker in prostate cancer. The aim of this study was to determine the clinical and prognostic role of p27 expression in hormone-naive prostate cancers. A tissue microarray containing samples from 4,699 prostate cancers with attached pathological, clinical follow-up and molecular data was analyzed for nuclear p27 expression by immunohistochemistry. p27 staining was negative in 18.6%, weak in 33.5%, moderate in 28.4% and strong in 19.5% of 3,701 interpretable cancer spots. Loss of p27 immunostaining was linked to tumors of low Gleason grade (P<0.0001) and ERG fusion-negative cancers (P<0.0001). p27 levels were not associated with other parameters, including tumor stage, nodal stage, preoperative prostate-specific antigen (PSA) levels, surgical margin status and cell proliferation (as measured by the Ki67 labeling index). p27 expression was also unrelated to clinical outcome in all cancers, as well as in the subsets of ERG fusion-positive and -negative cancers. Overall, the present data demonstrated that elevated p27 expression was often unrelated to prostate cancer phenotype. Furthermore, the lack of an effect of the p27 protein levels on PSA recurrence following radical prostatectomy indicated that factors other than p27 expression are likely to be the major determinants of prostate cancer recurrence. However, a subset of ERG-negative, low-grade tumors was frequently characterized by loss of p27, suggesting a role of this alteration for the development of these tumors.

10.
Exp Mol Pathol ; 95(2): 227-34, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23948277

ABSTRACT

The extracellular matrix metalloproteinase inducer CD147 has been suggested as a prognostic marker in prostate cancer. CD147 expression was analyzed by immunohistochemistry on a tissue microarray containing 11,152 prostate cancer specimens. Results were compared to tumor phenotype, biochemical recurrence, ERG status and deletions on PTEN, 3p13, 6q15 and 5q21. CD147 expression was strong in benign prostatic glands and often reduced in prostate cancers. CD147 immunostaining was found in 71.7% of 7628 interpretable cases. CD147 staining was considered strong in 34.6%, moderate in 24.3% and weak in 12.8% of cancers while 28.3% did not show any CD147 reactivity. Reduced CD147 staining was strongly associated with both TMPRSS2-ERG-rearrangement and ERG expression (p<0.0001 each). Within the subgroups of ERG positive and negative cancers, deletions of PTEN, 3p13, 6q15 and 5q21 were unrelated to the CD147 expression status. Decreased CD147 expression was significantly linked to high preoperative PSA values, high Gleason grade, advanced tumor stage (p<0.0001 each), and positive lymph node involvement (p=0.0026) in all cancers. There was a marginal, but statistically significant, association of reduced CD147 expression with early biochemical recurrence (p=0.0296). The significant reduction of CD147 expression in ERG positive prostate cancer provides further evidence for marked biological differences between "fusion type" and "non-fusion type" prostate cancer. Despite a weak association with PSA recurrence, CD147 cannot be considered a relevant prognostic biomarker.


Subject(s)
Basigin/biosynthesis , Biomarkers, Tumor/analysis , Neoplasm Recurrence, Local/metabolism , Oncogene Proteins, Fusion/genetics , Prostate-Specific Antigen/blood , Prostatic Neoplasms/metabolism , Adult , Aged , Basigin/analysis , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Tissue Array Analysis
11.
Mol Oncol ; 7(6): 1001-11, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23941784

ABSTRACT

Lysophosphatidylcholine acyltransferase 1 (LPCAT1) has been suggested to play a role in cancer. To assess its role in prostate cancer, LPCAT1 expression was analyzed on a tissue microarray containing samples from 11,152 prostate cancer patients. In benign prostate glands, LPCAT1 immunostaining was absent or weak. In prostate cancer, LPCAT1 positivity was found in 73.8% of 8786 interpretable tumors including 29.2% with strong expression. Increased LPCAT1 expression was associated with advanced tumor stage (pT3b/T4) (p < 0.0001), high Gleason score (≥4 + 4) (p < 0.0001), positive nodal involvement (p = 0.0002), positive surgical margin (p = 0.0005), and early PSA recurrence (p < 0.0001). High LPCAT1 expression was strongly linked to ERG-fusion type prostate cancer. Strong LPCAT1 staining was detected in 45.3% of ERG positive but in only 16.7% of ERG negative tumors (p < 0.0001). Within ERG negative cancers, LPCAT1 staining was strongly increased within the subgroup of PTEN deleted cancers (p < 0.0001). Further subgroup analyses revealed that associations of high LPCAT1 expression with PSA recurrence and unfavorable tumor phenotype were largely driven by ERG negative cancers (p < 0.0001) while these effects were substantially mitigated in ERG positive cancers (p = 0.0073). The prognostic impact of LPCAT1 expression was independent of histological and clinical parameters. It is concluded, that LPCAT1 measurement, either alone or in combination, may be utilized for better clinical decision-making. These data also highlight the potentially important role of lipid metabolism in prostate cancer biology.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/biosynthesis , Biomarkers, Tumor/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Neoplasm Recurrence, Local/enzymology , Prostatic Neoplasms/enzymology , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Aged , Biomarkers, Tumor/genetics , Follow-Up Studies , Humans , Lipid Metabolism/genetics , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Prognosis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/pathology , Retrospective Studies , Risk Factors
12.
Prostate ; 73(15): 1690-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23843146

ABSTRACT

BACKGROUND: The serine peptidase inhibitor, Kazal type 1 (SPINK1) has been suggested to define an aggressive molecular subtype of ERG-fusion negative prostate cancer. It was the aim of this study to further study the clinical relevance of SPINK1 expression and its relationship with other key genomic alterations of prostate cancer. METHODS: A tissue microarray containing more than 10,000 prostate cancers with clinical follow-up was used for immunohistochemical SPINK1 analysis. Data on ERG status as well as PTEN, 6q, 5q, and 3p deletions were available for comparison. RESULTS: SPINK1 expression was absent in benign prostate glands and detectable in 5.9% of 9,503 interpretable prostate cancers. Presence of SPINK1 expression was markedly more frequent in ERG negative (10.4%) than in ERG positive cancers (0.3%; P < 0.0001). However, SPINK1 expression was unrelated to tumor phenotype and biochemical recurrence in all cancers and in the subgroup of ERG negative cancers. Further subgroup analyses revealed, however, that--within ERG negative cancers--SPINK1 expression was significantly linked to deletions at 6q15 (P < 0.0001) and 5q21 (P = 0.0042). CONCLUSIONS: Our results exclude SPINK1 as a relevant prognostic prostate cancer biomarker. However, the data demonstrate that SPINK1 overexpression is tightly linked to the small subsets of 6q15- and 5q21-deleted ERG negative prostate cancers. These findings support the concept of molecularly defined subtypes of prostate cancers.


Subject(s)
Carrier Proteins/genetics , Gene Deletion , Neoplasm Recurrence, Local/genetics , Prostate/metabolism , Prostatic Neoplasms/genetics , Trans-Activators/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , Chromosomes, Human, Pair 5 , Chromosomes, Human, Pair 6 , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Prognosis , Prostate/pathology , Prostate-Specific Antigen/blood , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tissue Array Analysis , Transcriptional Regulator ERG , Trypsin Inhibitor, Kazal Pancreatic
13.
J Pathol ; 231(1): 130-41, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23794398

ABSTRACT

Deletion of 3p13 has been reported from about 20% of prostate cancers. The clinical significance of this alteration and the tumour suppressor gene(s) driving the deletion remain to be identified. We have mapped the 3p13 deletion locus using SNP array analysis and performed fluorescence in situ hybridization (FISH) analysis to search for associations between 3p13 deletion, prostate cancer phenotype and patient prognosis in a tissue microarray containing more than 3200 prostate cancers. SNP array analysis of 72 prostate cancers revealed a small deletion at 3p13 in 14 (19%) of the tumours, including the putative tumour suppressors FOXP1, RYBP and SHQ1. FISH analysis using FOXP1-specific probes revealed deletions in 16.5% and translocations in 1.2% of 1828 interpretable cancers. 3p13 deletions were linked to adverse features of prostate cancer, including advanced stage (p < 0.0001), high Gleason grade (p = 0.0125), and early PSA recurrence (p = 0.0015). In addition, 3p13 deletions were linked to ERG(+) cancers and to PTEN deletions (p < 0.0001 each). A subset analysis of ERG(+) tumours revealed that 3p13 deletions occurred independently from PTEN deletions (p = 0.3126), identifying tumours with 3p13 deletion as a distinct molecular subset of ERG(+) cancers. mRNA expression analysis confirmed that all 3p13 genes were down regulated by the deletion. Ectopic over-expression of FOXP1, RYBP and SHQ1 resulted in decreased colony-formation capabilities, corroborating a tumour suppressor function for all three genes. In summary, our data show that deletion of 3p13 defines a distinct and aggressive molecular subset of ERG(+) prostate cancers, which is possibly driven by inactivation of multiple tumour suppressors.


Subject(s)
Adenocarcinoma/genetics , Chromosome Deletion , Chromosomes, Human, Pair 3/genetics , Genes, Tumor Suppressor , Prostatic Neoplasms/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Cell Line, Tumor , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , Germany/epidemiology , Humans , Kaplan-Meier Estimate , Male , Neoplasm Recurrence, Local , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/metabolism , Polymorphism, Single Nucleotide , Prostate/metabolism , Prostate/pathology , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tissue Array Analysis
14.
Hum Pathol ; 44(9): 1789-96, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23642739

ABSTRACT

Molecular markers reliably predicting the aggressiveness of prostate cancer are currently lacking. Death-domain-associated protein (DAXX) has been implicated in the regulation of chromatin remodeling, transcription, and apoptosis that are integral to oncogenesis and cancer progression. DAXX expression was analyzed by immunohistochemistry on a tissue microarray containing 7478 prostate cancer specimens. Results were compared with tumor phenotype, biochemical recurrence, and v-ets erythroblastosis virus E26 oncogene homolog (ERG) status. DAXX expression was predominantly seen in the nucleus. DAXX expression was detectable in 4609 (80.6%) of 5718 interpretable cancers and considered strong in 5.9%, moderate in 45.8%, and weak in 28.9%. Strong DAXX expression was associated with both transmembrane protease, serine 2 (TMPRSS2)/ERG rearrangement and ERG expression (P < .0001 each). Strong DAXX expression was tightly linked to high Gleason grade, advanced pT stage, increased cell proliferation index, and early prostate-specific antigen recurrence (P < .0001 each). The prognostic role of DAXX expression was independent of Gleason grade, pT stage, and pN stage. Our study establishes DAXX as a novel independent prognosticator in prostate cancer and suggests an important role of DAXX expression for both prostate cancer development and progression. Furthermore, DAXX appears to exert biologically different effects in ERG-positive and ERG-negative prostate cancers.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/pathology , Nuclear Proteins/metabolism , Prostatic Neoplasms/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/surgery , Aged , Biomarkers, Tumor , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Proliferation , Co-Repressor Proteins , Humans , Male , Middle Aged , Molecular Chaperones , Neoplasm Grading , Neoplasm Recurrence, Local , Neoplasm Staging , Prognosis , Prostate-Specific Antigen/metabolism , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/surgery , Tissue Array Analysis , Trans-Activators/metabolism , Transcriptional Regulator ERG
15.
J Clin Pathol ; 66(7): 563-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23559350

ABSTRACT

BACKGROUND AND AIMS: Transcription factors of the forkhead box P (FOXP1-4) family have been implicated in various human cancer types before. The relevance and role of neuronal transcription factor FOXP2 in prostate cancer is unknown. METHODS: A tissue microarray containing samples from more than 11 000 prostate cancers from radical prostatectomy specimens with clinical follow-up data was analysed for FOXP2 expression by immunohistochemistry. FOXP2 data were also compared with pre-existing ERG fusion (by fluorescence in situ hybridisation and immunohistochemistry) and cell proliferation (Ki67 labelling index) data. RESULTS: There was a moderate to strong FOXP2 protein expression in basal and secretory cells of normal prostatic glands. As compared with normal cells, FOXP2 expression was lost or reduced in 25% of cancers. Strong FOXP2 expression was linked to advanced tumour stage, high Gleason score, presence of lymph node metastases and early tumour recurrence (p<0.0001; each) in ERG fusion-negative, but not in ERG fusion-positive cancers. High FOXP2 expression was linked to high Ki67 labelling index (p<0.0001) in all cancers irrespective of ERG fusion status. CONCLUSIONS: These data demonstrate that similar high FOXP2 protein levels as in normal prostate epithelium exert a 'paradoxical' oncogenic role in 'non fusion-type' prostate cancer. It may be speculated that interaction of FOXP2 with members of pathways that are specifically activated in 'non fusion-type' cancers may be responsible for this phenomenon.


Subject(s)
Adenocarcinoma/metabolism , Forkhead Transcription Factors/metabolism , Oncogene Proteins, Fusion/metabolism , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Forkhead Transcription Factors/genetics , Humans , Immunohistochemistry/methods , In Situ Hybridization, Fluorescence , Ki-67 Antigen/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local , Oncogene Proteins, Fusion/genetics , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Risk Factors , Tissue Array Analysis
16.
Cancer Res ; 73(9): 2795-805, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23492366

ABSTRACT

Deletions involving the chromosomal band 5q21 are among the most frequent alterations in prostate cancer. Using single-nucleotide polymorphism (SNP) arrays, we mapped a 1.3 megabase minimally deleted region including only the repulsive guidance molecule B (RGMB) and chromodomain helicase DNA-binding protein 1 (CHD1) genes. Functional analyses showed that CHD1 is an essential tumor suppressor. FISH analysis of 2,093 prostate cancers revealed a strong association between CHD1 deletion, prostate-specific antigen (PSA) biochemical failure (P = 0.0038), and absence of ERG fusion (P < 0.0001). We found that inactivation of CHD1 in vitro prevents formation of ERG rearrangements due to impairment of androgen receptor (AR)-dependent transcription, a prerequisite for ERG translocation. CHD1 is required for efficient recruitment of AR to responsive promoters and regulates expression of known AR-responsive tumor suppressor genes, including NKX3-1, FOXO1, and PPARγ. Our study establishes CHD1 as the 5q21 tumor suppressor gene in prostate cancer and shows a key role of this chromatin remodeling factor in prostate cancer biology.


Subject(s)
DNA Helicases/genetics , DNA-Binding Proteins/genetics , Gene Rearrangement , Genes, Tumor Suppressor , Prostatic Neoplasms/genetics , Trans-Activators/metabolism , Cell Line, Tumor , Chromosomes, Human, Pair 5 , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Male , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Prostate-Specific Antigen/metabolism , Receptors, Androgen/metabolism , Transcriptional Regulator ERG , Translocation, Genetic
17.
Mod Pathol ; 26(7): 975-83, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23370768

ABSTRACT

6q12-22 is the second most commonly deleted genomic region in prostate cancer. Mapping studies have described a minimally deleted area at 6q15, containing MAP3K7/TAK1, which was recently shown to have tumor suppressive properties. To determine prevalence and clinical significance of MAP3K7 alterations in prostate cancer, a tissue microarray containing 4699 prostate cancer samples was analyzed by fluorescence in situ hybridization. Heterozygous MAP3K7 deletions were found in 18.48% of 2289 interpretable prostate cancers. MAP3K7 deletions were significantly associated with advanced tumor stage (P<0.0001), high Gleason grade (P<0.0001), lymph node metastasis (P<0.0108) and early biochemical recurrence (P<0.0001). MAP3K7 alterations were typically limited to the loss of one allele as homozygous deletions were virtually absent and sequencing analyses revealed no evidence for MAP3K7 mutations in 15 deleted and in 14 non-deleted cancers. There was a striking inverse association of MAP3K7 deletions and TMPRSS2:ERG fusion status with 26.7% 6q deletions in 1125 ERG-negative and 11.1% 6q deletions in 1198 ERG-positive cancers (P<0.0001). However, the strong prognostic role of 6q deletions was retained in both ERG-positive and ERG-negative cancers (P<0.0001 each). In summary, our study identifies MAP3K7 deletion as a prominent feature in ERG-negative prostate cancer with strong association to tumor aggressiveness. MAP3K7 alterations are typically limited to one allele of the gene. Together with the demonstrated tumor suppressive function in cell line experiments and lacking evidence for inactivation through hypermethylation, these results indicate MAP3K7 as a gene for which haploinsufficency is substantially tumorigenic.


Subject(s)
MAP Kinase Kinase Kinases/genetics , Neoplasm Recurrence, Local/genetics , Prostatic Neoplasms/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Blotting, Western , Chromosomes, Human, Pair 6 , Gene Deletion , Humans , In Situ Hybridization, Fluorescence , MAP Kinase Kinase Kinases/blood , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Oncogene Proteins, Fusion/genetics , Polymerase Chain Reaction , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Tissue Array Analysis
18.
Cancer Cell ; 23(2): 159-70, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23410972

ABSTRACT

Early-onset prostate cancer (EO-PCA) represents the earliest clinical manifestation of prostate cancer. To compare the genomic alteration landscapes of EO-PCA with "classical" (elderly-onset) PCA, we performed deep sequencing-based genomics analyses in 11 tumors diagnosed at young age, and pursued comparative assessments with seven elderly-onset PCA genomes. Remarkable age-related differences in structural rearrangement (SR) formation became evident, suggesting distinct disease pathomechanisms. Whereas EO-PCAs harbored a prevalence of balanced SRs, with a specific abundance of androgen-regulated ETS gene fusions including TMPRSS2:ERG, elderly-onset PCAs displayed primarily non-androgen-associated SRs. Data from a validation cohort of > 10,000 patients showed age-dependent androgen receptor levels and a prevalence of SRs affecting androgen-regulated genes, further substantiating the activity of a characteristic "androgen-type" pathomechanism in EO-PCA.


Subject(s)
Gene Rearrangement , Genomics , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Serine Endopeptidases/genetics , Trans-Activators/genetics , Adult , Aged , Aged, 80 and over , Computational Biology , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Transcriptional Regulator ERG
19.
Int J Cancer ; 132(6): 1333-40, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-22886792

ABSTRACT

Prevalence and clinical significance of mammalian target of rapamycin (mTOR) phosphorylation at the serine 2448 is disputed in prostate cancer. A tissue microarray containing 3,261 prostate cancers and 49 normal prostate samples with clinical follow-up data was analyzed for p(Ser2448)-mTOR expression by immunohistochemistry. Moderate to strong p(Ser2448)-mTOR staining was found in all (n = 49) normal prostate tissues, but was lost in 24% or weak in 29% cancers. Moderate and strong staining was found in 36 and 11% of tumors. Loss of p(Ser2448)-mTOR staining was significantly linked to advanced stage (p = 0.0027), high-grade (p = 0.0045), nodal positive cancers (p = 0.0483), early tumor recurrence (p < 0.0001, independently from stage and grade, p = 0.0016), lack of Ets-related gene (ERG) fusion (p < 0.0001), reduced androgen receptor expression (p < 0.0001 each) and increased cell proliferation (p = 0.0092) in all cancers and in the subset of ERG-fusion-positive cancers. Loss of p(Ser2448)-mTOR expression was linked to tumor metastasis (p = 0.0275) in ERG-fusion-positive cancers only. Molecular subset analysis using pre-existing phosphatase and tensin homolog (PTEN) deletion data revealed that loss of p(Ser2448) -mTOR expression is of prognostic relevance and defines a subpopulation of PTEN-deleted and ERG-fusion-positive cancers with a particular poor outcome. The results of our study strongly suggest that loss of p(Ser2448)-mTOR expression is a marker for activated AKT/mTOR signaling. Tumors with concomitant PTEN deletion and activated mTOR signaling indicated by loss of p(Ser2448)-mTOR expression characterize a small (4%) but clinically significant subset of prostate cancers that might optimally benefit from anti-mTOR therapies.


Subject(s)
Gene Fusion , Prostatic Neoplasms/etiology , TOR Serine-Threonine Kinases/physiology , Trans-Activators/genetics , Disease Progression , Gene Deletion , Humans , Male , PTEN Phosphohydrolase/genetics , Phosphorylation , Prognosis , Proportional Hazards Models , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , TOR Serine-Threonine Kinases/analysis , Transcriptional Regulator ERG
20.
Mod Pathol ; 26(5): 733-42, 2013 May.
Article in English | MEDLINE | ID: mdl-23196798

ABSTRACT

The aim of this study was to determine whether cysteine-rich secretory protein 3 (CRISP3) expression is linked to clinically or molecularly relevant subgroups of prostate cancer. A tissue microarray representing samples from >10,000 prostate cancers from radical prostatectomy specimens with clinical follow-up data were analyzed for CRISP3 expression by immunohistochemistry. CRISP3 expression was also compared with key genomic alterations of prostate cancer. CRISP3 staining was found as weak in 15%, moderate in 8.5%, and strong in 7.2% of prostate cancers, whereas no expression was detected in normal prostate. Strong CRISP3 expression was linked to advanced tumor stage, high Gleason score, and positive surgical margin status (P<0.0001 each). There was a marked accumulation of high CRISP3 expression in PTEN-deleted ERG-positive tumors (P<0.0001). A total of, 21.7% of ERG-positive and PTEN-deleted cancers had strong CRISP3 expression, but only 10.4% of ERG-positive cancers without PTEN deletion (P<0.0001). The rate of high CRISP3 expression was 2.5% in ERG-negative cancers (P=0.0001; vs ERG-positive cancers). Accordingly, CRISP3 overexpression was associated with early prostate-specific antigen recurrence in all tumors (P=0.0013) as well as in ERG-negative (P=0.004) and ERG-positive cancers (P=0.0318). CRISP3 expression did not retain prognostic significance in models also involving PTEN deletions. Strong CRISP3 expression is associated with unfavorable tumor phenotype and early recurrence in prostate cancers. The tight link of strong CRISP3 expression to the ERG fusion-positive prostate cancers with PTEN deletions provides further evidence for the existence of molecularly distinct subgroups of prostate cancers.


Subject(s)
Biomarkers, Tumor/analysis , Neoplasm Recurrence, Local/metabolism , Prostatic Neoplasms/metabolism , Salivary Proteins and Peptides/biosynthesis , Seminal Plasma Proteins/biosynthesis , Adult , Aged , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Oncogene Proteins, Fusion/genetics , PTEN Phosphohydrolase/genetics , Prognosis , Prostate-Specific Antigen/blood , Prostatic Neoplasms/genetics , Salivary Proteins and Peptides/analysis , Seminal Plasma Proteins/analysis , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...