Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Genes Chromosomes Cancer ; 55(8): 617-25, 2016 08.
Article in English | MEDLINE | ID: mdl-27074291

ABSTRACT

Amplification of the 9p13.3 chromosomal region occurs in a subset of prostate cancers (PCs); however, the target gene or genes of this amplification have remained unidentified. The aim of this study was to investigate the 9p13.3 amplification in more detail to identify genes that are potentially advantageous for cancer cells. We narrowed down the minimally amplified area and assessed the frequency of the 9p13.3 amplification. Of the clinical samples from untreated PCs that were examined (n = 134), 9.7% showed high-level amplification, and 32.1% showed low-level amplification. Additionally, in clinical samples from castration-resistant PCs (n = 70), high- and low-level amplification was seen in 14.3% and 44.3% of the samples, respectively. We next analyzed the protein-coding genes in this chromosomal region for both their expression in clinical PC samples as well as their potential as growth regulators in PC cells. We found that the 9p13.3 amplification harbors several genes that are able to affect the growth of PC cells when downregulated using siRNA. Of these, UBAP2 was the most prominently upregulated gene in the clinical prostate tumor samples. © 2016 Wiley Periodicals, Inc.


Subject(s)
Carrier Proteins/genetics , Chromosomes, Human, Pair 9/genetics , Gene Amplification/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms/genetics , Aged , Animals , Cell Line, Tumor , Disease-Free Survival , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Middle Aged , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/pathology , Xenograft Model Antitumor Assays
2.
PLoS One ; 11(3): e0150241, 2016.
Article in English | MEDLINE | ID: mdl-26939004

ABSTRACT

Microseminoprotein-beta (MSMB, MSMB) is an abundant secretory protein contributed by the prostate, and is implicated as a prostate cancer (PC) biomarker based on observations of its lower expression in cancerous cells compared with benign prostate epithelium. However, as the current literature on MSMB is inconsistent, we assessed the expression of MSMB at the protein and mRNA levels in a comprehensive set of different clinical stages of PC. Immunohistochemistry using monoclonal and polyclonal antibodies against MSMB was used to study protein expression in tissue specimens representing prostatectomies (n = 261) and in diagnostic needle biopsies from patients treated with androgen deprivation therapy (ADT) (n = 100), and in locally recurrent castration-resistant PC (CRPC) (n = 105) and CRPC metastases (n = 113). The transcript levels of MSMB, nuclear receptor co-activator 4 (NCOA4) and MSMB-NCOA4 fusion were examined by qRT-PCR in prostatectomy samples and by RNA-sequencing in benign prostatic hyperplasia, PC, and CRPC samples. We also measured serum MSMB levels and genotyped the single nucleotide polymorphism rs10993994 using DNA from the blood of 369 PC patients and 903 controls. MSMB expression in PC (29% of prostatectomies and 21% of needle biopsies) was more frequent than in CRPC (9% of locally recurrent CRPCs and 9% of CRPC metastases) (p<0.0001). Detection of MSMB protein was inversely correlated with the Gleason score in prostatectomy specimens (p = 0.024). The read-through MSMB-NCOA4 transcript was detected at very low levels in PC. MSMB levels in serum were similar in cases of PC and controls but were significantly associated with PC risk when adjusted for age at diagnosis and levels of free or total PSA (p<0.001). Serum levels of MSMB in both PC patients and controls were significantly associated with the rs10993994 genotype (p<0.0001). In conclusion, decreased expression of MSMB parallels the clinical progression of PC and adjusted serum MSMB levels are associated with PC risk.


Subject(s)
Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms/blood , Prostatic Neoplasms/metabolism , Prostatic Secretory Proteins/blood , Prostatic Secretory Proteins/metabolism , Androgens/metabolism , Biopsy, Needle , Cell Line, Tumor , Disease-Free Survival , Gene Expression Regulation, Neoplastic , Genotype , Humans , Immunohistochemistry , Male , Neoplasm Metastasis , Neoplasm Proteins/blood , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local , Nuclear Receptor Coactivators/metabolism , Polymorphism, Single Nucleotide , Prostatectomy , Prostatic Hyperplasia/metabolism , Sequence Analysis, RNA
3.
J Oral Pathol Med ; 45(5): 329-37, 2016 May.
Article in English | MEDLINE | ID: mdl-26436875

ABSTRACT

BACKGROUND: CIP2A, an inhibitor of PP2A tumour suppressor function, is a widely overexpressed biomarker of aggressive disease and poor therapy response in multiple human cancer types. METHODS: CIP2A and DPPA4 copy number alterations and expression were analysed by fluorescence in situ hybridisation (FISH) and immunohistochemistry (IHC) in different cell lines and a tissue microarray of 52 HNSCC patients. Results were correlated with patient survival and other clinicopathological data. RESULTS: CIP2A and DPPA4 copy number increase occurred at a relatively high frequency in human HNSCC patient samples. CIP2A but not DPPA4 FISH status was significantly associated with patient survival. CIP2A detection by combining IHC with FISH yielded superior resolution in the prognostication of HNSCC. CONCLUSIONS: CIP2A copy number increase is associated with poor patient survival in human HNSCC. We suggest that the reliability and prognostic value of CIP2A detection can be improved by performing FISH analysis to CIP2A IHC positive tumours.


Subject(s)
Autoantigens/biosynthesis , Autoantigens/genetics , Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Disease-Free Survival , Female , Gene Dosage , HeLa Cells , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence/methods , Intracellular Signaling Peptides and Proteins , Male , Middle Aged , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Prognosis , Reproducibility of Results , Squamous Cell Carcinoma of Head and Neck , Tissue Array Analysis/methods
4.
Oncotarget ; 6(23): 19661-70, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-25965834

ABSTRACT

Residual androgen receptor (AR)-signaling and presence of cancer stem-like cells (SCs) are the two emerging paradigms for clinically challenging castration-resistant prostate cancer (CRPC). Therefore, identification of AR-target proteins that are also overexpressed in the cancer SC population would be an attractive therapeutic approach.Our analysis of over three hundred clinical samples and patient-derived prostate epithelial cultures (PPECs), revealed Cancerous inhibitor of protein phosphatase 2A (CIP2A) as one such target. CIP2A is significantly overexpressed in both hormone-naïve prostate cancer (HN-PC) and CRPC patients . CIP2A is also overexpressed, by 3- and 30-fold, in HN-PC and CRPC SCs respectively. In vivo binding of the AR to the intronic region of CIP2A and its functionality in the AR-moderate and AR-high expressing LNCaP cell-model systems is also demonstrated. Further, we show that AR positively regulates CIP2A expression, both at the mRNA and protein level. Finally, CIP2A depletion reduced cell viability and colony forming efficiency of AR-independent PPECs as well as AR-responsive LNCaP cells, in which anchorage-independent growth is also impaired.These findings identify CIP2A as a common denominator for AR-signaling and cancer SC functionality, highlighting its potential therapeutic significance in the most clinically challenging prostate pathology: castration-resistant prostate cancer.


Subject(s)
Autoantigens/metabolism , Membrane Proteins/metabolism , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/therapy , Androgen Antagonists/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Autoantigens/genetics , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cell Survival , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Humans , Intracellular Signaling Peptides and Proteins , Introns , Male , Membrane Proteins/genetics , Molecular Targeted Therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Binding , RNA Interference , RNA, Messenger/metabolism , Receptors, Androgen/drug effects , Receptors, Androgen/metabolism , Signal Transduction , Time Factors , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Up-Regulation
5.
Oncotarget ; 6(8): 6235-50, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25749039

ABSTRACT

Prostate cancer is the third most common cause of male cancer death in developed countries, and one of the most comprehensively characterized human cancers. Roughly 60% of prostate cancers harbor gene fusions that juxtapose ETS-family transcription factors with androgen regulated promoters. A second subtype, characterized by SPINK1 overexpression, accounts for 15% of prostate cancers. Here we report the discovery of a new prostate cancer subtype characterized by rearrangements juxtaposing the SMAD inhibitor SKIL with androgen regulated promoters, leading to increased SKIL expression. SKIL fusions were found in 6 of 540 (1.1%) prostate cancers and 1 of 27 (3.7%) cell lines and xenografts. 6 of 7 SKIL-positive cancers were negative for ETS overexpression, suggesting mutual exclusivity with ETS fusions. SKIL knockdown led to growth arrest in PC-3 and LNCaP cell line models of prostate cancer, and its overexpression led to increased invasiveness in RWPE-1 cells. The role of SKIL as a prostate cancer oncogene lends support to recent studies on the role of TGF-ß signaling as a rate-limiting step in prostate cancer progression. Our findings highlight SKIL as an oncogene and potential therapeutic target in 1-2% of prostate cancers, amounting to an estimated 10,000 cancer diagnoses per year worldwide.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Prostatic Neoplasms/classification , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins/genetics , Animals , Cell Line, Tumor , Cohort Studies , Gene Knockdown Techniques , Gene Rearrangement , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Middle Aged , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Transcriptome , Transfection
6.
Prostate ; 74(16): 1647-54, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25252136

ABSTRACT

BACKGROUND: TMPRSS2-ERG gene fusion is the most frequent genetic alteration in prostate cancer. However, information about its distribution in lymph node positive prostate cancers and the prognostic significance in these advanced tumors is unknown. METHODS: Gene fusion status was determined by fluorescence in situ hybridization on a tissue-microarray constructed from 119 hormone-naïve nodal positive, surgically treated prostate cancers containing samples from the primary tumors and corresponding lymph node metastases. Data were correlated with various tumor features (Gleason score, stage, cancer volume, nodal tumor burden) and biochemical recurrence-free, disease-specific, and overall survival. RESULTS: TMPRSS2-ERG fusion was detected in 43.5% of the primary tumors. Conversely, only 29.9% of the metastasizing components showed the fusion. Concordance in TMPRSS2-ERG status between primary tumors and metastases was 70.9% (Kappa 0.39); 20.9% and 8.1% of the patients showed the mutation solely in their primary tumors and metastases, respectively. TMPRSS2-ERG fusion was not correlated with specific histopathological tumor features but predicted favorable biochemical recurrence-free, disease-specific and overall survival independently when present in the primary tumor (P < 0.05 each). CONCLUSION: TMPRSS2-ERG fusion is more frequent in primary prostate cancer than in corresponding metastases suggesting no selection of fusion-positive cells in the metastatic process. The gene fusion in primary tumors independently predicts favorable outcome.


Subject(s)
Gene Fusion/genetics , Lymphatic Metastasis/genetics , Prostatic Neoplasms/genetics , Serine Endopeptidases/genetics , Trans-Activators/genetics , Aged , Cohort Studies , Follow-Up Studies , Genotype , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Male , Middle Aged , Prevalence , Prognosis , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Retrospective Studies , Serine Endopeptidases/metabolism , Survival Rate , Tissue Array Analysis , Trans-Activators/metabolism , Transcriptional Regulator ERG
7.
BMC Res Notes ; 7: 547, 2014 Aug 18.
Article in English | MEDLINE | ID: mdl-25135188

ABSTRACT

BACKGROUND: The aim of the study was to characterize a recurrent amplification at chromosomal region 1p21-22 in bladder cancer. METHODS: ArrayCGH (aCGH) was performed to identify DNA copy number variations in 7 clinical samples and 6 bladder cancer cell lines. FISH was used to map the amplicon at 1p21-22 in the cell lines. Gene expression microarrays and qRT-PCR were used to study the expression of putative target genes in the region. RESULTS: aCGH identified an amplification at 1p21-22 in 10/13 (77%) samples. The minimal region of the amplification was mapped to a region of about 1 Mb in size, containing a total of 11 known genes. The highest amplification was found in SCaBER squamous cell carcinoma cell line. Four genes, TMED5, DR1, RPL5 and EVI5, showed significant overexpression in the SCaBER cell line compared to all the other samples tested. Oncomine database analysis revealed upregulation of DR1 in superficial and infiltrating bladder cancer samples, compared to normal bladder. CONCLUSIONS: In conclusions, we have identified and mapped chromosomal amplification at 1p21-22 in bladder cancer as well as studied the expression of the genes in the region. DR1 was found to be significantly overexpressed in the SCaBER, which is a model of squamous cell carcinoma. However, the overexpression was found also in a published clinical sample cohort of superficial and infiltrating bladder cancers. Further studies with more clinical material are needed to investigate the role of the amplification at 1p21-22.


Subject(s)
Chromosome Mapping , Chromosomes, Human, Pair 1 , Urinary Bladder Neoplasms/genetics , Base Sequence , Cell Line, Tumor , Comparative Genomic Hybridization , DNA Primers , Humans , In Situ Hybridization, Fluorescence , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Urinary Bladder Neoplasms/pathology
8.
Cancer Epidemiol Biomarkers Prev ; 22(12): 2333-44, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24083995

ABSTRACT

BACKGROUND: The associations of ERG overexpression with clinical behavior and molecular pathways of prostate cancer are incompletely known. We assessed the association of ERG expression with AR, PTEN, SPINK1, Ki-67, and EZH2 expression levels, deletion, and mutations of chromosomal region 3p14 and TP53, and clinicopathologic variables. METHODS: The material consisted of 326 prostatectomies, 166 needle biopsies from men treated primarily with endocrine therapy, 177 transurethral resections of castration-resistant prostate cancers (CRPC), and 114 CRPC metastases obtained from 32 men. Immunohistochemistry, FISH, and sequencing was used for the measurements. RESULTS: ERG expression was found in about 45% of all patient cohorts. In a multivariate analysis, ERG expression showed independent value of favorable prognosis (P = 0.019). ERG positivity was significantly associated with loss of PTEN expression in prostatectomy (P = 0.0348), and locally recurrent CRPCs (P = 0.0042). Loss of PTEN expression was associated (P = 0.0085) with shorter progression-free survival in ERG-positive, but not in negative cases. When metastases in each subject were compared, consistent ERG, PTEN, and AR expression as well as TP53 mutations were found in a majority of subjects. CONCLUSIONS: A similar frequency of ERG positivity from early to late stage of the disease suggests lack of selection of ERG expression during disease progression. The prognostic significance of PTEN loss solely in ERG-positive cases indicates interaction of these pathways. The finding of consistent genetic alterations in different metastases suggests that the major genetic alterations take place in the primary tumor. IMPACT: Interaction of PTEN and ERG pathways warrants further studies.


Subject(s)
Biomarkers, Tumor/metabolism , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms, Castration-Resistant/enzymology , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Trans-Activators/biosynthesis , Cohort Studies , Disease-Free Survival , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Neoplasm Metastasis , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Paraffin Embedding , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/surgery , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/surgery , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptional Regulator ERG , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
PLoS One ; 6(10): e26595, 2011.
Article in English | MEDLINE | ID: mdl-22028916

ABSTRACT

ARLTS1 is a recently characterized tumor suppressor gene at 13q14.3, a region frequently deleted in both sporadic and hereditary prostate cancer (PCa). ARLTS1 variants, especially Cys148Arg (T442C), increase susceptibility to different cancers, including PCa. In this study the role of Cys148Arg substitution was investigated as a risk factor for PCa using both genetic and functional analysis. Cys148Arg genotypes and expression of the ARLTS1 were explored in a large set of familial and unselected PCa cases, clinical tumor samples, xenografts, prostate cancer cell lines and benign prostatic hyperplasia (BPH) samples. The frequency of the variant genotype CC was significantly higher in familial (OR = 1.67, 95% CI = 1.08-2.56, P = 0.019) and unselected patients (OR = 1.52, 95% CI = 1.18-1.97, P = 0.001) and the overall risk was increased (OR = 1.54, 95% CI = 1.20-1.98, P = 0.0007). Additional analysis with clinicopathological data revealed an association with an aggressive disease (OR = 1.28, 95% CI = 1.05-∞, P = 0.02). The CC genotype of the Cys148Arg variant was also contributing to the lowered ARLTS1 expression status in lymphoblastoid cells from familial patients. In addition significantly lowered ARLTS1 expression was observed in clinical tumor samples compared to BPH samples (P = 0.01). The ARLTS1 co-expression signature based on previously published microarray data was generated from 1587 cancer samples confirming the low expression of ARLTS1 in PCa and showed that ARLTS1 expression was strongly associated with immune processes. This study provides strong confirmation of the important role of ARLTS1 Cys148Arg variant as a contributor in PCa predisposition and a potential marker for aggressive disease outcome.


Subject(s)
ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Genetic Predisposition to Disease/genetics , Polymorphism, Genetic/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Animals , Base Sequence , Cell Line, Tumor , Cell Transformation, Neoplastic , Chromosome Aberrations , Chromosomes, Human, Pair 13/genetics , DNA Copy Number Variations/genetics , Humans , Male , Mice , Prostatic Hyperplasia/enzymology , Prostatic Hyperplasia/genetics , Prostatic Neoplasms/enzymology
10.
Genes Chromosomes Cancer ; 50(7): 499-509, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21472816

ABSTRACT

MicroRNAs (miRNAs) are small, non-coding RNAs that negatively regulate the expression of protein coding genes. In this study, we screened highly informative prostate cancer cell lines and xenografts (n = 42) for miRNA gene copy number and expression changes. The expression profiling showed distinction between cell lines and xenografts as well as between androgen sensitive and independent models. Only a few copy number alterations that were associated with expression changes were identified. Most importantly, the miR-15a-miR-16-1 locus was found to be homozygously deleted in two samples leading to the abolishment of miR-15a, but not miR-16, expression. miR-16 is also expressed from another genomic locus. Mutation screening of the miR-15a-miR-16-1 gene in the model systems as well as clinical samples (n = 50) revealed no additional mutations. In conclusion, our data indicate that putative tumor suppressors, miR-15a and miR-16-1, are homozygously deleted in a subset of prostate cancers, further suggesting that these miRNAs could be important in the development of prostate cancer.


Subject(s)
Gene Deletion , Genetic Loci , Homozygote , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Cell Line, Tumor , Cluster Analysis , DNA Copy Number Variations/genetics , Gene Expression Profiling , Humans , Male , Prostatic Neoplasms/metabolism , Transplantation, Heterologous
11.
Clin Cancer Res ; 16(10): 2845-51, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20442300

ABSTRACT

PURPOSE: The aim of the study was to examine whether TMPRSS2:ERG fusion or SPINK1 protein expression is associated with hormone responsiveness of prostate cancer and can thus be used as a biomarker. EXPERIMENTAL DESIGN: Diagnostic needle biopsies from prostate cancer patients primarily treated by endocrine therapy were evaluated for TMPRSS2:ERG fusion with fluorescence in situ hybridization and SPINK1 protein expression with immunohistochemistry. RESULTS: The frequency of TMPRSS2:ERG fusion in 178 biopsies of hormonally treated patients was 34%. Of the fusion-positive cases, 71% showed deletion between the two genes, and 23% showed gain of the fusion. The fusion was associated with high Ki-67 staining (P=0.001), age at diagnosis (P=0.024), and tumor area (P=0.006), but not with Gleason score, T stage, M stage, prostate-specific antigen (PSA), or progression-free survival. Strong positive SPINK1 expression was found in 11% (21 of 186) of the biopsies. SPINK1-positive cases had significantly shorter progression-free survival compared with SPINK1-negative cases (P=0.001). The expression was not associated with any other clinicopathologic variables studied. In a multivariate analysis, SPINK1 expression showed independent prognostic value, with a relative risk of 2.3 (95% confidence interval, 1.1-4.6). SPINK1 expression and the fusion were not associated with each other. CONCLUSIONS: There was no association between TMPRSS2:ERG fusion and prognosis, suggesting that TMPRSS2:ERG rearrangement does not implicate hormone dependence of the cancer. SPINK1 expression, found in approximately 10% of prostate cancers, was associated with aggressive form of the disease and could serve as a biomarker in endocrine-treated prostate cancer.


Subject(s)
Biomarkers, Tumor/genetics , Carrier Proteins/genetics , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Aged , Antineoplastic Agents, Hormonal/therapeutic use , Disease-Free Survival , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Male , Prognosis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/mortality , Trypsin Inhibitor, Kazal Pancreatic
12.
Methods Mol Biol ; 505: 263-77, 2009.
Article in English | MEDLINE | ID: mdl-19117150

ABSTRACT

Genetic alterations underlying the development of cancer include large chromosomal aberrations, such as amplifications, deletions and translocations as well as small changes in sequence, i.e. mutations. Thus, different methods are needed to reveal various types of genetic changes. Fluorescence in situ hybridisation (FISH) is a versatile technique for detecting chromosomal alterations either in cultured cells or even in formalin-fixed paraffin-embedded tissue. For screening mutations, denaturing high-performance liquid chromatography (DHPLC) provides a relatively fast, cheap and sensitive option. The only special requirement is the HPLC equipment suitable for the analysis. As a screening tool, it does not reveal the actual base pair change, which in the end needs to be done by sequencing. FISH and DHPLC can both be utilized in research as well as in clinical diagnostic laboratories.


Subject(s)
Chromatography, High Pressure Liquid/methods , DNA Mutational Analysis/methods , In Situ Hybridization, Fluorescence/methods , Prostatic Neoplasms/genetics , Cell Line, Tumor , Chromatography, High Pressure Liquid/instrumentation , DNA Mutational Analysis/instrumentation , Humans , In Situ Hybridization, Fluorescence/instrumentation , Male
13.
Int J Cancer ; 123(7): 1601-9, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18649357

ABSTRACT

Lack of good models has complicated investigations on the mechanisms of prostate cancer. By far, the most commonly used transgenic mouse model of prostate cancer is TRAMP, which, however, has not been fully characterized for genetic and epigenetic aberrations. Here, we screened TRAMP-derived C2 cell line for the alterations using different microarray approaches, and compared it to human prostate cancer. TRAMP-C2 had relatively few genomic copy number alterations according to array comparative genomic hybridization (aCGH). However, the gene copy number and expression were significantly correlated (p < 0.001). Screening genes for promoter hypermethylation using demethylation treatment with 5-aza-2'-deoxycytidine and subsequent expression profiling indicated 43 putatively epigenetically silenced genes. Further studies revealed that clusterin is methylated in the TRAMP-C2 cell line, as well as in the human prostate cancer cell line LNCaP. Its expression was found to be significantly reduced (p < 0.01) in untreated and hormone-refractory human prostate carcinomas. Together with known function of clusterin, the data suggest an epigenetic component in the regulation of clusterin in prostate cancer.


Subject(s)
Adenocarcinoma/genetics , Clusterin/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Cell Line, Tumor , Chromosome Aberrations , DNA Methylation , Gene Silencing , Humans , Male , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
14.
Clin Cancer Res ; 14(11): 3395-400, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18519769

ABSTRACT

PURPOSE: Our aim was to assess the frequency of ERG overexpression and TMPRSS2:ERG rearrangement in prostate cancer and their association with clinicopathologic variables and outcome. EXPERIMENTAL DESIGN: The presence of the TMPRSS2:ERG rearrangement was studied by reverse transcription-PCR and fluorescence in situ hybridization in 19 prostate cancer xenografts and 7 prostate cancer cell lines. The expression of ERG was studied in the xenografts and cell lines and in 49 freshly frozen clinical prostate samples by quantitative reverse transcription-PCR. The frequency of the TMPRSS2:ERG fusion in clinical prostate cancer (n = 253) on tissue microarrays was assessed by three-color fluorescence in situ hybridization. RESULTS: Seven of 19 (37%) of the xenografts overexpressed ERG and had TMPRSS2:ERG rearrangement. Two xenografts, representing small cell carcinomas, also contained the fusion but did not express ERG. In clinical tumor specimens, the overexpression of ERG was associated with the rearrangement (P = 0.0019). Fifty of 150 (33%) of the prostatectomy specimens and 28 of 76 (37%) of the hormone-refractory prostate cancers on the tissue microarrays carried the TMPRSS2:ERG rearrangement. It was associated with longer progression-free survival in patients treated by prostatectomy (P = 0.019), and according to multivariate analysis, it was an independent predictor of favorable outcome (relative risk, 0.54; 95% confidence interval, 0.30-0.98). The fusion was not associated with Gleason score, pT stage, diagnostic prostate-specific antigen, or cell proliferation activity in prostatectomy specimens nor with the AR gene amplification in hormone-refractory tumors. CONCLUSIONS: The TMPRSS2:ERG rearrangement can be found in about one third of prostate cancers. A subgroup of prostate cancer patients with a good prognosis may be identified by the rearrangement.


Subject(s)
Biomarkers, Tumor/genetics , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Animals , Cell Line, Tumor , Disease-Free Survival , Humans , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Male , Mice , Middle Aged , Neoplasm Staging , Prognosis , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction
15.
Front Biosci ; 12: 3287-301, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17485299

ABSTRACT

Prostate cancer incidence is steadily increasing in Western industrialized countries where it has become the most common male malignancy and second most common cause of cancer death among men. Despite efforts to understand the mechanisms of prostate cancer development and progression, the reasons for the disease remain unclear. Although recurrent DNA copy number aberrations in prostate cancer have been well documented in the past 15 years, most of the target genes for these aberrations remain to be identified. The most common DNA copy number aberrations are losses in chromosomes 5q, 6q, 8p, 10q, 13q, 16q, 17p, and 18q, and gains in 7p/q, 8q, 9p, and Xq. In addition, a chromosomal rearrangement in 21q has been observed in over 50% of prostate cancers. The target genes for two common chromosomal aberrations have been identified: the androgen receptor (AR) gene at Xq12, and TMPRSS2 and ERG at 21q. Putative target genes for other copy number aberrations include: NKX3-1 (8p loss), PTEN and MXI1 (10q loss), FOXO1A (13q loss), CDH1 and ATBF1 (16q loss), MCM7 and EZH2 (7q gain), TCEB1, EIF3S3 and MYC (8q gain). The identification of target genes for the chromosomal aberrations will provide new prognostic markers and therapeutic targets for future drug development.


Subject(s)
Chromosome Aberrations , Prostatic Neoplasms/genetics , Chromosome Mapping , DNA, Neoplasm/genetics , Humans , Loss of Heterozygosity , Male
16.
Int J Cancer ; 119(6): 1322-9, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16642477

ABSTRACT

The aim of this study was to screen genetic as well as expression alterations in prostate cancer. Array comparative genomic hybridization (aCGH) to a 16K cDNA microarray was performed to analyze DNA sequence copy number alterations in 5 prostate cancer cell lines and 13 xenografts. The aCGH confirmed the previously implicated common gains and losses, such as gains at 1q, 7, 8q, 16p and 17q and losses at 2q, 4p/q, 6q, 8p, 13q, 16q, 17p and 18q, which have previously been identified by chromosomal CGH (cCGH). Because of the higher resolution of aCGH, the minimal commonly altered regions were significantly narrowed-down. For example, the gain of 8q was mapped to three independent regions, 8q13.3-q21.11, 8q22.2 and 8q24.13-q24.3. In addition, a novel recurrent gain at 9p13-q21 was identified. The concomitant expression analysis indicated that genome-wide DNA sequence copy number (gene dosage) was significantly associated with the expression level (p < 0.0001). The analyses indicated several individual genes whose expression was associated with the gene copy number. For example, gains of PTK2 and FZD6, were associated with the increased expression, whereas losses of TNFRSF10B (alias DR5) and ITGA4 with decreased expression. In conclusion, the aCGH mapping data will aid in the identification of genes altered in prostate cancer. The combined expression and copy number analysis suggested that even a low-level copy number change may have significant effect on gene expression, and thus on the development of prostate cancer.


Subject(s)
Chromosome Aberrations , Gene Expression Profiling , Prostatic Neoplasms/genetics , Animals , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Transplantation, Heterologous , Tumor Cells, Cultured
17.
Genes Chromosomes Cancer ; 45(7): 639-45, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16575874

ABSTRACT

Overexpression of the polycomb group protein enhancer of zeste homologue 2 (EZH2) has been found in several malignancies, including prostate cancer, with an aggressive phenotype. Amplification of the gene has previously been demonstrated in several malignancies, but not in prostate cancer. Our goal was to evaluate the gene copy number and expression alterations of EZH2 in prostate cancer. The copy number of EZH2 in cell lines (LNCaP, DU145, PC-3, 22Rv1), xenografts (n = 10), and clinical tumors (n = 191) was studied with fluorescence in situ hybridization. All cell lines had a gain of EZH2. Eight of the ten xenografts showed an increased copy number of the gene, including one case of high-level amplification (>or=5 copies of the gene and EZH2/centromere ratio >or=2). 34/125 (27%) of untreated prostate carcinomas showed increased copy number, but only one case of low-level amplification (>or=5 copies of the gene and EZH2/centromere ratio <2), whereas half (25/46) of the hormone-refractory carcinomas showed increased copy number, including seven cases of low-level amplification and three cases of high-level amplification (P < 0.0001). Expression of EZH2 was significantly (P = 0.0009) higher in hormone-refractory prostate cancer compared with that in benign prostatic hyperplasia or untreated cancer, according to quantitative real-time RT-PCR assay. Also, the expression of EZH2 protein was found to be higher in hormone-refractory tumors than in hormone-naïve tumors by immunohistochemistry. The EZH2 gene amplification was significantly (P < 0.05) associated with increased EZH2 protein expression. The data show that amplification of the EZH2 gene is rare in early prostate cancer, whereas a fraction of late-stage tumors contains the gene amplification leading to the overexpression of the gene, thus indicating the importance of EZH2 in the progression of prostate cancer.


Subject(s)
DNA-Binding Proteins/genetics , Gene Amplification , Prostatic Neoplasms/genetics , Transcription Factors/genetics , Cell Line, Tumor , DNA, Neoplasm , Disease Progression , Enhancer of Zeste Homolog 2 Protein , Gene Dosage , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Male , Neoplasm Staging , Polycomb Repressive Complex 2 , Prostatic Hyperplasia/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
18.
Cell Oncol ; 26(5-6): 347-51, 2004.
Article in English | MEDLINE | ID: mdl-15623945

ABSTRACT

OBJECTIVE: Hypoxia Inducible Factor-1 (HIF-1) is an important transcription factor that stimulates tumour growth and metastases via several pathways, including angiogenesis and altered metabolism. Activation of HIF-1 depends on the levels of its alpha-subunit, which increase during hypoxia. Recent studies showed that the HIF-1alpha gene was amplified in prostate cancer, leading to overexpression of HIF-1alpha at normoxia. The aim of this study was to evaluate the presence of HIF-1alpha gene amplifications in invasive breast cancer as an explanation for HIF-1alpha protein overexpression. METHODS: Protein and gene expression of HIF-1alpha were analyzed on a tissue microarray of 94 breast cancers by immunohistochemistry and fluorescent in situ hybridization (FISH), respectively. RESULTS: Overexpression of HIF-1alpha protein was found in 58/94 (62%) of patients. No amplifications of the HIF-1alpha gene were detected. CONCLUSION: Increased protein levels of HIF-1alpha are not associated with amplification of the HIF-1alpha gene in human breast cancer. Therefore, other mechanisms than gene amplification must be responsible for HIF-alpha overexpression at normoxia.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis , Protein Array Analysis/methods , Transcription Factors/genetics , Breast Neoplasms/pathology , Female , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Neoplasm Invasiveness , Oxygen/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism
19.
Cancer Res ; 64(5): 1589-94, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14996714

ABSTRACT

We report a previously unrecognized prostate-specific protein, PrLZ (prostate leucine zipper), a new member of the Tumor Protein D52 (TPD52) family. The gene for PrLZ was localized at chromosome 8q21.1, a locus most frequently amplified in human prostate cancer. Multiple tissue analyses demonstrated PrLZ predominantly in the prostate gland. Although its expression was enhanced by androgens in androgen receptor-expressing cells, PrLZ was detected in all of the human prostate cancer cell lines, regardless of androgen receptor status. Monoclonal anti-PrLZ antibodies were produced and intense immunohistochemical staining of PrLZ was observed in prostate epithelial cells in intraepithelial neoplasia and prostate cancer, whereas lower-level staining was detected in normal and benign epithelial components of the prostate gland. As the only prostate-specific gene identified in the most frequently amplified genomic region in prostate cancer, PrLZ may be the link between chromosome 8q amplification and malignant transformation of the prostate epithelia.


Subject(s)
Androgens/pharmacology , Chromosomes, Human, Pair 8 , Gene Amplification , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Biomarkers, Tumor , Cell Line, Tumor , Chromosome Mapping , Gene Expression Regulation, Neoplastic , Humans , Male
20.
Lab Invest ; 82(5): 629-37, 2002 May.
Article in English | MEDLINE | ID: mdl-12004003

ABSTRACT

Functional significance of several oncogenes is mediated by overexpression. To identify overexpressed genes in prostate cancer, we analyzed expression of 1081 transcripts in three prostate cancer cell lines (PC-3, DU145, and LNCaP) using cDNA microarray hybridization. The cDNA microarray analyses were validated by quantitative real-time RT-PCR. On average, 64% of the genes were expressed at detectable levels in the cell lines. Next, the expression profiles were combined with the data on DNA sequence copy number alterations in the cell lines obtained by comparative genomic hybridization. The genes for Elongin C and urokinase type plasminogen-activator, both located in the regions of amplification in the PC-3 cell line (8q21 and 10q22, respectively), were found to be overexpressed in the PC-3. Amplification and overexpression of urokinase type plasminogen-activator in prostate cancer has previously been reported. Here, fluorescence in situ hybridization on tissue microarray showed high-level amplification of the Elongin C gene in 8 (23%) of 35 hormone-refractory carcinomas but in none of the untreated prostate carcinomas (n = 35). Finally, it was shown that the Elongin C gene was overexpressed and amplified also in breast cancer cell line SK-Br-3. The results indicate that Elongin C is a putative target gene for 8q amplification.


Subject(s)
Carcinoma/genetics , Carcinoma/metabolism , Gene Amplification , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , DNA Primers/chemistry , DNA Probes/chemistry , DNA, Neoplasm/analysis , Elongin , Female , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Male , Oligonucleotide Array Sequence Analysis/methods , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...