Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters

Country/Region as subject
Publication year range
1.
BMC Fam Pract ; 19(1): 32, 2018 02 17.
Article in English | MEDLINE | ID: mdl-29454309

ABSTRACT

BACKGROUND: Most health authorities do not recommend screening for prostate cancer with PSA tests in asymptomatic patients who are not at increased risk. However, opportunistic screening for prostate cancer is still wanted by many patients and it is widely used in primary care clinics, with potential for overdiagnosis and overtreatment. Better tools for risk assessment have been called for, to better target such opportunistic screening. Our aim was to explore perceptions about prostate cancer risk and subsequent opportunistic screening among patients who were not at increased risk of prostate cancer after a first PSA test plus a genetic lifetime risk assessment. METHODS: We undertook semi-structured patient interviews with recording and verbatim transcription of interviews. Data were analysed thematically. RESULTS: Three themes were identified: uncertainty of the nature of prostate cancer; perceived benefits of testing; and conflicting public health recommendations. Prostate cancer was spoken of as an inescapable risk in older age. The aphorism "you die with it, not from it" was prominent in the interviews but patients focused on the benefits of testing now rather than the future risks associated with treatment relating to potential overdiagnosis. Many expressed frustration with perceived mixed messages about early detection of cancer, in which on one side men feel that they are encouraged to seek medical testing to act responsibly regarding the most common cancer disease in men, and on the other side they are asked to refrain from opportunistic testing for prostate cancer. Taken together, personal risks of prostate cancer were perceived as high in spite of a normal PSA test and a genetic lifetime risk assessment showing no increased risk. CONCLUSION: Patients saw prostate cancer risk as high and increasing with age. They focused on the perceived benefit of early detection using PSA testing. It was also commonly acknowledged that most cases are indolent causing no symptoms and not shortening life expectancy. There was a frustration with mixed messages about the benefit of early detection and risk of overdiagnosis. These men's genetic lifetime risk assessment showing no increased risk did not appear to influence current intentions to get PSA testing in the future.


Subject(s)
Early Detection of Cancer , Genetic Predisposition to Disease , Health Knowledge, Attitudes, Practice , Prostatic Neoplasms/diagnosis , Adult , Age Factors , Early Detection of Cancer/methods , Humans , Interviews as Topic , Male , Medical Overuse , Middle Aged , Prostate-Specific Antigen/blood , Prostatic Neoplasms/genetics , Qualitative Research , Risk Assessment
2.
Int J Mol Sci ; 18(9)2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28930171

ABSTRACT

Overdiagnosis and overtreatment of clinically insignificant tumors remains a major problem in prostate cancer (PC) due to suboptimal diagnostic and prognostic tools. Thus, novel biomarkers are urgently needed. In this study, we investigated the biomarker potential of Trefoil factor 3 (TFF3) promoter methylation and RNA expression levels for PC. Initially, by quantitative methylation specific PCR (qMSP) analysis of a large radical prostatectomy (RP) cohort (n = 292), we found that the TFF3 promoter was significantly hypomethylated in PC compared to non-malignant (NM) prostate tissue samples (p < 0.001) with an AUC (area under the curve) of 0.908 by receiver operating characteristics (ROC) curve analysis. Moreover, significant TFF3 promoter hypomethylation (p ≤ 0.010) as well as overexpression (p < 0.001) was found in PC samples from another large independent patient sample set (498 PC vs. 67 NM) analyzed by Illumina 450K DNA methylation arrays and/or RNA sequencing. TFF3 promoter methylation and transcriptional expression levels were inversely correlated, suggesting that epigenetic mechanisms contribute to the regulation of gene activity. Furthermore, low TFF3 expression was significantly associated with high ERG, ETS transcription factor (ERG) expression (p < 0.001), as well as with high Gleason score (p < 0.001), advanced pathological T-stage (p < 0.001), and prostate-specific antigen (PSA) recurrence after RP (p = 0.013; univariate Cox regression analysis). There were no significant associations between TFF3 promoter methylation levels, ERG status, or PSA recurrence in these RP cohorts. In conclusion, our results demonstrated diagnostic biomarker potential of TFF3 promoter hypomethylation for PC as well as prognostic biomarker potential of TFF3 RNA expression. To the best of our knowledge, this is the most comprehensive study of TFF3 promoter methylation and transcriptional expression in PC to date.


Subject(s)
Biomarkers, Tumor/genetics , DNA Methylation , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Trefoil Factor-3/genetics , Adult , Aged , Cohort Studies , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prostate/metabolism , Prostate/pathology , Prostate/surgery , Prostatectomy , Prostatic Neoplasms/surgery
3.
RNA ; 20(2): 236-51, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24344320

ABSTRACT

The past decade has shown mammalian genomes to be pervasively transcribed and identified thousands of noncoding (nc) transcripts. It is currently unclear to what extent these transcripts are of functional importance, as experimental functional evidence exists for only a small fraction. Here, we characterize the expression and evolutionary conservation properties of 12,115 known and novel nc transcripts, including structural RNAs, long nc RNAs (lncRNAs), antisense RNAs, EvoFold predictions, ultraconserved elements, and expressed nc regions. Expression levels are evaluated across 12 human tissues using a custom-designed microarray, supplemented with RNAseq. Conservation levels are evaluated at both the base level and at the syntenic level. We combine these measures with epigenetic mark annotations to identify subsets of novel nc transcripts that show characteristics similar to known functional ncRNAs. Few novel nc transcripts show both high expression and conservation levels. However, overall, we observe a positive correlation between expression and both conservation and epigenetic annotations, suggesting that a subset of the expressed transcripts are under purifying selection and likely functional. The identified subsets of expressed and conserved novel nc transcripts may form the basis for further functional characterization.


Subject(s)
RNA, Untranslated/genetics , Transcriptome , Base Sequence , Chromatin/genetics , Conserved Sequence , Expressed Sequence Tags , Humans , Inverted Repeat Sequences , Molecular Sequence Annotation , Oligonucleotide Array Sequence Analysis , Open Reading Frames , Organ Specificity , RNA, Untranslated/metabolism
4.
Proteomics ; 14(6): 699-712, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24376083

ABSTRACT

Cancer cells secrete soluble factors and various extracellular vesicles, including exosomes, into their tissue microenvironment. The secretion of exosomes is speculated to facilitate local invasion and metastatic spread. Here, we used an in vivo metastasis model of human bladder carcinoma cell line T24 without metastatic capacity and its two isogenic derivate cell lines SLT4 and FL3, which form metastases in the lungs and liver of mice, respectively. Cultivation in CLAD1000 bioreactors rather than conventional culture flasks resulted in a 13- to 16-fold increased exosome yield and facilitated quantitative proteomics of fractionated exosomes. Exosomes from T24, SLT4, and FL3 cells were partitioned into membrane and luminal fractions and changes in protein abundance related to the gain of metastatic capacity were identified by quantitative iTRAQ proteomics. We identified several proteins linked to epithelial-mesenchymal transition, including increased abundance of vimentin and hepatoma-derived growth factor in the membrane, and casein kinase II α and annexin A2 in the lumen of exosomes, respectively, from metastatic cells. The change in exosome protein abundance correlated little, although significant for FL3 versus T24, with changes in cellular mRNA expression. Our proteomic approach may help identification of proteins in the membrane and lumen of exosomes potentially involved in the metastatic process.


Subject(s)
Epithelial-Mesenchymal Transition , Exosomes/pathology , Proteome/analysis , Urinary Bladder Neoplasms/pathology , Urinary Bladder/pathology , Animals , Cell Line, Tumor , Exosomes/metabolism , Humans , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Mass Spectrometry , Mice , Neoplasm Metastasis/pathology , Proteome/metabolism , Urinary Bladder/metabolism , Urinary Bladder Neoplasms/metabolism , Vimentin/analysis , Vimentin/metabolism
5.
BMC Cancer ; 14: 859, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25416653

ABSTRACT

BACKGROUND: Prostate cancer is one of the leading causes of cancer death amongst men in economically advanced countries. The disease is characterized by a greatly varying clinical course, where some patients harbor non- or slowly-progressive disease, others highly aggressive disease. There is a great lack of markers to differentiate between aggressive and indolent disease. Markers that could help to identify patients needing curative treatment while sparing those who do not. METHODS: MicroRNA profiling of 672 microRNAs using multiplex RT-qPCR was performed using 36 prostate cancer samples to evaluate the association of microRNAs and biochemical recurrence after radical prostatectomy. RESULTS: Among 31 microRNAs associated with recurrence, we identified miR-449b, which was further validated in an independent cohort of 163 radical prostatectomy patients. Patients expressing miR-449b had a significantly higher risk of recurrence (HR = 1.57; p = 0.028), and miR-449b was shown to be an independent predictor of recurrence after prostatectomy (HR = 1.9; p = 0.003) when modeled with known risk factors of recurrent disease in multivariate analysis. CONCLUSION: High miR-449b expression was shown to be an independent predictor of biochemical recurrence after radical prostatectomy.


Subject(s)
Gene Expression , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Aged , Biomarkers , Cohort Studies , Gene Expression Profiling , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prostatectomy , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Recurrence , Reproducibility of Results
6.
Int J Cancer ; 131(10): 2320-31, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22362417

ABSTRACT

Tumor-associated macrophages (TAMs) represent a distinct malignancy-promoting phenotype suggested to play a key role in tumor formation and metastasis. We aimed to investigate the expression of the monocyte/macrophage-restricted receptor CD163 in bladder tumor biopsies and assess the potential mechanism inducing the CD163 expression in tumor cells. A high CD163 mRNA expression (n = 87) was significantly associated with a poor 13-year overall survival (log-rank test, χ(2) = 8.931; p = 0.0028). Moreover, CD163 mRNA expression was significantly increased in muscle invasive (T2-T4), p = 0.017, and aggressive (grade III/IV) cancers (p = 0.015). The expression strongly correlated with local expression of IL-6 (r = 0.72; p <0.0001) and IL-10 (r = 0.75; p <0.0001), mediators known to induce CD163 expression in vitro. CD163 immunostaining (n = 46) confirmed the association between dense TAM infiltration and histologically advanced disease. In 39% of the biopsies, CD163 immunoreactivity was also observed in tumor cells, and CD163-expressing metastatic cells were identified in lymph node biopsies (n = 8). Bladder cancer cell lines did not express CD163; however, when cocultured with macrophages the bladder cancer cell expression of CD163 was significantly induced in an IL-6/IL-10 independent manner. In conclusion, we show a strong association between CD163 mRNA expression in bladder cancer biopsies and poor patient outcome. CD163 expression was not confined to the infiltrating TAMs, but was also expressed by a significant portion of the malignant cells in both tumors and lymph nodes. CD163 expressing tumor cells may constitute a subpopulation of tumor cells with a phenotypic shift associated with epithelial-to-mesenchymal transition (EMT) and increased metastatic activity induced by TAMs.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Macrophages/metabolism , Receptors, Cell Surface/metabolism , Urinary Bladder Neoplasms/metabolism , Aged , Aged, 80 and over , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lymph Nodes/metabolism , Lymph Nodes/pathology , Macrophages/immunology , Male , Middle Aged , Neoplasm Staging , Receptors, Cell Surface/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
7.
Int J Cancer ; 129(8): 1848-58, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21154748

ABSTRACT

Genomic alterations play important roles in colorectal cancer (CRC) carcinogenesis. Here, we aimed to identify and characterize recurrent copy-number alterations (CNAs) associated with clinical outcome of CRC by the use of single nucleotide polymorphism arrays, genomic quantitative PCR (qPCR) and fluorescence in situ hybridization (FISH). Colorectal neoplasia specimens and paired germline samples from 144 patients (40 adenomas and 104 carcinomas) as well as 40 CRC cell lines were investigated. This large dataset revealed frequent loss, including homozygous loss, at chr16p13.2 (from 5.9 to 7.42Mb). The loss was observed in 30% of adenomas and even more frequently in carcinomas, 56%, indicating that the loss define a subset of adenomas with a propensity for invasion. Consistent with this, the loss occurred twice as frequent in villous (40%) as in tubular adenomas (20%). The loss occurred independently of microsatellite stability and could be validated by qPCR in an independent sample cohort (n = 71). In Stage II/III, microsatellite stable (MSS) CRC it was associated with poor recurrence free survival (hazard ratio 2.4; p = 0.02; Multivariate Cox regression analysis). No transcriptional consequences of the losses were observed, and the only gene, A2BP1, located in the region showed no mutations. Correlation with other CNAs was established for chr3p22 in carcinomas and chr20p (inverse) in adenomas. FISH documented the chr16p13.2 region to be involved in complex structural rearrangements that included translocation to chr3p22 in some cases. The findings indicate that structural rearrangements involving chr16p13.2 are very frequent in colorectal neoplasia, often lead to homozygous deletion, and are associated with poor clinical outcome.


Subject(s)
Adenoma/genetics , Carcinoma , Chromosomes, Human, Pair 16 , Colorectal Neoplasms/genetics , DNA Copy Number Variations , Aged , Carcinoma/genetics , Cell Line, Tumor , Disease-Free Survival , Female , Humans , Male , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Prognosis
8.
Tumour Biol ; 32(4): 671-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21399973

ABSTRACT

FK506 binding protein 65 (FKBP65) belongs to a group of proteins termed immunophilins that have a high binding affinity to immunosuppressant drugs as FK506 (tacrolimus) and rapamycin (sirolimus). Treatment of female premenopausal women with tacrolimus, which binds to FKBP65, has been reported to be followed by a strongly increased risk of ovarian cysts. We performed the present study to reveal how FKBP65 is expressed in the ovary and in ovarian tumors and to see if this expression might be related to ovarian tumor development, a relationship we have found in colorectal cancer. Biopsies from prospectively collected samples from ovaries and benign, borderline, and invasive ovarian tumors were analyzed for expression of FKBP65 by immunohistochemistry. The expression was compared to survival and several clinicopathological parameters. FKBP65 is strongly expressed in ovarian epithelium and in benign ovarian tumor cells. In the ovary, a positive staining was also found in endothelial cells of blood vessels. In non-invasive and in invasive malignant tumor cells, a decreased staining was observed, which was not correlated to stage, histology, or survival. A significant inversed correlation to expression of p53 was found. The differential expression of FKBP65 indicates a role in ovarian physiology as well as in ovarian tumor development. Our observations and the chromosomal localization of the FKBP65 gene indicate a tumor suppressor function of the FKBP65 protein in ovarian carcinogenesis.


Subject(s)
Epithelial Cells/metabolism , Ovarian Neoplasms/metabolism , Ovary/metabolism , Tacrolimus Binding Proteins/biosynthesis , Blotting, Western , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality
9.
Int J Cancer ; 127(8): 1857-65, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20112343

ABSTRACT

Trefoil factors, mucin-associated peptides, are overexpressed in prostate cancer (PC). We hypothesized that promoter methylation contributes to the regulation of trefoil factors (TFF1, TFF2 and TFF3) in human prostate cells. Here we show hypomethylation of promoter regions of TFF1 and TFF3 in PC cell lines with significant TFF expression as compared to benign immortalized prostate cell lines and PC cell lines not expressing trefoil factor. The most striking difference was observed for CpG sites located close to the AUG start codon overlapping several putative binding sites for cellular transcription factors. TFF2 was hypermethylated and had no or very low expression in all prostate cell lines investigated. Treatment of methylated cell lines with 5-aza-2'-deoxycytidine restored TFF expression in cell lines not expressing TFF and increased expression significantly in low-expressing cell lines. In clinical samples, methylation of the promoter/enhancer regions of TFF1 and TFF3 was significantly lower in PC compared to benign prostatic hyperplasia. The present study shows an inverse relation between promoter methylation and expression of trefoil factors. Preliminary analysis on clinical samples suggests that this regulatory mechanism is responsible for the increased levels of TFF1 and TFF3 observed in PC. The overexpression and promoter hypomethylation of trefoil factors may serve as biomarkers in PC.


Subject(s)
DNA Methylation , Peptides/genetics , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Prognosis , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , Trefoil Factor-1 , Trefoil Factor-2 , Trefoil Factor-3 , Tumor Cells, Cultured , Up-Regulation
10.
Clin Cancer Res ; 15(4): 1400-10, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19228741

ABSTRACT

PURPOSE: This study investigates SLC18A2 (vesicular monoamine transporter 2) expression in prostate adenocarcinoma and examines its potential as a predictive marker for prostate cancer patient outcome after radical prostatectomy. EXPERIMENTAL DESIGN: Expression and single nucleotide polymorphism microarray analyses identified SLC18A2 as both down-regulated and subject to common loss-of-heterozygosity in prostate cancer. Down-regulated SLC18A2 expression was validated on tissue microarrays containing benign and malignant prostate specimens from an independent patient group (n=738). Furthermore, SLC18A2 immunoreactivity in radical prostatectomy tumor specimens (n=506) was correlated to clinicopathologic characteristics and recurrence-free survival. The possibility of SLC18A2 silencing by aberrant DNA methylation in prostate cancer cells was investigated by bisulfite sequencing. RESULTS: Tissue microarray analysis revealed markedly lower cytoplasmic SLC18A2 staining in cancer compared with nonmalignant prostate tissue samples, confirming RNA expression profiling results. Furthermore, multivariate analysis identified cytoplasmic SLC18A2 immunoreactivity as a novel predictor of biochemical recurrence following prostatectomy (hazard ratio, 0.485; 95% confidence interval, 0.333-0.709; P<0.001) independent of prostate-specific antigen, Gleason score, tumor stage, and surgical margin status. SLC18A2 showed loss-of-heterozygosity in 23% of the tumors and was densely hypermethylated in 15 of 17 (88%) prostate cancer samples plus 6 of 6 prostate cancer cell lines. In contrast, SLC18A2 was unmethylated in 4 of 4 adjacent nonmalignant prostate and 3 of 5 benign prostatic hyperplasia tissue samples, whereas 2 of 5 benign prostatic hyperplasia samples had monoallelic hypermethylation. Methylation and histone deacetylase inhibitory agents rescued SLC18A2 expression in three prostate cancer cell lines. CONCLUSIONS: SLC18A2 silencing by DNA hypermethylation and/or allelic loss is a frequent event in prostate cancer and a novel independent predictor of biochemical recurrence after prostatectomy.


Subject(s)
Gene Silencing , Neoplasm Recurrence, Local/genetics , Prostatectomy , Prostatic Neoplasms/genetics , Vesicular Monoamine Transport Proteins/genetics , DNA Methylation , Humans , Loss of Heterozygosity , Male , Multivariate Analysis , Prostatic Neoplasms/mortality , Prostatic Neoplasms/surgery , Tissue Array Analysis , Vesicular Monoamine Transport Proteins/analysis , Vesicular Monoamine Transport Proteins/physiology
11.
BMC Cancer ; 9: 149, 2009 May 16.
Article in English | MEDLINE | ID: mdl-19445696

ABSTRACT

BACKGROUND: Non-muscle invasive bladder neoplasms with invasion of the lamina propria (stage T1) or high grade of dysplasia are at "high risk" of progression to life-threatening cancer. However, the individual course is difficult to predict. Chromosomal instability (CI) is associated with high tumor stage and grade, and possibly with the risk of progression. METHODS: To investigate the relationship between CI and subsequent disease progression, we performed a case-control-study of 125 patients with "high-risk" non-muscle invasive bladder neoplasms, 67 with later disease progression, and 58 with no progression. Selection criteria were conservative (non-radical) resections and full prospective clinical follow-up (> 5 years). We investigated primary lesions in 59, and recurrent lesions in 66 cases.We used Affymetrix GeneChip Mapping 10 K and 50 K SNP microarrays to evaluate genome wide chromosomal imbalance (loss-of-heterozygosity and DNA copy number changes) in 48 representative tumors. DNA copy number changes of 15 key instability regions were further investigated using QPCR in 101 tumors (including 25 tumors also analysed on 50 K SNP microarrays). RESULTS: Chromosomal instability did not predict any higher risk of subsequent progression. Stage T1 and high-grade tumors had generally more unstable genomes than tumors of lower stage and grade (mostly non-primary tumors following a "high-risk" tumor). However, about 25% of the "high-risk" tumors had very few alterations. This was independent of subsequent progression. Recurrent lesions represent underlying field disease. A separate analysis of these lesions did neither reflect any difference in the risk of progression. Of specific chromosomal alterations, a possible association between loss of chromosome 8p11 and the risk of progression was found. However, the predictive value was limited by the heterogeneity of the changes. CONCLUSION: Chromosomal instability (CI) was associated with "high risk" tumors (stage T1 or high-grade), but did not predict subsequent progression. Recurrences after "high-risk" tumors had fewer chromosomal alterations, but there was no association with the risk of progression in this group either. Thus, the prediction of progression of "high risk" non-muscle invasive bladder tumors using chromosomal changes is difficult. Loss of chromosome 8p11 may play a role in the progression process. About 25% of the "high risk" tumors were chromosomal stable.


Subject(s)
Chromosome Aberrations , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Aged , Aged, 80 and over , Case-Control Studies , Disease Progression , Female , Gene Dosage , Genome, Human , Humans , Loss of Heterozygosity , Male , Middle Aged , Neoplasm Staging , Polymerase Chain Reaction/methods , Polymorphism, Single Nucleotide
12.
Cancer Res ; 67(6): 2559-67, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17363574

ABSTRACT

Heat shock protein 70-2 (Hsp70-2) is a chaperone protein essential for the growth of spermatocytes and cancer cells. Here, we show that Hsp70-2 depletion triggers lysosomal membrane permeabilization and cathepsin-dependent cell death and identify lens epithelium-derived growth factor (LEDGF) as an Hsp70-2-regulated guardian of lysosomal stability in human cancer. Knockdown of LEDGF in cancer cells induces destabilization of lysosomal membranes followed by caspase-independent and Bcl-2-resistant cell death. Accordingly, ectopic LEDGF stabilizes lysosomes and protects cancer cells against cytotoxicity induced by anticancer agents that trigger the lysosomal cell death pathway. Remarkably, ectopic LEDGF also increases the tumorigenic potential of human cancer cells in immunodeficient mice, and LEDGF expression is increased in human breast and bladder carcinomas correlating with that of Hsp70-2 in invasive bladder cancer. Taken together, these data reveal LEDGF as an oncogenic protein that controls a caspase-independent lysosomal cell death pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Breast Neoplasms/metabolism , HSP70 Heat-Shock Proteins/physiology , Lysosomes/metabolism , Transcription Factors/physiology , Urinary Bladder Neoplasms/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/deficiency , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Death/physiology , Cell Lineage , Cell Membrane Permeability , Down-Regulation , Female , HSP70 Heat-Shock Proteins/deficiency , HeLa Cells , Humans , Lysosomes/pathology , Mice , Mice, SCID , Transcription Factors/biosynthesis , Transcription Factors/deficiency , Transfection , Transplantation, Heterologous , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
13.
Int J Cancer ; 122(3): 509-19, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17943724

ABSTRACT

Loss of heterozygosity (LOH) at 6q is a frequent chromosomal aberration in prostate adenocarcinoma; however, a possible target gene remains to be identified. Findings in this study indicate that the FYN tyrosine kinase gene at 6q21 is a new candidate tumor suppressor in prostate cancer. Initially, single nucleotide polymorphism microarray analysis of 40 microdissected prostate adenocarcinoma samples revealed 25% LOH at the FYN locus. Furthermore, Western blot analysis and real-time reverse transcriptase PCR (RT-PCR) showed significantly lower FYN expression in prostate cancer tissue than in benign prostate hyperplasia (BPH), as well as in 6 prostate adenocarcinoma cell lines compared with that in BPH-1 cells. By immunohistochemistry, FYN protein was detected in nonmalignant prostate epithelium, but not in cancerous glands. Moreover, genomic bisulfite sequencing revealed frequent aberrant methylation of a large CpG island in the FYN promoter region in both adenocarcinoma cell lines (3 of 5 cell lines tested) and primary prostate cancer (12 of 18 tumors). Methylation was generally of moderate density, affecting preferentially the 3' region of the CpG island. Dense hypermethylation of the entire CpG island, consistent with gene silencing, was detected in 2 of 18 tumors (11%). No methylation was found in BPH-1 cells or nonmalignant prostate tissue samples (0 of 7). These results indicate that FYN is downregulated in prostate cancer by both chromosomal deletion and promoter hypermethylation, and therefore is a novel prostate tumor suppressor gene candidate.


Subject(s)
Chromosome Deletion , DNA Methylation , Gene Expression Regulation, Neoplastic , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-fyn/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Blotting, Western , Cell Line, Tumor , CpG Islands , Down-Regulation , Enzyme Induction , Humans , Male , Middle Aged , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-fyn/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Nat Commun ; 9(1): 3664, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30202008

ABSTRACT

Point mutations in cancer have been extensively studied but chromosomal gains and losses have been more challenging to interpret due to their unspecific nature. Here we examine high-resolution allelic imbalance (AI) landscape in 1699 colorectal cancers, 256 of which have been whole-genome sequenced (WGSed). The imbalances pinpoint 38 genes as plausible AI targets based on previous knowledge. Unbiased CRISPR-Cas9 knockout and activation screens identified in total 79 genes within AI peaks regulating cell growth. Genetic and functional data implicate loss of TP53 as a sufficient driver of AI. The WGS highlights an influence of copy number aberrations on the rate of detected somatic point mutations. Importantly, the data reveal several associations between AI target genes, suggesting a role for a network of lineage-determining transcription factors in colorectal tumorigenesis. Overall, the results unravel the contribution of AI in colorectal cancer and provide a plausible explanation why so few genes are commonly affected by point mutations in cancers.


Subject(s)
Allelic Imbalance , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , CRISPR-Cas Systems , Chromosome Aberrations , Chromosomes, Human, Pair 8 , Colorectal Neoplasms/pathology , DNA Copy Number Variations , Denmark , Gene Expression Profiling , Genomics , Genotype , Humans , Loss of Heterozygosity , Microsatellite Repeats , Phenotype , Point Mutation , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Small Interfering/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Whole Genome Sequencing
16.
Front Biosci ; 12: 2063-73, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17127444

ABSTRACT

Bladder cancer is the fifth most common neoplasm in industrialized countries. Due to frequent recurrences of the superficial form of this disease, bladder cancer ranks as one of the most common cancers. Despite the description of a large number of tumor markers for bladder cancers, none have individually contributed to the management of the disease. However, the development of high-throughput techniques for simultaneous assessment of a large number of markers has allowed classification of tumors into clinically relevant molecular subgroups beyond those possible by pathological classification. Here, we review the recent advances in high-throughput molecular marker identification for superficial and invasive bladder cancers.


Subject(s)
Biomarkers, Tumor/analysis , Urinary Bladder Neoplasms/diagnosis , Biomarkers, Tumor/genetics , Carcinoma/classification , Carcinoma/diagnosis , Carcinoma/pathology , Female , Gene Expression Profiling , Humans , Male , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Prognosis , Protein Array Analysis , Urinary Bladder Neoplasms/classification , Urinary Bladder Neoplasms/pathology
17.
Genome Biol ; 18(1): 38, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28222791

ABSTRACT

The study of epigenetic heterogeneity at the level of individual cells and in whole populations is the key to understanding cellular differentiation, organismal development, and the evolution of cancer. We develop a statistical method, epiG, to infer and differentiate between different epi-allelic haplotypes, annotated with CpG methylation status and DNA polymorphisms, from whole-genome bisulfite sequencing data, and nucleosome occupancy from NOMe-seq data. We demonstrate the capabilities of the method by inferring allele-specific methylation and nucleosome occupancy in cell lines, and colon and tumor samples, and by benchmarking the method against independent experimental data.


Subject(s)
DNA Methylation , Epigenomics/methods , High-Throughput Nucleotide Sequencing , Sequence Analysis, DNA , Software , Alleles , CpG Islands , Gene Expression Profiling , Genotype , Nucleosomes/metabolism , Polymorphism, Single Nucleotide , Protein Binding , Reproducibility of Results
18.
Sci Rep ; 7: 40636, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28084441

ABSTRACT

Prostate cancer (PC) diagnosis is based on histological evaluation of prostate needle biopsies, which have high false negative rates. Here, we investigated if cancer-associated epigenetic field effects in histologically normal prostate tissue may be used to increase sensitivity for PC. We focused on nine genes (AOX1, CCDC181 (C1orf114), GABRE, GAS6, HAPLN3, KLF8, MOB3B, SLC18A2, and GSTP1) known to be hypermethylated in PC. Using quantitative methylation-specific PCR, we analysed 66 malignant and 134 non-malignant tissue samples from 107 patients, who underwent ultrasound-guided prostate biopsy (67 patients had at least one cancer-positive biopsy, 40 had exclusively cancer-negative biopsies). Hypermethylation was detectable for all genes in malignant needle biopsy samples (AUC: 0.80 to 0.98), confirming previous findings in prostatectomy specimens. Furthermore, we identified a four-gene methylation signature (AOX1xGSTP1xHAPLN3xSLC18A2) that distinguished histologically non-malignant biopsies from patients with vs. without PC in other biopsies (AUC = 0.65; sensitivity = 30.8%; specificity = 100%). This signature was validated in an independent patient set (59 PC, 36 adjacent non-malignant, and 9 normal prostate tissue samples) analysed on Illumina 450 K methylation arrays (AUC = 0.70; sensitivity = 40.6%; specificity = 100%). Our results suggest that a novel four-gene signature may be used to increase sensitivity for PC diagnosis through detection of epigenetic field effects in histologically non-malignant prostate tissue samples.


Subject(s)
DNA Methylation , Genetic Heterogeneity , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Aged , Aged, 80 and over , Biomarkers, Tumor , Biopsy, Needle , Epigenesis, Genetic , Epigenomics/methods , Humans , Male , Middle Aged , Neoplasm Grading , Prostatic Neoplasms/surgery , ROC Curve , Reproducibility of Results
19.
Eur Urol ; 71(6): 961-969, 2017 06.
Article in English | MEDLINE | ID: mdl-28069289

ABSTRACT

BACKGROUND: Disease surveillance in patients with bladder cancer is important for early diagnosis of progression and metastasis and for optimised treatment. OBJECTIVE: To develop urine and plasma assays for disease surveillance for patients with FGFR3 and PIK3CA tumour mutations. DESIGN, SETTING, AND PARTICIPANTS: Droplet digital polymerase chain reaction (ddPCR) assays were developed and tumour DNA from two patient cohorts was screened for FGFR3 and PIK3CA hotspot mutations. One cohort included 363 patients with non-muscle-invasive bladder cancer (NMIBC). The other cohort included 468 patients with bladder cancer undergoing radical cystectomy (Cx). Urine supernatants (NMIBC n=216, Cx n=27) and plasma samples (NMIBC n=39, Cx n=27) from patients harbouring mutations were subsequently screened using ddPCR assays. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Progression-free survival, recurrence-free survival, and overall survival were measured. Fisher's exact test, the Wilcoxon rank-sum test and Cox regression analysis were applied. RESULTS AND LIMITATIONS: In total, 36% of the NMIBC patients (129/363) and 11% of the Cx patients (44/403) harboured at least one FGFR3 or PIK3CA mutation. Screening of DNA from serial urine supernatants from the NMIBC cohort revealed that high levels of tumour DNA (tDNA) were associated with later disease progression in NMIBC (p=0.003). Furthermore, high levels of tDNA in plasma samples were associated with recurrence in the Cx cohort (p=0.016). A positive correlation between tDNA levels in urine and plasma was observed (correlation coefficient 0.6). The retrospective study design and low volumes of plasma available for analysis were limitations of the study. CONCLUSIONS: Increased levels of FGFR3 and PIK3CA mutated DNA in urine and plasma are indicative of later progression and metastasis in bladder cancer. PATIENT SUMMARY: Urine and plasma from patients with bladder cancer may be monitored for diagnosis of progression and metastasis using mutation assays.


Subject(s)
Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Mutation , Receptor, Fibroblast Growth Factor, Type 3/genetics , Urinary Bladder Neoplasms/genetics , Aged , Biomarkers, Tumor/blood , Biomarkers, Tumor/urine , Circulating Tumor DNA/blood , Circulating Tumor DNA/urine , Class I Phosphatidylinositol 3-Kinases/blood , Class I Phosphatidylinositol 3-Kinases/urine , Cystectomy , DNA Mutational Analysis , Disease Progression , Disease-Free Survival , Female , Genetic Predisposition to Disease , Humans , Kaplan-Meier Estimate , Liquid Biopsy , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Recurrence, Local , Phenotype , Polymerase Chain Reaction , Predictive Value of Tests , Proportional Hazards Models , Receptor, Fibroblast Growth Factor, Type 3/blood , Receptor, Fibroblast Growth Factor, Type 3/urine , Retrospective Studies , Risk Factors , Time Factors , Treatment Outcome , Urinary Bladder Neoplasms/blood , Urinary Bladder Neoplasms/surgery , Urinary Bladder Neoplasms/urine
20.
Sci Rep ; 7(1): 395, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28341852

ABSTRACT

Aberrant expression of long non-coding RNAs (lncRNAs) has been regarded as a critical component in bladder cancer (BC) and lncRNAs have been associated with BC development and progression although their overall expression and functional significance is still unclear. The aim of our study was to identify novel lncRNAs with a functional role in BC carcinogenesis. RNA-sequencing was used to identify aberrantly expressed lncRNAs in 8 normal and 72 BC samples. We identified 89 lncRNAs that were significantly dys-regulated in BC. Five lncRNAs; LINC00958, LINC01296, LINC00355, LNC-CMC1-1 and LNC-ALX1-2 were selected for further analyses. Silencing of LINC00958 or LINC01296 in vitro reduced both cell viability and migration. Knock-down of LINC00958 also affected invasion and resistance to anoikis. These cellular effects could be linked to direct/indirect regulation of protein coding mRNAs involved in cell death/survival, proliferation and cellular movement. Finally, we showed that LINC00958 binds proteins involved in regulation and initiation of translation and in post-transcriptional modification of RNA, including Metadherin, which has previously been associated with BC. Our analyses identified novel lncRNAs in BC that likely act as oncogenic drivers contributing to an aggressive cancerous phenotype likely through interaction with proteins involved in initiation of translation and/or post-transcriptional modification of RNA.


Subject(s)
Gene Expression Regulation, Neoplastic , Oncogenes , RNA, Long Noncoding/genetics , Urinary Bladder Neoplasms/genetics , Cell Line, Tumor , Cell Survival , Gene Expression Profiling , Humans , Transcriptome , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL