Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
NPJ Genom Med ; 5: 41, 2020.
Article in English | MEDLINE | ID: mdl-33062306

ABSTRACT

Germline copy number variants (CNVs) and single-nucleotide polymorphisms (SNPs) form the basis of inter-individual genetic variation. Although the phenotypic effects of SNPs have been extensively investigated, the effects of CNVs is relatively less understood. To better characterize mechanisms by which CNVs affect cellular phenotype, we tested their association with variable CpG methylation in a genome-wide manner. Using paired CNV and methylation data from the 1000 genomes and HapMap projects, we identified genome-wide associations by methylation quantitative trait locus (mQTL) analysis. We found individual CNVs being associated with methylation of multiple CpGs and vice versa. CNV-associated methylation changes were correlated with gene expression. CNV-mQTLs were enriched for regulatory regions, transcription factor-binding sites (TFBSs), and were involved in long-range physical interactions with associated CpGs. Some CNV-mQTLs were associated with methylation of imprinted genes. Several CNV-mQTLs and/or associated genes were among those previously reported by genome-wide association studies (GWASs). We demonstrate that germline CNVs in the genome are associated with CpG methylation. Our findings suggest that structural variation together with methylation may affect cellular phenotype.

2.
Cancer Med ; 8(15): 6709-6716, 2019 11.
Article in English | MEDLINE | ID: mdl-31503420

ABSTRACT

BACKGROUND: MicroRNAs have recently emerged as promising circulating biomarkers in diverse cancer types, including ovarian cancer. We utilized conditional, doxycycline-induced fallopian tube (FT)-derived cancer models to identify changes in miRNA expression in tumors and plasma, and further validated the murine findings in high-grade ovarian cancer patient samples. METHODS: We analyzed 566 biologically informative miRNAs in doxycycline-induced FT and metastatic tumors as well as plasma samples derived from murine models bearing inactivation of Brca, Tp53, and Pten genes. We identified miRNAs that showed a consistent pattern of dysregulated expression and validated our results in human patient serum samples. RESULTS: We identified six miRNAs that were significantly dysregulated in doxycycline-induced FTs (P < .05) and 130 miRNAs differentially regulated in metastases compared to normal fallopian tissues (P < .05). Furthermore, we validated miR-21a-5p, miR-146a-5p, and miR-126a-3p as dysregulated in both murine doxycycline-induced FT and metastatic tumors, as well as in murine plasma and patient serum samples. CONCLUSIONS: In summary, we identified changes in miRNA expression that potentially accompany tumor development in murine models driven by commonly found genetic alterations in cancer patients. Further studies are required to test both the function of these miRNAs in driving the disease and their utility as potential biomarkers for diagnosis and/or disease progression.


Subject(s)
Doxycycline/adverse effects , Fallopian Tubes/pathology , Gene Expression Profiling/methods , MicroRNAs/genetics , Ovarian Neoplasms/genetics , Animals , Biomarkers, Tumor/genetics , Fallopian Tubes/chemistry , Fallopian Tubes/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Middle Aged , Neoplasm Transplantation , Ovarian Neoplasms/pathology
3.
Clin Cancer Res ; 23(3): 735-745, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27503198

ABSTRACT

PURPOSE: Chronic lymphocytic leukemia (CLL) with 17p deletion typically progresses quickly and is refractory to most conventional therapies. However, some del(17p) patients do not progress for years, suggesting that del(17p) is not the only driving event in CLL progression. We hypothesize that other concomitant genetic abnormalities underlie the clinical heterogeneity of del(17p) CLL. EXPERIMENTAL DESIGN: We profiled the somatic mutations and copy number alterations (CNA) in a large group of del(17p) CLLs as well as wild-type CLL and analyzed the genetic basis of their clinical heterogeneity. RESULTS: We found that increased somatic mutation number associates with poor overall survival independent of 17p deletion (P = 0.003). TP53 mutation was present in 81% of del(17p) CLL, mostly clonal (82%), and clonal mutations with del(17p) exhibit shorter overall survival than subclonal mutations with del(17p) (P = 0.019). Del(17p) CLL has a unique driver mutation profile, including NOTCH1 (15%), RPS15 (12%), DDX3X (8%), and GPS2 (6%). We found that about half of del(17p) CLL cases have recurrent deletions at 3p, 4p, or 9p and that any of these deletions significantly predicts shorter overall survival. In addition, the number of CNAs, but not somatic mutations, predicts shorter time to treatment among patients untreated at sampling. Indolent del(17p) CLLs were characterized by absent or subclonal TP53 mutation and few CNAs, with no difference in somatic mutation number. CONCLUSIONS: We conclude that del(17p) has a unique genomic profile and that clonal TP53 mutations, 3p, 4p, or 9p deletions, and genomic complexity are associated with shorter overall survival. Clin Cancer Res; 23(3); 735-45. ©2016 AACR.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 17/ultrastructure , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Adult , Aged , Aged, 80 and over , Chromosome Breakage , Chromosomes, Human, Pair 17/genetics , Clone Cells , Disease Progression , Female , Gene Dosage , Germ-Line Mutation , Humans , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Male , Middle Aged , Mutation , Polymorphism, Single Nucleotide , Saliva/chemistry , Tumor Suppressor Protein p53/genetics , Exome Sequencing
4.
J Formos Med Assoc ; 114(1): 81-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25618588

ABSTRACT

Mosaicism with an isodicentric 8 with a breakpoint at p23.3 [idic(8)(p23.3)] is very rare. We report the first prenatal case on a male fetus, in which obstetric ultrasound revealed multiple congenital anomalies at 28 weeks of gestation. Cytogenetic analysis of amniocytes showed mos 45,XY,-8,psu idic(8)(p23.3)[16]/46,XY,psu idic(8)(p23.3)[4], and that of cord blood lymphocytes revealed mos 46,XY, psu idic(8)(p23.3)[37]/45,XY,-8,psu idic(8)(p23.3)[13]. Fluorescence in situ hybridization studies revealed that the break-reunion occurred at the cytoband 8p23.3 within the physical position 2.08 Mb from the 8p telomere. Chromosomal microarray analyses further assigned the duplication/deletion breakpoint at 2.16 Mb (Agilent 244K) and at 2.19 Mb (Affymetrix SNP6.0). Analysis of microsatellite DNA indicated that the psu idic(8)(p23.3) was derived from the maternal chromosome 8. Together, these findings indicate that the fetus was nullisomic for ~2.2 Mb from 8pter, trisomic for the rest of chromosome 8 in mosaic condition, and likely had breaks in MYOM2 repeats of the maternal chromosome 8.


Subject(s)
Abnormalities, Multiple/diagnosis , Homologous Recombination/genetics , Prenatal Diagnosis , Trisomy/diagnosis , Uniparental Disomy/diagnosis , Adult , Chromosomes, Human, Pair 8 , Cytogenetic Analysis , Female , Gestational Age , Humans , In Situ Hybridization, Fluorescence , Magnetic Resonance Imaging , Mosaicism , Pregnancy
5.
Cancer Cell ; 24(6): 751-65, 2013 Dec 09.
Article in English | MEDLINE | ID: mdl-24332043

ABSTRACT

High-grade serous ovarian carcinoma presents significant clinical and therapeutic challenges. Although the traditional model of carcinogenesis has focused on the ovary as a tumor initiation site, recent studies suggest that there may be additional sites of origin outside the ovary, namely the secretory cells of the fallopian tube. Our study demonstrates that high-grade serous tumors can originate in fallopian tubal secretory epithelial cells and also establishes serous tubal intraepithelial carcinoma as the precursor lesion to high-grade serous ovarian and peritoneal carcinomas in animal models targeting the Brca, Tp53, and Pten genes. These findings offer an avenue to address clinically important questions that are critical for cancer prevention and early detection in women carrying BRCA1 and BRCA2 mutations.


Subject(s)
Cell Transformation, Neoplastic , Cystadenocarcinoma, Serous/etiology , Fallopian Tube Neoplasms/pathology , Genes, BRCA1 , Genes, BRCA2 , Ovarian Neoplasms/etiology , Precancerous Conditions/pathology , Animals , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Epithelium/pathology , Female , Genes, p53 , Integrases/genetics , Mice , Mice, Inbred C57BL , Neoplasm Grading , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , PAX8 Transcription Factor , PTEN Phosphohydrolase/genetics , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/physiology
6.
Cell ; 149(5): 959-61, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22632962
7.
Proc Natl Acad Sci U S A ; 109(17): 6686-91, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22496589

ABSTRACT

Copy number variants (CNVs) are a recently recognized class of human germ line polymorphisms and are associated with a variety of human diseases, including cancer. Because of the strong genetic influence on prostate cancer, we sought to identify functionally active CNVs associated with susceptibility of this cancer type. We queried low-frequency biallelic CNVs from 1,903 men of Caucasian origin enrolled in the Tyrol Prostate Specific Antigen Screening Cohort and discovered two CNVs strongly associated with prostate cancer risk. The first risk locus (P = 7.7 × 10(-4), odds ratio = 2.78) maps to 15q21.3 and overlaps a noncoding enhancer element that contains multiple activator protein 1 (AP-1) transcription factor binding sites. Chromosome conformation capture (Hi-C) data suggested direct cis-interactions with distant genes. The second risk locus (P = 2.6 × 10(-3), odds ratio = 4.8) maps to the α-1,3-mannosyl-glycoprotein 4-ß-N-acetylglucosaminyltransferase C (MGAT4C) gene on 12q21.31. In vitro cell-line assays found this gene to significantly modulate cell proliferation and migration in both benign and cancer prostate cells. Furthermore, MGAT4C was significantly overexpressed in metastatic versus localized prostate cancer. These two risk associations were replicated in an independent PSA-screened cohort of 800 men (15q21.3, combined P = 0.006; 12q21.31, combined P = 0.026). These findings establish noncoding and coding germ line CNVs as significant risk factors for prostate cancer susceptibility and implicate their role in disease development and progression.


Subject(s)
Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 15 , Gene Dosage , Genetic Predisposition to Disease , Prostatic Neoplasms/genetics , Case-Control Studies , Cell Line, Tumor , Cell Proliferation , Cohort Studies , Humans , Male , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Prostatic Neoplasms/pathology
8.
J Clin Oncol ; 29(17): 2391-6, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21537050

ABSTRACT

PURPOSE: Prostate-specific antigen screening has led to enormous overtreatment of prostate cancer because of the inability to distinguish potentially lethal disease at diagnosis. We reasoned that by identifying an mRNA signature of Gleason grade, the best predictor of prognosis, we could improve prediction of lethal disease among men with moderate Gleason 7 tumors, the most common grade, and the most indeterminate in terms of prognosis. PATIENTS AND METHODS: Using the complementary DNA-mediated annealing, selection, extension, and ligation assay, we measured the mRNA expression of 6,100 genes in prostate tumor tissue in the Swedish Watchful Waiting cohort (n = 358) and Physicians' Health Study (PHS; n = 109). We developed an mRNA signature of Gleason grade comparing individuals with Gleason ≤ 6 to those with Gleason ≥ 8 tumors and applied the model among patients with Gleason 7 to discriminate lethal cases. RESULTS: We built a 157-gene signature using the Swedish data that predicted Gleason with low misclassification (area under the curve [AUC] = 0.91); when this signature was tested in the PHS, the discriminatory ability remained high (AUC = 0.94). In men with Gleason 7 tumors, who were excluded from the model building, the signature significantly improved the prediction of lethal disease beyond knowing whether the Gleason score was 4 + 3 or 3 + 4 (P = .006). CONCLUSION: Our expression signature and the genes identified may improve our understanding of the de-differentiation process of prostate tumors. Additionally, the signature may have clinical applications among men with Gleason 7, by further estimating their risk of lethal prostate cancer and thereby guiding therapy decisions to improve outcomes and reduce overtreatment.


Subject(s)
Gene Expression Profiling , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Aged , Aged, 80 and over , Cell Differentiation , Disease Progression , Humans , Male , Middle Aged
9.
Neoplasia ; 12(12): 1031-40, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21170267

ABSTRACT

To elucidate the role of ETS gene fusions in castration-resistant prostate cancer (CRPC), we characterized the transcriptome of 54 CRPC tumor samples from men with locally advanced or metastatic disease. Trefoil factor 3 (TFF3) emerged as the most highly differentially regulated gene with respect to ERG rearrangement status and resistance to hormone ablation therapy. Conventional chromatin immunoprecipitation (ChIP)-polymerase chain reaction and ChIP followed by DNA sequencing (ChIP-seq) revealed direct binding of ERG to ETS binding sites in the TFF3 promoter in ERG-rearranged prostate cancer cell lines. These results were confirmed in ERG-rearranged hormone-naive prostate cancer (HNPC) and CRPC tissue samples. Functional studies demonstrated that ERG has an inhibitory effect on TFF3 expression in hormone-naive cancer but not in the castration-resistant state. In addition, we provide evidence suggesting an effect of androgen receptor signaling on ERG-regulated TFF3 expression. Furthermore, TFF3 overexpression enhances ERG-mediated cell invasion in CRPC prostate cancer cells. Taken together, our findings reveal a novel mechanism for enhanced tumor cell aggressiveness resulting from ERG rearrangement in the castration-resistant setting through TFF3 gene expression.


Subject(s)
Peptides/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Trans-Activators/metabolism , Chromatin Immunoprecipitation , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Invasiveness/genetics , Neoplasms, Hormone-Dependent , Peptides/metabolism , Polymerase Chain Reaction , Sequence Analysis, DNA , Signal Transduction , Trans-Activators/genetics , Transcription Factors , Transcriptional Regulator ERG , Trefoil Factor-3
10.
Br J Haematol ; 151(4): 336-45, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20812997

ABSTRACT

Approximately 10% of patients with chronic lymphocytic leukaemia (CLL) have a family history of the disease or a related lymphoproliferative disorder, yet the relationship of familial CLL to genomic abnormalities has not been characterized in detail. We therefore studied 75 CLL patients, half familial and half sporadic, using high-resolution array comparative genomic hybridization (CGH), in order to better define the relationship of genomic abnormalities to familial disease and other biological prognostic factors. Our results showed that the most common high-risk deletion in CLL, deletion 11q, was significantly associated with sporadic disease. Comparison of familial to sporadic disease additionally identified a copy number variant region near the centromere on 14q, proximal to IGH@, in which gains were associated both with familial CLL, and with mutated IGHV and homozygous deletion of 13q. Homozygous deletion of 13q was also found to be associated with mutated IGHV and low expression of ZAP-70, and a significantly longer time to first treatment compared to heterozygous deletion or lack of alteration. This study is the first high resolution effort to investigate and report somatic genetic differences between familial and sporadic CLL.


Subject(s)
Chromosome Aberrations , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Adult , Aged , Chromosome Deletion , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 14/genetics , Comparative Genomic Hybridization/methods , Female , Genetic Predisposition to Disease , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged
11.
BMC Med Genomics ; 3: 8, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20233430

ABSTRACT

BACKGROUND: Current prostate cancer prognostic models are based on pre-treatment prostate specific antigen (PSA) levels, biopsy Gleason score, and clinical staging but in practice are inadequate to accurately predict disease progression. Hence, we sought to develop a molecular panel for prostate cancer progression by reasoning that molecular profiles might further improve current clinical models. METHODS: We analyzed a Swedish Watchful Waiting cohort with up to 30 years of clinical follow up using a novel method for gene expression profiling. This cDNA-mediated annealing, selection, ligation, and extension (DASL) method enabled the use of formalin-fixed paraffin-embedded transurethral resection of prostate (TURP) samples taken at the time of the initial diagnosis. We determined the expression profiles of 6100 genes for 281 men divided in two extreme groups: men who died of prostate cancer and men who survived more than 10 years without metastases (lethals and indolents, respectively). Several statistical and machine learning models using clinical and molecular features were evaluated for their ability to distinguish lethal from indolent cases. RESULTS: Surprisingly, none of the predictive models using molecular profiles significantly improved over models using clinical variables only. Additional computational analysis confirmed that molecular heterogeneity within both the lethal and indolent classes is widespread in prostate cancer as compared to other types of tumors. CONCLUSIONS: The determination of the molecularly dominant tumor nodule may be limited by sampling at time of initial diagnosis, may not be present at time of initial diagnosis, or may occur as the disease progresses making the development of molecular biomarkers for prostate cancer progression challenging.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Biomarkers, Tumor/genetics , Disease Progression , Humans , Male
12.
Nucleic Acids Res ; 38(10): 3275-86, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20156996

ABSTRACT

The detection of copy number variants (CNV) by array-based platforms provides valuable insight into understanding human diversity. However, suboptimal study design and data processing negatively affect CNV assessment. We quantitatively evaluate their impact when short-sequence oligonucleotide arrays are applied (Affymetrix Genome-Wide Human SNP Array 6.0) by evaluating 42 HapMap samples for CNV detection. Several processing and segmentation strategies are implemented, and results are compared to CNV assessment obtained using an oligonucleotide array CGH platform designed to query CNVs at high resolution (Agilent). We quantitatively demonstrate that different reference models (e.g. single versus pooled sample reference) used to detect CNVs are a major source of inter-platform discrepancy (up to 30%) and that CNVs residing within segmental duplication regions (higher reference copy number) are significantly harder to detect (P < 0.0001). After adjusting Affymetrix data to mimic the Agilent experimental design (reference sample effect), we applied several common segmentation approaches and evaluated differential sensitivity and specificity for CNV detection, ranging 39-77% and 86-100% for non-segmental duplication regions, respectively, and 18-55% and 39-77% for segmental duplications. Our results are relevant to any array-based CNV study and provide guidelines to optimize performance based on study-specific objectives.


Subject(s)
DNA Copy Number Variations , Oligonucleotide Array Sequence Analysis/methods , Algorithms , Base Sequence , Humans , Oligonucleotide Array Sequence Analysis/standards , Oligonucleotide Probes/chemistry , Polymorphism, Single Nucleotide
13.
Cancer Epidemiol Biomarkers Prev ; 19(1): 229-39, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20056642

ABSTRACT

PURPOSE: Dihydrotestosterone (DHT) is an important factor in prostate cancer (PCA) genesis and disease progression. Given PCA's strong genetic component, we evaluated the possibility that variation in genes involved in DHT metabolism influence PCA risk. EXPERIMENTAL DESIGN: We investigated copy number variants (CNV) and single nucleotide polymorphisms (SNP). We explored associations between CNV of uridine diphospho-glucuronosyltransferase (UGT) genes from the 2B subclass, given their prostate specificity and/or involvement in steroid metabolism and PCA risk. We also investigated associations between SNPs in genes (HSD3B1, SRD5A1/2, and AKR1C2) involved in the conversion of testosterone to DHT, and in DHT metabolism and PCA risk. The population consisted of 426 men (205 controls and 221 cases) who underwent prostate-specific antigen screening as part of a PCA early detection program in Tyrol, Austria. RESULTS: No association between CNV in UGT2B17 and UGT2B28 and PCA risk was identified. Men carrying the AA genotype at SNP rs6428830 (HSD3B1) had an odds ratio (OR) of 2.0 [95% confidence intervals (95% CI), 1.1-4.1] compared with men with GG, and men with AG or GG versus AA in rs1691053 (SRD5A1) had an OR of 1.8 (95% CI, 1.04-3.13). Individuals carrying both risk alleles had an OR of 3.1 (95% CI, 1.4-6.7) when compared with men carrying neither (P = 0.005). Controls with the AA genotype on rs7594951 (SRD5A2) tended toward higher serum DHT levels (P = 0.03). CONCLUSIONS: This is the first study to implicate the 5alpha-reductase isoform 1 (SRD5A1) and PCA risk, supporting the rationale of blocking enzymatic activity of both isoforms of 5alpha-reductase for PCA chemoprevention.


Subject(s)
Dihydrotestosterone/metabolism , Genetic Predisposition to Disease , Prostatic Neoplasms/genetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Adult , Aged , Biomarkers, Tumor/genetics , DNA-Binding Proteins/genetics , Gene Dosage , Genotype , Glucuronosyltransferase/genetics , Humans , Hydroxysteroid Dehydrogenases/genetics , Male , Middle Aged , Minor Histocompatibility Antigens , Polymorphism, Single Nucleotide , Prostate-Specific Antigen/blood , Prostatic Neoplasms/metabolism , Risk Factors , Testosterone/metabolism
15.
Cancer Res ; 69(7): 2734-8, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19293179

ABSTRACT

Chromosomal rearrangements account for all erythroblast transformation-specific (ETS) family member gene fusions that have been reported in prostate cancer and have clinical, diagnostic, and prognostic implications. Androgen-regulated genes account for the majority of the 5' genomic regulatory promoter elements fused with ETS genes. TMPRSS2-ERG, TMPRSS2-ETV1, and SLC45A3-ERG rearrangements account for roughly 90% of ETS fusion prostate cancer. ELK4, another ETS family member, is androgen regulated, involved in promoting cell growth, and highly expressed in a subset of prostate cancer, yet the mechanism of ELK4 overexpression is unknown. In this study, we identified a novel ETS family fusion transcript, SLC45A3-ELK4, and found it to be expressed in both benign prostate tissue and prostate cancer. We found high levels of SLC45A3-ELK4 mRNA restricted to a subset of prostate cancer samples. SLC45A3-ELK4 transcript can be detected at high levels in urine samples from men at risk for prostate cancer. Characterization of the fusion mRNA revealed a major variant in which SLC45A3 exon 1 is fused to ELK4 exon 2. Based on quantitative PCR analyses of DNA, unlike other ETS fusions described in prostate cancer, the expression of SLC45A3-ELK4 mRNA is not exclusive to cases harboring a chromosomal rearrangement. Treatment of LNCaP cancer cells with a synthetic androgen (R1881) revealed that SLC45A3-ELK4, and not endogenous ELK4, mRNA expression is androgen regulated. Altogether, our findings show that SLC45A3-ELK4 mRNA expression is heterogeneous, highly induced in a subset of prostate cancers, androgen regulated, and most commonly occurs through a mechanism other than chromosomal rearrangement (e.g., trans-splicing).


Subject(s)
Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , RNA, Messenger/genetics , Cell Line, Tumor , Chromosome Deletion , Chromosomes, Human, Pair 1 , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Situ Hybridization, Fluorescence , Male , Metribolone/pharmacology , Oncogene Proteins, Fusion/biosynthesis , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-ets/biosynthesis , Proto-Oncogene Proteins c-ets/genetics , RNA, Messenger/biosynthesis , Testosterone Congeners/pharmacology , Transcription, Genetic
16.
Genes Chromosomes Cancer ; 48(4): 366-80, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19156837

ABSTRACT

Emerging molecular and clinical data suggest that ETS fusion prostate cancer represents a distinct molecular subclass, driven most commonly by a hormonally regulated promoter and characterized by an aggressive natural history. The study of the genomic landscape of prostate cancer in the light of ETS fusion events is required to understand the foundation of this molecularly and clinically distinct subtype. We performed genome-wide profiling of 49 primary prostate cancers and identified 20 recurrent chromosomal copy number aberrations, mainly occurring as genomic losses. Co-occurring events included losses at 19q13.32 and 1p22.1. We discovered three genomic events associated with ERG rearranged prostate cancer, affecting 6q, 7q, and 16q. 6q loss in nonrearranged prostate cancer is accompanied by gene expression deregulation in an independent dataset and by protein deregulation of MYO6. To analyze copy number alterations within the ETS genes, we performed a comprehensive analysis of all 27 ETS genes and of the 3 Mbp genomic area between ERG and TMPRSS2 (21q) with an unprecedented resolution (30 bp). We demonstrate that high-resolution tiling arrays can be used to pin-point breakpoints leading to fusion events. This study provides further support to define a distinct molecular subtype of prostate cancer based on the presence of ETS gene rearrangements.


Subject(s)
Chromosome Aberrations , Gene Rearrangement , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-ets/genetics , Trans-Activators/genetics , Cell Line, Tumor , Chromosome Mapping , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Male , Oligonucleotide Array Sequence Analysis , Transcriptional Regulator ERG
17.
Diagn Mol Pathol ; 17(4): 207-13, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18936709

ABSTRACT

The aim of the current study was to analyze gene expression profiles in benign prostatic hyperplasia and to compare them with phenotypic properties. Thirty-seven specimens of benign prostatic hyperplasia were obtained from symptomatic patients undergoing surgery. RNA was extracted and hybridized to Affymetrix Chips containing 54,000 gene expression probes. Gene expression profiles were analyzed using cluster, TreeView, and significance analysis of microarrays softwares. In an initial unsupervised analysis, our 37 samples clustered hierarchically in 2 groups of 18 and 19 samples, respectively. Five clinical parameters were statistically different between the 2 groups: in group 1 compared with group 2, patients had larger prostate glands, had higher prostate specific antigen levels, were more likely to be treated by alpha blockers, to be operated by prostatectomy, and to have major irritative symptoms. The sole independent parameter associated with this dichotome clustering, however, was the prostate gland volume. Therefore, the role of prostate volume was explored in a supervised analysis. Gene expression of prostate glands <60 mL and >60 mL were compared using significance analysis of microarrays and 227 genes were found differentially expressed between the 2 groups (>2 change and false discovery rate of <5%). Several specific pathways including growth factors genes, cell cycle genes, apoptose genes, inflammation genes, and androgen regulated genes, displayed major differences between small and large prostate glands.


Subject(s)
Gene Expression Profiling , Prostate/pathology , Prostatic Hyperplasia/pathology , Aged , Aged, 80 and over , Cluster Analysis , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Organ Size , Prostate/physiology , Prostate-Specific Antigen/blood , Prostatic Hyperplasia/physiopathology , Up-Regulation
18.
J Natl Cancer Inst ; 100(11): 815-25, 2008 Jun 04.
Article in English | MEDLINE | ID: mdl-18505969

ABSTRACT

BACKGROUND: The majority of prostate cancers harbor gene fusions of the 5'-untranslated region of the androgen-regulated transmembrane protease serine 2 (TMPRSS2) promoter with erythroblast transformation-specific transcription factor family members. The common fusion between TMPRESS2 and v-ets erythroblastosis virus E26 oncogene homolog (avian) (ERG) is associated with a more aggressive clinical phenotype, implying the existence of a distinct subclass of prostate cancer defined by this fusion. METHODS: We used complementary DNA-mediated annealing, selection, ligation, and extension to determine the expression profiles of 6144 transcriptionally informative genes in archived biopsy samples from 455 prostate cancer patients in the Swedish Watchful Waiting cohort (1987-1999) and the United States-based Physicians(') Health Study cohort (1983-2003). A gene expression signature for prostate cancers with the TMPRSS2-ERG fusion was determined using partitioning and classification models and used in computational functional analysis. Cell proliferation and TMPRSS2-ERG expression in androgen receptor-negative (NCI-H660) prostate cancer cells after treatment with vehicle or estrogenic compounds were assessed by viability assays and quantitative polymerase chain reaction, respectively. All statistical tests were two-sided. RESULTS: We identified an 87-gene expression signature that distinguishes TMPRSS2-ERG fusion prostate cancer as a discrete molecular entity (area under the curve = 0.80, 95% confidence interval [CI] = 0.792 to 0.81; P < .001). Computational analysis suggested that this fusion signature was associated with estrogen receptor (ER) signaling. Viability of NCI-H660 cells decreased after treatment with estrogen (viability normalized to day 0, estrogen vs vehicle at day 8, mean = 2.04 vs 3.40, difference = 1.36, 95% CI = 1.12 to 1.62) or ERbeta agonist (ERbeta agonist vs vehicle at day 8, mean = 1.86 vs 3.40, difference = 1.54, 95% CI = 1.39 to 1.69) but increased after ERalpha agonist treatment (ERalpha agonist vs vehicle at day 8, mean = 4.36 vs 3.40, difference = 0.96, 95% CI = 0.68 to 1.23). Similarly, expression of TMPRSS2-ERG decreased after ERbeta agonist treatment (fold change over internal control, ERbeta agonist vs vehicle at 24 hours, NCI-H660, mean = 0.57- vs 1.0-fold, difference = 0.43-fold, 95% CI = 0.29- to 0.57-fold) and increased after ERalpha agonist treatment (ERalpha agonist vs vehicle at 24 hours, mean = 5.63- vs 1.0-fold, difference = 4.63-fold, 95% CI = 4.34- to 4.92-fold). CONCLUSIONS: TMPRSS2-ERG fusion prostate cancer is a distinct molecular subclass. TMPRSS2-ERG expression is regulated by a novel ER-dependent mechanism.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/agonists , Estrogens/metabolism , Neoplasms, Hormone-Dependent/metabolism , Oncogene Proteins, Fusion/metabolism , Prostatic Neoplasms/metabolism , Signal Transduction , Antineoplastic Agents, Hormonal/therapeutic use , Area Under Curve , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Chromatin Immunoprecipitation , DNA, Complementary/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Health Surveys , Humans , Male , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/pathology , Nitriles/pharmacology , Phenols , Physicians/statistics & numerical data , Polymerase Chain Reaction , Propionates/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Pyrazoles/pharmacology , Serine Endopeptidases/metabolism , Signal Transduction/drug effects , Sweden/epidemiology , Transfection
19.
Cancer Res ; 68(7): 2286-91, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18381435

ABSTRACT

Early detection and diagnosis of prostate cancer is key to designing effective treatment strategies. Microarrays have resulted in the discovery of hepsin (HPN) as a biomarker for detection of prostate cancer. In this study, we explore the development of HPN imaging probes for detection of prostate cancer. We used phage display to isolate HPN binding peptides with 190 + 2.2 nmol/L affinity in monomeric form and high specificity. The identified peptides were able to detect human prostate cancer on tissue microarrays and in cell-based assays. HPN-targeted imaging agents were synthesized by conjugating multiple peptides to fluorescent nanoparticles to further improve avidity through multivalency and to improve pharmacokinetics. When injected into mouse xenograft models, HPN-targeted nanoparticles bound specifically to HPN-expressing LNCaP xenografts compared with non-HPN-expressing PC3 xenografts. HPN imaging may provide a new method for detection of prostate cancer.


Subject(s)
Biomarkers, Tumor/analysis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/enzymology , Serine Endopeptidases/analysis , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , Peptide Library , Prostatic Neoplasms/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics , Transfection
20.
Cancer Res ; 67(21): 10296-303, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17974971

ABSTRACT

Microarrays have been used to identify genes involved in cancer progression. We have now developed an algorithm that identifies dysregulated pathways from multiple expression array data sets without a priori definition of gene expression thresholds. Integrative microarray analysis of pathways (IMAP) was done using existing expression array data from localized and metastatic prostate cancer. Comparison of metastatic cancer and localized disease in multiple expression array profiling studies using the IMAP approach yielded a list of about 100 pathways that were significantly dysregulated (P < 0.05) in prostate cancer metastasis. The pathway that showed the most significant dysregulation, HIV-I NEF, was validated at both the transcript level and the protein level by quantitative PCR and immunohistochemical analysis, respectively. Validation by unsupervised analysis on an independent data set using the gene expression signature from the HIV-I NEF pathway verified the accuracy of our method. Our results indicate that this pathway is especially dysregulated in hormone-refractory prostate cancer.


Subject(s)
Oligonucleotide Array Sequence Analysis/methods , Prostatic Neoplasms/pathology , Signal Transduction/physiology , Gene Expression Regulation, Neoplastic , Humans , Male , NF-kappa B/physiology , NF-kappa B p50 Subunit/physiology , Neoplasm Metastasis , Prostatic Neoplasms/genetics , nef Gene Products, Human Immunodeficiency Virus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL