Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Nat Commun ; 12(1): 5307, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489465

ABSTRACT

Prostate cancer is heterogeneous and patients would benefit from methods that stratify those who are likely to respond to systemic therapy. Here, we employ single-cell assays for transposase-accessible chromatin (ATAC) and RNA sequencing in models of early treatment response and resistance to enzalutamide. In doing so, we identify pre-existing and treatment-persistent cell subpopulations that possess regenerative potential when subjected to treatment. We find distinct chromatin landscapes associated with enzalutamide treatment and resistance that are linked to alternative transcriptional programs. Transcriptional profiles characteristic of persistent cells are able to stratify the treatment response of patients. Ultimately, we show that defining changes in chromatin and gene expression in single-cell populations from pre-clinical models can reveal as yet unrecognized molecular predictors of treatment response. This suggests that the application of single-cell methods with high analytical resolution in pre-clinical models may powerfully inform clinical decision-making.


Subject(s)
Chromatin/chemistry , DNA, Neoplasm/genetics , Drug Resistance, Neoplasm/genetics , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Transcriptome , Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Cell Line, Tumor , Chromatin/metabolism , DNA, Neoplasm/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Proteins/metabolism , Nitriles/therapeutic use , Phenylthiohydantoin/therapeutic use , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Survival Analysis , Exome Sequencing
2.
Nat Commun ; 11(1): 5070, 2020 10 08.
Article in English | MEDLINE | ID: mdl-33033260

ABSTRACT

The evolutionary progression from primary to metastatic prostate cancer is largely uncharted, and the implications for liquid biopsy are unexplored. We infer detailed reconstructions of tumor phylogenies in ten prostate cancer patients with fatal disease, and investigate them in conjunction with histopathology and tumor DNA extracted from blood and cerebrospinal fluid. Substantial evolution occurs within the prostate, resulting in branching into multiple spatially intermixed lineages. One dominant lineage emerges that initiates and drives systemic metastasis, where polyclonal seeding between sites is common. Routes to metastasis differ between patients, and likely genetic drivers of metastasis distinguish the metastatic lineage from the lineage that remains confined to the prostate within each patient. Body fluids capture features of the dominant lineage, and subclonal expansions that occur in the metastatic phase are non-uniformly represented. Cerebrospinal fluid analysis reveals lineages not detected in blood-borne DNA, suggesting possible clinical utility.


Subject(s)
Cell Lineage , Liquid Biopsy , Prostatic Neoplasms/pathology , Body Fluids/metabolism , Chromosomes, Human, Pair 8/genetics , Clone Cells , DNA Copy Number Variations/genetics , DNA, Neoplasm/genetics , Genetic Loci , Humans , Male , Middle Aged , Neoplasm Metastasis , Phylogeny
3.
Transl Oncol ; 11(5): 1160-1170, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30056367

ABSTRACT

Ovarian cancer has the highest mortality rate of all gynecologic malignancies. Identification of new biomarkers is highly needed due to its late diagnosis and high recurrence rate. The objective of this study was to identify mechanisms of therapy resistance and potential biomarkers by analyzing mRNA and protein expression from samples derived from patients with platinum-sensitive and -resistant ovarian cancer (total cohort n = 53). The data revealed new candidates for targeted therapies, such as GREB1 and ROR2. We showed that the development of platinum resistance correlated with upregulation of ROR2, whereas GREB1 was downregulated. Moreover, we demonstrated that high levels of ROR2 in platinum-resistant samples were associated with upregulation of Wnt5a, STAT3 and NF-kB levels, suggesting that a crosstalk between the non-canonical Wnt5a-ROR2 and STAT3/NF-kB signaling pathways. Upregulation of ROR2, Wnt5a, STAT3 and NF-kB was further detected in a platinum-resistant cell-line model. The results of the present study provided insight into molecular mechanisms associated with platinum resistance that could be further investigated to improve treatment strategies in this clinically challenging gynecological cancer.

4.
Prostate Cancer Prostatic Dis ; 19(4): 367-373, 2016 12.
Article in English | MEDLINE | ID: mdl-27502739

ABSTRACT

BACKGROUND: To evaluate the association between use of metformin and other antidiabetic drugs with tumor characteristics and survival in surgically managed prostate cancer (PCa) patients. METHODS: The study population included 1314 men who underwent radical prostatectomy at the Tampere University Hospital during 1995-2009. Causes of deaths were collected from the Finnish Cancer Registry. Individual-level data on medication use during 1995-2009 was obtained from national prescription database. Fasting blood glucose and hemoglobin A1c values during the study period were gathered from hospital district database. Gleason grade and pathological stage were compared by drug use before surgery and separately by metformin usage. Risk of biochemical recurrence, all-cause death and PCa-specific death were calculated using Cox proportional hazard regression with adjustment for age, tumor characteristics, glycemic control and use of other drug groups. RESULTS: High-grade tumors were more common among antidiabetic drug users (P=0.032), including metformin users (P=0.012). Despite this, no difference in PSA levels was observed. Men who had used antidiabetic drugs before surgery had an increased risk of Gleason 7-10 disease (odds ratio (OR) 1.83, 95% confidence interval (CI) 1.04-3.23). The risk of high-grade PCa was higher among metformin users compared with other antidiabetic drug users (OR 3.11, 95% CI 1.16-8.33). During the median follow-up of 8.6 years after surgery, 551 men had biochemical recurrence and 244 died, 32 owing to PCa. Generally, no association with risk of disease recurrence was observed. Risk of death was increased by preoperative use of antidiabetic drugs (hazard ratio 1.81 95% CI 1.03-3.19), but no survival associations for postoperative use of antidiabetic drugs or metformin were observed. CONCLUSION: Diabetic men have more high-grade PCa at lower PSA levels, but that does not have a clear impact on disease-specific survival in the short term even when glycemic control is being considered.


Subject(s)
Diabetes Mellitus/drug therapy , Hypoglycemic Agents/therapeutic use , Prostatic Neoplasms/pathology , Adult , Aged , Blood Glucose/drug effects , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Glycated Hemoglobin/metabolism , Humans , Male , Metformin/therapeutic use , Middle Aged , Neoplasm Grading/methods , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Prostate/drug effects , Prostate/pathology , Prostate-Specific Antigen/blood , Prostatectomy/methods , Prostatic Neoplasms/mortality , Prostatic Neoplasms/surgery
5.
Cancer Res ; 75(11): 2326-36, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25858144

ABSTRACT

Altered microRNA (miRNA; miR) expression is associated with tumor formation and progression of various solid cancers. A major challenge in miRNA expression profiling of bulk tumors is represented by the heterogeneity of the subpopulations of cells that constitute the organ, as well as the tumor tissue. Here, we analyzed the expression of miRNAs in a subpopulation of epithelial stem/progenitor-like cells in human prostate cancer [prostate cancer stem cell (PCSC)] and compared their expression profile to more differentiated cancer cells. In both cell lines and clinical prostate cancer specimens, we identified that miR-25 expression in PCSCs was low/absent and steadily increased during their differentiation into cells with a luminal epithelial phenotype. Functional studies revealed that overexpression of miR-25 in prostate cancer cell lines and selected subpopulation of highly metastatic and tumorigenic cells (ALDH(high)) strongly affected the invasive cytoskeleton, causing reduced migration in vitro and metastasis via attenuation of extravasation in vivo. Here, we show, for the first time, that miR-25 can act as a tumor suppressor in highly metastatic PCSCs by direct functional interaction with the 3'-untranslated regions of proinvasive αv- and α6-integrins. Taken together, our observations suggest that miR-25 is a key regulator of invasiveness in human prostate cancer through its direct interactions with αv- and α6-integrin expression.


Subject(s)
Integrin alpha6/biosynthesis , Integrin alphaV/biosynthesis , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic/genetics , Humans , Integrin alpha6/genetics , Integrin alphaV/genetics , Male , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology
6.
Br J Cancer ; 106(4): 768-74, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22240788

ABSTRACT

BACKGROUND: The aim of this study was to investigate the hypothesis that changes in circulating microRNAs (miRs) represent potentially useful biomarkers for the diagnosis, staging and prediction of outcome in prostate cancer. METHODS: Real-time polymerase chain reaction analysis of 742 miRs was performed using plasma-derived circulating microvesicles of 78 prostate cancer patients and 28 normal control individuals to identify differentially quantified miRs. RESULTS: A total of 12 miRs were differentially quantified in prostate cancer patients compared with controls, including 9 in patients without metastases. In all, 11 miRs were present in significantly greater amounts in prostate cancer patients with metastases compared with those without metastases. The association of miR-141 and miR-375 with metastatic prostate cancer was confirmed using serum-derived exosomes and microvesicles in a separate cohort of patients with recurrent or non-recurrent disease following radical prostatectomy. An analysis of five selected miRs in urine samples found that miR-107 and miR-574-3p were quantified at significantly higher concentrations in the urine of men with prostate cancer compared with controls. CONCLUSION: These observations suggest that changes in miR concentration in prostate cancer patients may be identified by analysing various body fluids. Moreover, circulating miRs may be used to diagnose and stage prostate cancer.


Subject(s)
MicroRNAs/blood , Prostatic Neoplasms/genetics , Aged , Biomarkers, Tumor/genetics , Humans , Male , MicroRNAs/urine , Neoplasm Metastasis , Prognosis , Prostate-Specific Antigen/analysis , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology
7.
Oncogene ; 31(41): 4460-71, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-22266859

ABSTRACT

The androgen receptor (AR) signaling pathway is involved in the emergence of castration-resistant prostate cancer (CRPC). Here, we identified several androgen-regulated microRNAs (miRNAs) that may contribute to the development of CRPC. Seven miRNAs, miR-21, miR-32, miR-99a, miR-99b, miR-148a, miR-221 and miR-590-5p, were found to be differentially expressed in CRPC compared with benign prostate hyperplasia (BPH) according to microarray analyses. Significant growth advantage for LNCaP cells transfected with pre-miR-32 and pre-miR-148a was found. miR-32 was demonstrated to reduce apoptosis, whereas miR-148a enhanced proliferation. Androgen regulation of miR-32 and miR-148a was confirmed by androgen stimulation of the LNCaP cells followed by expression analyses. The AR-binding sites in proximity of these miRNAs were demonstrated with chromatin immunoprecipitation (ChIP). To identify target genes for the miRNAs, mRNA microarray analyses were performed with LNCaP cells transfected with pre-miR-32 and pre-miR-148a. Expression of BTG2 and PIK3IP1 was reduced in the cells transfected with pre-miR-32 and pre-miR-148a, respectively. Also, the protein expression was reduced according to western blot analysis. BTG2 and PIK3IP1 were confirmed to be targets by 3'UTR-luciferase assays. Finally, immunostainings showed a statistically significant (P<0.0001) reduction of BTG2 protein in CRPCs compared with untreated prostate cancer (PC). The lack of BTG2 staining was also associated (P<0.01) with a short progression-free time in patients who underwent prostatectomy. In conclusion, androgen-regulated miR-32 is overexpressed in CRPC, leading to reduced expression of BTG2. Thus, miR-32 is a potential marker for aggressive disease and is a putative drug target in PC.


Subject(s)
Gene Expression Regulation, Neoplastic , Immediate-Early Proteins/genetics , MicroRNAs/metabolism , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , Tumor Suppressor Proteins/genetics , Androgens/physiology , Binding Sites , Case-Control Studies , Cell Line, Tumor , Cell Proliferation , Humans , Immediate-Early Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/genetics , Neoplasms, Hormone-Dependent/genetics , Oligonucleotide Array Sequence Analysis , Prognosis , Prostatic Neoplasms/genetics , Receptors, Androgen/metabolism , Regulatory Sequences, Nucleic Acid , Signal Transduction , Transcriptome , Tumor Suppressor Proteins/metabolism
8.
Oncogene ; 31(17): 2153-63, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-21909140

ABSTRACT

Androgen receptor (AR) is overexpressed in the majority of castration-resistant prostate cancers (CRPCs). Our goal was to study the effect of AR overexpression on the chromatin binding of the receptor and to identify AR target genes that may be important in the emergence of CRPC. We have established two sublines of LNCaP prostate cancer (PC) cell line, one overexpressing AR 2-3-fold and the other 4-5-fold compared with the control cells. We used chromatin immunoprecipitation (ChIP) and deep-sequencing (seq) to identify AR-binding sites (ARBSs). We found that the number of ARBSs and the AR-binding strength were positively associated with the level of AR when cells were stimulated with low concentrations of androgens. In cells overexpressing AR, the chromatin binding of the receptor took place in 100-fold lower concentration of the ligand than in control cells. We confirmed the association of AR level and chromatin binding in two PC xenografts, one containing AR gene amplification with high AR expression, and the other with low expression. By combining the ChIP-seq and expression profiling, we identified AR target genes that are upregulated in PC. Of them, the expression of ZWINT, SKP2 (S-phase kinase-associated protein 2 (p45)) and FEN1 (flap structure-specific endonuclease 1) was demonstrated to be increased in CRPC, while the expression of SNAI2 was decreased in both PC and CRPC. FEN1 protein expression was also associated with poor prognosis in prostatectomy-treated patients. Finally, the knock-down of FEN1 with small interfering RNA inhibited the growth of LNCaP cells. Our data demonstrate that the overexpression of AR sensitizes the receptor binding to chromatin, thus, explaining how AR signaling pathway is reactivated in CRPC cells.


Subject(s)
Chromatin/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Animals , Binding Sites , Cell Line, Tumor , Flap Endonucleases/genetics , Gene Amplification , Gene Expression Profiling , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Nuclear Proteins/genetics , Nucleic Acid Amplification Techniques , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , S-Phase Kinase-Associated Proteins/genetics , Transplantation, Heterologous
9.
Oncogene ; 31(8): 978-91, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-21765474

ABSTRACT

Prostate cancer (PCa) is the most frequent male malignancy and the second most common cause of cancer-related death in Western countries. Current clinical and pathological methods are limited in the prediction of postoperative outcome. It is becoming increasingly evident that small non-coding RNA (ncRNA) species are associated with the development and progression of this malignancy. To assess the diversity and abundance of small ncRNAs in PCa, we analyzed the composition of the entire small transcriptome by Illumina/Solexa deep sequencing. We further analyzed the microRNA (miRNA) expression signatures of 102 fresh-frozen patient samples during PCa progression by miRNA microarrays. Both platforms were cross-validated by quantitative reverse transcriptase-PCR. Besides the altered expression of several miRNAs, our deep sequencing analyses revealed strong differential expression of small nucleolar RNAs (snoRNAs) and transfer RNAs (tRNAs). From microarray analysis, we derived a miRNA diagnostic classifier that accurately distinguishes normal from cancer samples. Furthermore, we were able to construct a PCa prognostic predictor that independently forecasts postoperative outcome. Importantly, the majority of miRNAs included in the predictor also exhibit high sequence counts and concordant differential expression in Illumina PCa samples, supported by quantitative reverse transcriptase-PCR. Our findings provide miRNA expression signatures that may serve as an accurate tool for the diagnosis and prognosis of PCa.


Subject(s)
Biomarkers, Tumor/metabolism , MicroRNAs/metabolism , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Transcriptome , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Gene Expression , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , MicroRNAs/genetics , Middle Aged , Oligonucleotide Array Sequence Analysis , Primary Cell Culture , Prognosis , Prostate/metabolism , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , ROC Curve , Real-Time Polymerase Chain Reaction
10.
Oncogene ; 27(36): 4865-76, 2008 Aug 21.
Article in English | MEDLINE | ID: mdl-18469865

ABSTRACT

Transcriptional control by androgens via androgen receptor (AR) is strongly involved in prostate cancer development, but the critical target genes have remained elusive. We have characterized E twenty-six-like transcription factor 4 (ELK4) (also known as serum response factor accessory protein 1) as a novel AR target in human prostate cancer cells. In-silico screening identified three putative AR response elements (AREs) within -10 kb from the transcription start site of ELK4. Both ARE1 at -167/-153 and ARE2 at -481/-467 bound AR in vitro and mediated androgen induction as isolated elements in transcription assays in non-prostate cells. However, merely the ARE2 that cooperates with a proximal forkhead box A1-binding site was critical for the AR-dependent activation of ELK4 promoter in prostate cancer cells. Preferential loading of holo-AR onto the ARE2 and concomitant recruitment of RNA polymerase II onto the ELK4 promoter was confirmed in prostate cancer cells by chromatin immunoprecipitation. Database searches indicated that the expression of ELK4 is markedly increased in prostate cancers relative to normal prostates. Moreover, prostate cancer tissue immunostainings showed that nuclear ELK4 levels are significantly increased in androgen-refractory prostate cancers compared to untreated tumours. Reduction of the amount of ELK4 in LNCaP cells by RNAi retarded cell growth. In conclusion, ELK4 is a direct AR target in prostate cancer cells. Androgens may thus contribute to the growth of prostate cancer via influencing ELK4 levels.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , ets-Domain Protein Elk-4/metabolism , Androgens/physiology , Cell Line, Tumor , Humans , Male , Promoter Regions, Genetic , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , ets-Domain Protein Elk-4/biosynthesis , ets-Domain Protein Elk-4/genetics
11.
Rheumatology (Oxford) ; 46(10): 1538-46, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17726036

ABSTRACT

OBJECTIVES: Cytokines play a key pathogenic role in rheumatoid arthritis (RA). Several cytokines signal through the JAK-STAT pathway, which is negatively regulated by the suppressors of cytokine signalling (SOCS) proteins. Since SOCS protein levels can profoundly modulate cellular responses to cytokines, we have investigated their expression in chronic RA. METHODS: The levels of SOCS1-3 and CIS1 mRNA in peripheral blood (PB) and synovial fluid (SF) mononuclear cells (MCs), purified T cells and monocytes from RA patients and healthy volunteers were studied using quantitative reverse transcriptase polymerase chain reaction (RT-PCR). SOCS mRNA and protein expression in synovial tissues were examined by RT-PCR and immunohistochemistry. RESULTS: The levels of SOCS1 and SOCS3 were significantly increased in PBMCs from RA patients when compared with healthy volunteers. These differences were mainly due to up-regulation of SOCS1 in PB T cells and of SOCS3 in PB monocytes. In addition, SOCS2 was up-regulated in PB T cells. Interestingly, SF T cells expressed lower and SF macrophages higher levels of SOCS molecules than their PB counterparts. Similarly, while a significant portion of macrophages in synovial tissues expressed SOCS1 and SOCS3 proteins, the majority of T cells remained SOCS negative. Finally, SOCS1 was up-regulated in the synovial membranes from patients with RA when compared with osteoarthritis. CONCLUSIONS: SOCS expression levels are profoundly altered in RA, and the profile of SOCS expression is dependent on both the cell type as well as the cellular compartment.


Subject(s)
Arthritis, Rheumatoid/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Adult , Aged , Aged, 80 and over , Cells, Cultured , Cytokines/pharmacology , Female , Gene Expression Regulation , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Monocytes/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Synovial Fluid/metabolism , Synovial Membrane/metabolism , T-Lymphocyte Subsets/metabolism , Up-Regulation
12.
J Pathol ; 212(4): 395-401, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17503439

ABSTRACT

BAG-1L (Bcl-2-associated anthanogene 1) has been found to interact with androgen receptor (AR), and has been suggested to be involved in the development of prostate cancer. In order to determine the presence of genetic and/or expression alterations of BAG-1L in prostate cancer, we analysed human prostate cancer cell lines and xenografts as well as patient samples of untreated, hormone-naïve, and hormone-refractory prostate carcinomas for sequence variations using denaturing high-performance liquid chromatography (DHPLC), for gene copy number using fluorescence in situ hybridization (FISH), and for expression using both quantitative RT-PCR and immunostaining. Only one sequence variation was found in all 37 cell lines and xenografts analysed. BAG-1 gene amplification was detected in two xenografts. In addition, gene amplification was found in 6 of 81 (7.4%) hormone-refractory clinical tumours, whereas no amplification was found in any of the 130 untreated tumours analysed. Additionally, gain of the BAG-1 gene was observed in 27.2% of the hormone-refractory tumours and in 18.5% of the untreated carcinomas. In a set of 263 patient samples, BAG-1L protein expression was significantly higher in hormone-refractory tumours than in primary tumours (p = 0.002). Altogether, these data suggest that amplification and overexpression of BAG-1L may be involved in the progression of prostate cancer.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Transcription Factors/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , Cell Line, Tumor , Chromatography, High Pressure Liquid/methods , DNA Mutational Analysis/methods , DNA-Binding Proteins/genetics , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Proteins/genetics , Neoplasm Transplantation , Orchiectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription Factors/genetics , Transplantation, Heterologous , Treatment Failure
13.
Br J Cancer ; 96(2): 352-6, 2007 Jan 29.
Article in English | MEDLINE | ID: mdl-17242703

ABSTRACT

Germline mutations in the aryl hydrocarbon receptor interacting protein (AIP) gene were recently identified in individuals with pituitary adenoma predisposition (PAP). These patients have prolactin (PRL) or growth hormone (GH) oversecreting pituitary adenomas, the latter exhibiting acromegaly or gigantism. Loss-of-heterozygosity (LOH) analysis revealed that AIP is lost in PAP tumours, suggesting that it acts as a tumour-suppressor gene. Aryl hydrocarbon receptor interacting protein is involved in several pathways, but it is best characterised as a cytoplasmic partner of the aryl hydrocarbon receptor (AHR). To examine the possible role of AIP in the genesis of common cancers, we performed somatic mutation screening in a series of 373 colorectal cancers (CRCs), 82 breast cancers, and 44 prostate tumour samples. A missense R16H (47G>A) change was identified in two CRC samples, as well as in the respective normal tissues, but was absent in 209 healthy controls. The remaining findings were silent, previously unreported, changes of the coding, non-coding, or untranslated regions of AIP. These results suggest that somatic AIP mutations are not common in CRC, breast, and prostate cancers.


Subject(s)
Breast Neoplasms/genetics , Colorectal Neoplasms/genetics , Mutation , Prostatic Neoplasms/genetics , Proteins/genetics , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Humans , Intracellular Signaling Peptides and Proteins , Loss of Heterozygosity , Male , Middle Aged , Molecular Sequence Data , Proteins/chemistry , Sequence Homology, Amino Acid
14.
Oncogene ; 25(9): 1358-66, 2006 Mar 02.
Article in English | MEDLINE | ID: mdl-16331274

ABSTRACT

The incidence of prostate cancer is increasing in western countries because of population aging. Prostate cancer begins as an androgen-dependent disease, but it can become androgen independent at a later stage or in tumors recurring after an antihormonal treatment. Although many genetic events have been described to be involved in androgen-dependent and/or -independent prostate cancer growth, little is known about the contribution of epigenetic events. Here we have examined the possibility that the methyl-CpG-binding protein MECP2 might play a role in controlling the growth of prostate cancer cells. Inhibition of MECP2 expression by stable short hairpin RNA stopped the growth of both normal and cancer human prostate cells. In addition, ectopic expression of the MECP2 conferred a growth advantage to human prostate cancer cells. More importantly, this expression allowed androgen-dependent cells to grow independently of androgen stimulation and to retain tumorigenic properties in androgen-depleted conditions. Analysis of signaling pathways showed that this effect is independent of androgen receptor signaling. Instead, MECP2 appears to act by maintaining a constant c-myc level during antihormonal treatment. We further show that MECP2-expressing cells possess a functional p53 pathway and are still responsive to chemotherapeutic drugs.


Subject(s)
Cell Proliferation , Methyl-CpG-Binding Protein 2/physiology , Prostatic Neoplasms/pathology , Androgens/physiology , Antineoplastic Agents, Hormonal/pharmacology , Cell Survival , Humans , Male , Prostate/cytology , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Androgen , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured
15.
J Autoimmun ; 22(3): 249-65, 2004 May.
Article in English | MEDLINE | ID: mdl-15041046

ABSTRACT

In coeliac disease in the jejunum of a genetically susceptible person wheat gliadin and related prolamins from rye and barley trigger an immunological reaction, which induces small-bowel mucosal transformations, villous atrophy and crypt hyperplasia. Though CD4+ specific T cells, intraepithelial lymphocytes, infiltrating plasma cells and autoantibodies are known to have an effect on the coeliac disease, the pathogenic mechanisms leading to the tissue injury remain to be elucidated. Our aim was to find novel gene transcripts, which might have a role in coeliac disease pathogenesis. The gene expression in duodenal biopsy samples from untreated coeliac patients (n=4), patients on gluten-free diet (n=4) and healthy controls (n=4) was studied by cDNA microarray analysis. The method allows monitoring of the expression of thousands of genes simultaneously. Compared to healthy controls, the expression of 156 and 60 genes was changed in untreated and treated coeliac disease, respectively. Between treated and untreated coeliac disease, 98 genes had altered expression. Of the 5184 genes or expressed sequence tags, altogether 263 were affected. Many of these genes are directly or indirectly connected to T-cell activation, B-cell maturation or epithelial cell differentiation. By the microarray method, numerous genes were found to evince altered mRNA expression in coeliac disease. The method holds promise in exploring the pathogenetic mechanisms in the small bowel and may reveal new target genes for the therapy of coeliac disease.


Subject(s)
Celiac Disease/genetics , Gene Expression , Jejunum/pathology , Adult , Aged , Biopsy , Celiac Disease/etiology , Celiac Disease/pathology , Chromosome Mapping , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
16.
Br J Cancer ; 90(5): 1041-6, 2004 Mar 08.
Article in English | MEDLINE | ID: mdl-14997205

ABSTRACT

The long arm of chromosome 8 is one of the most common regions of amplification in cancers of several organs, especially carcinomas of the breast and prostate. TRPS1, MYC and EIF3S3 genes are located in one of the minimal regions of amplification, 8q23-q24, and have been suggested to be the target genes of the amplification. Here, our goal was to study copy number and expression of the three genes in order to investigate the significance of the genes in breast and prostate cancer. By using fluorescence in situ hybridisation (FISH), we first found that TRPS1 and EIF3S3 were amplified together in about one-third of hormone-refractory prostate carcinomas. Next, we analysed the mRNA expression of the three genes by real-time quantitative RT-PCR and the gene copy number by FISH in six breast and five prostate cancer cell lines. Breast cancer cell line, SK-Br-3, which contained the highest copy number of all three genes, showed overexpression of only EIF3S3. Finally, the expression levels of TRPS1, EIF3S3 and MYC were measured in freshly frozen clinical samples of benign prostate hyperplasia (BPH), as well as untreated and hormone-refractory prostate carcinoma. The TRPS1 and MYC expression levels were similar in all prostate tumour groups, whereas EIF3S3 expression was higher (P=0.029) in prostate carcinomas compared to BPH. The data suggest that the expression of EIF3S3 is increased in prostate cancer, and that one of the mechanisms underlying the overexpression is the amplification of the gene.


Subject(s)
Breast Neoplasms/genetics , DNA-Binding Proteins , Eukaryotic Initiation Factor-3/genetics , Gene Dosage , Genes, myc/physiology , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Breast Neoplasms/metabolism , Chromosomes, Human, Pair 8/genetics , DNA Probes/genetics , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Eukaryotic Initiation Factor-3/metabolism , Female , Gene Amplification/genetics , Humans , In Situ Hybridization, Fluorescence , Male , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors , Tumor Cells, Cultured
18.
Am J Pathol ; 159(6): 2089-94, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11733359

ABSTRACT

Gain of the long arm of chromosome 8 (8q) is one of the most common gains found in the advanced prostate cancer by comparative genomic hybridization. We have previously identified a putative target gene for the 8q gain, EIF3S3, that encodes a p40 subunit of eukaryotic translation initiation factor 3 (eIF3). Here, we studied the frequency of the EIF3S3 amplification in different stages of prostate cancer and co-amplification of EIF3S3 and oncogene MYC. In addition, prognostic utility of the EIF3S3 copy number alteration was evaluated. The analyses were done with fluorescence in situ hybridization and tissue microarray technology. High-level amplification of EIF3S3 was found in 11 of 125 (9%) of pT1/pT2 tumors, 12 of 44 (27%) of pT3/pT4 tumors, and 8 of 37 (22%) of lymph node metastases as well as in 26 of 78 (33%) and 15 of 30 (50%) of hormone refractory locally recurrent tumors and metastases, respectively. The amplification was associated with high Gleason score (P < 0.001). One of the 79 tumors with EIF3S3 amplification had only two copies of MYC, whereas all tumors with amplification of MYC had also amplification of EIF3S3 indicating common co-amplification of the genes. Gain of EIF3S3 was associated with poor cancer-specific survival in incidentally found prostate carcinomas (P = 0.023). In the analyses of prostatectomy-treated patients, the amplification was not statistically significantly associated with progression-free time. In conclusion, amplification of EIF3S3 gene is common in late-stage prostate cancer suggesting that it may be functionally involved in the progression of the disease.


Subject(s)
Peptide Initiation Factors/genetics , Prostatic Neoplasms/pathology , Aged , Aged, 80 and over , Eukaryotic Initiation Factor-3 , Gene Amplification , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasm Staging , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Survival Analysis , Survival Rate
19.
Cancer Genet Cytogenet ; 128(1): 31-4, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11454426

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1) is a transcription factor that regulates the expression of genes associated with adaptation to reduced oxygen pressure. Increased expression of HIF-1alpha gene (HIF1A) has been found in the majority of prostate carcinomas. In addition, the PC-3 prostate cancer cell line has been shown to express the gene even under normoxic conditions. By comparative genomic hybridization (CGH), we have earlier shown that the PC-3 cell line contains a high-level amplification in the chromosomal region harboring the HIF1A gene. Here, we first fine mapped the gene to locus 14q23 by fluorescence in situ hybridization (FISH). The gene was then shown to be highly amplified in the PC-3 cell line. Subsequently, the copy number of the HIF1A gene was studied in 5 other prostate cancer cell lines (LNCaP, DU-145, NCI-H660, Tsu-Pr, JCA-1) and in 117 prostate tumors representing both hormone-dependent and -refractory disease as well as primary and metastatic lesions. No high-level amplifications of the HIF1A gene were found. Additional copies of the gene were seen in all of the cell lines and in 36% of the tumors. There was no association between the tumor type and the copy number alterations of the gene. In conclusion, high-level amplification of the HIF1A gene may explain the overexpression of the gene in the PC-3 prostate cancer cell line. However, such high-level amplification seems to be very rare in prostate cancer.


Subject(s)
Chromosome Mapping , Chromosomes, Human, Pair 14/genetics , DNA-Binding Proteins/genetics , Helix-Loop-Helix Motifs/genetics , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Transcription Factors/genetics , DNA-Binding Proteins/metabolism , Gene Amplification , Gene Dosage , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , In Situ Hybridization, Fluorescence , Male , Nuclear Proteins/metabolism , Nucleic Acid Hybridization , Tumor Cells, Cultured
20.
Cancer Res ; 61(14): 5340-4, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454671

ABSTRACT

Prostate cancer is the most common male malignancy in the United States as well as in many European countries. It is curable as long as it is localized, but the invasion of prostate cancer and formation of metastasis turn it into a life-threatening disease. Urokinase-type plasminogen activator (uPA) is believed to play a key role in tissue degradation and cell migration under various normal and pathological conditions, including cancer invasion and metastasis. Increased expression of uPA has been reported in various malignancies including prostate cancer. However, the mechanisms of the overexpression have remained poorly understood. Here, we report increased copy number of uPA gene in 3 of 13 hormone-refractory prostate carcinomas, including 1 high-level amplification. Real-time quantitative reverse transcription-PCR showed that the increased expression of uPA coincided with the amplification of the gene in these tumors. Matrigel invasion assay showed that prostate cancer cell line PC-3, containing amplification of the uPA gene, was more sensitive to the urokinase inhibitor, amiloride, than DU145 or LNCaP cell lines, which do not have the amplification. The findings suggest that one of the mechanisms underlying the overexpression of the uPA is the amplification of the gene, which is associated with the increased invasive potential of the cells.


Subject(s)
Prostatic Neoplasms/pathology , Urokinase-Type Plasminogen Activator/genetics , Blotting, Northern , Gene Amplification , Humans , In Situ Hybridization, Fluorescence , Male , Nucleic Acid Hybridization , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...