Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 133
1.
Oncogene ; 34(5): 568-77, 2015 Jan 29.
Article En | MEDLINE | ID: mdl-24488012

Gene fusions, mainly between TMPRSS2 and ERG, are frequent early genomic rearrangements in prostate cancer (PCa). In order to discover novel genomic fusion events, we applied whole-genome paired-end sequencing to identify structural alterations present in a primary PCa patient (G089) and in a PCa cell line (PC346C). Overall, we identified over 3800 genomic rearrangements in each of the two samples as compared with the reference genome. Correcting these structural variations for polymorphisms using whole-genome sequences of 46 normal samples, the numbers of cancer-related rearrangements were 674 and 387 for G089 and PC346C, respectively. From these, 192 in G089 and 106 in PC346C affected gene structures. Exclusion of small intronic deletions left 33 intergenic breaks in G089 and 14 in PC346C. Out of these, 12 and 9 reassembled genes with the same orientation, capable of generating a feasible fusion transcript. Using PCR we validated all the reliable predicted gene fusions. Two gene fusions were in-frame: MPP5-FAM71D in PC346C and ARHGEF3-C8ORF38 in G089. Downregulation of FAM71D and MPP5-FAM71D transcripts in PC346C cells decreased proliferation; however, no effect was observed in the RWPE-1-immortalized normal prostate epithelial cells. Together, our data showed that gene rearrangements frequently occur in PCa genomes but result in a limited number of fusion transcripts. Most of these fusion transcripts do not encode in-frame fusion proteins. The unique in-frame MPP5-FAM71D fusion product is important for proliferation of PC346C cells.


Cell Proliferation/genetics , Membrane Proteins/genetics , Nucleoside-Phosphate Kinase/genetics , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Male , Membrane Proteins/biosynthesis , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Nucleoside-Phosphate Kinase/biosynthesis , Oncogene Proteins, Fusion/isolation & purification , Prostatic Neoplasms/pathology , Rho Guanine Nucleotide Exchange Factors/biosynthesis , Rho Guanine Nucleotide Exchange Factors/genetics
2.
Sex Dev ; 7(5): 223-34, 2013.
Article En | MEDLINE | ID: mdl-23774508

Androgen receptor (AR) mutations in androgen insensitivity syndrome (AIS) are associated with a variety of clinical phenotypes. The aim of the present study was to compare the molecular properties and potential pathogenic nature of 8 novel and 3 recurrent AR variants with a broad variety of functional assays. Eleven AR variants (p.Cys177Gly, p.Arg609Met, p.Asp691del, p.Leu701Phe, p.Leu723Phe, p.Ser741Tyr, p.Ala766Ser, p.Arg775Leu, p.Phe814Cys, p.Lys913X, p.Ile915Thr) were analyzed for hormone binding, transcriptional activation, cofactor binding, translocation to the nucleus, nuclear dynamics, and structural conformation. Ligand-binding domain variants with low to intermediate transcriptional activation displayed aberrant Kd values for hormone binding and decreased nuclear translocation. Transcriptional activation data, FxxFF-like peptide binding and DNA binding correlated well for all variants, except for p.Arg609Met, p.Leu723Phe and p.Arg775Leu, which displayed a relatively higher peptide binding activity. Variants p.Cys177Gly, p.Asp691del, p.Ala766Ser, p.Phe814Cys, and p.Ile915Thr had intermediate or wild type values in all assays and showed a predominantly nuclear localization in living cells. All transcriptionally inactive variants (p.Arg609Met, p.Leu701Phe, p.Ser741Tyr, p.Arg775Leu, p.Lys913X) were unable to bind to DNA and were associated with complete AIS. Three variants (p.Asp691del, p.Arg775Leu, p.Ile915Thr) still displayed significant functional activities in in vitro assays, although the clinical phenotype was associated with complete AIS. The data show that molecular phenotyping based on 5 different functional assays matched in most (70%) but not all cases.


Androgen-Insensitivity Syndrome/genetics , Receptors, Androgen/genetics , Humans , Male , Mutation
3.
Oncogene ; 31(8): 978-91, 2012 Feb 23.
Article En | MEDLINE | ID: mdl-21765474

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.


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
4.
Br J Cancer ; 102(10): 1491-4, 2010 May 11.
Article En | MEDLINE | ID: mdl-20407443

BACKGROUND: The phosphatidylinositol 3-kinase (PI3K)-AKT pathway is activated in many cancers. Mutational hotspots in AKT1 and in the regulatory and catalytic subunits of PI3K have been detected in multiple tumour types. In AKT1, the E17K substitution leads to a PI3K-independent activation of AKT1. METHODS: A mutational profiling of AKT1 and of the mutational hotspots in PIK3CA and PIK3R1 was carried out in samples from primary and recurrent prostate tumours. RESULTS: We show that, in prostate cancer, AKT1(E17K) had a prevalence of 1.4%. The mutation seemed to be associated with a favourable clinical course but it was not associated with a specific tumour growth pattern. Activating mutations in PIK3CA or PIK3R1 were not found in prostate cancer. CONCLUSION: The E17K substitution in AKT1 is rare in prostate cancer. It seems associated with a favourable clinical outcome but not with a specific histology of the tumour.


Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Aged , DNA Mutational Analysis , Gene Expression , Gene Expression Profiling , Humans , Male , Middle Aged , Polymerase Chain Reaction , Prognosis
5.
Prostate ; 70(2): 135-46, 2010 Feb 01.
Article En | MEDLINE | ID: mdl-19784964

BACKGROUND: The tumor suppressor PTEN regulates many biological processes. A well-known downstream effector of PTEN is phospho-Akt. Although PTEN is the most frequently inactivated gene in prostate cancer, its mode of action is not fully understood. We studied the association of regulated PTEN expression with changes in biological function and gene expression profiles. METHODS: PTEN-negative LNCaP cells were stably transfected with wild-type PTEN cDNA under inducible control, resulting in LNCaP/PTEN cells. Microarray analysis was used to monitor gene expression changes upon induction of PTEN. Expression of selected individual genes was studied in Q-PCR and siRNA experiments. Cell-cycle distribution was analyzed by flow cytometry. RESULTS: Induced expression of PTEN in LNCaP/PTEN cells significantly inhibited cell proliferation, at least partly due to cell-cycle arrest at the G1 phase. Expression profiling combined with pathway analysis revealed that PTEN-dependent G1 growth arrest was associated with an altered mRNA expression of the G1 cell-cycle regulators Cdc25a, E2F2, cyclin G2, and RBL2/p130. Specific inhibition of Akt signaling by siRNA resulted in downregulation of both E2F2 and Cdc25a mRNA expression and upregulation of the FOXO target cyclin G2, similar to the effect observed by PTEN induction. However, Akt did not mediate the PTEN-dependent RBL2/p130 mRNA expression in LNCaP/PTEN cells. CONCLUSIONS: The results indicate that PTEN dependent gene expression is important in cell-cycle regulation and is mediated by both Akt-dependent and -independent mechanisms.


Cell Cycle Proteins/genetics , Cell Cycle/genetics , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Profiling , Humans , Male , Oligonucleotide Array Sequence Analysis , Transfection
6.
Sex Dev ; 3(5): 237-44, 2009.
Article En | MEDLINE | ID: mdl-19851057

Mutations in the androgen receptor (AR) gene, rendering the AR protein partially or completely inactive, cause androgen insensitivity syndrome, which is a form of a 46,XY disorder of sex development (DSD). We present 3 novel AR variants found in a cohort of Indonesian DSD patients: p.I603N, p.P671S, and p.Q738R. The aim of this study was to determine the possible pathogenic nature of these newly found unclassified variants. To investigate the effect of these variants on AR function, we studied their impact on transcription activation, AR ligand-binding domain interaction with an FxxLF motif containing peptide, AR subcellular localization, and AR nuclear dynamics and DNA-binding. AR-I603N had completely lost its transcriptional activity due to disturbed DNA-binding capacity and did not show the 114-kDa hyperphosphorylated AR protein band normally detectable after hormone binding. The patient with AR-I603N displays a partial androgen insensitivity syndrome phenotype, which is explained by somatic mosaicism. A strongly reduced transcriptional activity was observed for AR-Q738R, together with diminished interaction with an FxxLF motif containing peptide. AR-P671S also showed reduced transactivation ability, but no change in DNA- or FxxLF-binding capacity and interferes with transcriptional activity for as yet unclear reasons.


Disorders of Sex Development/genetics , Mutation/genetics , Receptors, Androgen/genetics , Androgen-Insensitivity Syndrome/genetics , Child , Child, Preschool , Humans , Indonesia , Male
7.
J Pathol ; 208(5): 699-707, 2006 Apr.
Article En | MEDLINE | ID: mdl-16402365

PTEN is frequently inactivated during the development of many cancers, including prostate cancer, and both bi-allelic and mono-allelic PTEN inactivation may contribute to tumorigenesis. PTEN mutations in clinical cancer specimens can easily be recorded but mono- or bi-allelic gene deletions are often difficult to assess. We performed a comprehensive study to detect PTEN inactivation in 40 locally progressive clinical prostate cancer specimens obtained by transurethral resection of the prostate, utilizing a variety of complementary technical approaches. The methods to detect PTEN deletion included allelotype analysis, dual-colour FISH and array-based CGH. We also applied a novel semi-quantitative approach, assessing the PTEN-WT (wild-type): PTEN-Psi (pseudogene) ratio (WPR). Structural analysis of PTEN was performed by single-strand conformational polymorphism (PCR-SSCP) and sequencing. PTEN protein expression was assessed by immunohistochemistry. Our data predict complete PTEN inactivation in 12 samples (30%), nine of these by bi-allelic deletion. Loss of one PTEN copy was also detected by several methodologies but the number could not be accurately assessed. Immunohistochemistry indicated the absence of PTEN protein in 15 samples, and heterogeneous expression of the protein in eight tumours. Taken together, these data show that bi-allelic deletion is a major mechanism of PTEN inactivation in locally progressive prostate cancer.


Gene Deletion , Gene Silencing , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Chromosomes, Human, Pair 10/genetics , DNA, Neoplasm/genetics , Disease Progression , Humans , In Situ Hybridization, Fluorescence , Male , Nucleic Acid Hybridization/methods , PTEN Phosphohydrolase/metabolism , Polymerase Chain Reaction/methods , Polymorphism, Single-Stranded Conformational , Prostatic Neoplasms/metabolism
8.
Med Vet Entomol ; 18(4): 313-22, 2004 Dec.
Article En | MEDLINE | ID: mdl-15641996

The role of volatile semiochemicals in mediating the location and selection within herds of Holstein-Friesian heifers by nuisance and disease-transmitting cattle flies was investigated using coupled gas chromatography-electrophysiology (GC-EAG), coupled gas chromatography-mass spectrometry (GC-MS), electrophysiology (EAG), laboratory behaviour and field studies. Using volatile extracts collected by air entrainment from heifers in the Netherlands, a number of active peaks were located by coupled GC-EAG for Musca autumnalis (de Geer) (Diptera: Muscidae) and Haematobia irritans (L.) (Diptera: Muscidae). Volatile samples were also collected from two heifers in Denmark shown in previous counting experiments to differ significantly in their fly loads. Coupled GC-EAG using Ha. irritans antennae revealed differences in the EAG response to the samples, with additional EAG activity in the sample collected from the heifer with the lower fly load. To identify more EAG active compounds, volatiles were also collected from 48-h-old urine by air entrainment. In total, 23 compounds were located and identified by coupled GC-EAG and GC-MS. Further electrophysiological testing of these compounds with five fly species [M. autumnalis, Ha. irritans, Hydrotaea irritans (L.) (Diptera: Muscidae), Stomoxys calcitrans (L.) (Diptera: Musicidae) and Wohlfahrtia magnifica (Schiner) (Diptera: Sarcophagidae)] showed that only some of the compounds were physiologically active across the range of flies tested. These included 1-octen-3-ol, 6-methyl-5-hepten-2-one, (Z)-3-hexen-1-ol, naphthalene, and all EAG active compounds identified from urine. Compounds showing significant EAG activity were tested for behavioural activity using a wind-tunnel designed for measuring upwind flight behaviour. At certain concentrations, 1-octen-3-ol, 6-methyl-5-hepten-2-one and 3-octanol increased upwind flight, whereas naphthalene, propyl butanoate and linalool reduced upwind flight. In field studies using small herds of heifers ranked according to their fly load, individual slow-release formulations of 1-octen-3-ol and 6-methyl-5-hepten-2-one, when applied to low and high fly loading heifers, reduced fly loads on these individuals. This study provides evidence for the hypothesis that the natural differential attractiveness within herds of Holstein-Freisian heifers, i.e. a single host species, for cattle flies is partly due to differences in volatile semiochemicals emitted from the host. It is suggested that this phenomenon applies to other vertebrate host species and their associated insect pests.


Cattle/physiology , Cattle/parasitology , Diptera/physiology , Host-Parasite Interactions , Pheromones/physiology , Animals , Behavior, Animal/physiology , Cattle Diseases/parasitology , Chromatography, Gas , Ectoparasitic Infestations/veterinary , Electrophysiology , Female , Male
10.
Cancer Res ; 60(22): 6479-81, 2000 Nov 15.
Article En | MEDLINE | ID: mdl-11103816

Mutations of the androgen receptor (AR) gene have been reported in prostate cancer, usually from tumor tissue specimens from late-stage, androgen-independent cancer. Occasionally, germ-line mutations have been found, but a link between AR mutations and predisposition to human prostate cancer has not been firmly established. Recently, two independent studies reported the same germ-line mutation at codon 726 in exon E (CGC to CTC) in two apparently unrelated Finnish prostate cancer patients. This arginine to leucine substitution was reported to alter the transactivational specificity of the AR protein. In the present study, the R726L mutation was analyzed by allele-specific oligohybridization in DNA specimens from 418 consecutive prostate cancer patients who reported a negative family history (sporadic group) and from 106 patients with a positive family history (hereditary group). The population frequency of the R726L mutation in blood donors was 3 of 900 (0.33%). In contrast, eight (1.91%) mutations (odds ratio = 5.8; P = 0.006) were found in the sporadic group, and two (1.89%) mutations were found in the hereditary group (odds ratio = 5.8; P = 0.09). Suggestive evidence of the segregation of the mutation with prostate cancer was seen in these two families. The present study indicates that the R726L substitution in the AR may confer an up to 6-fold increased risk of prostate cancer and may contribute to cancer development in up to 2% of Finnish prostate cancer patients. These results warrant additional large-scale studies of the significance of rare mutations and polymorphisms in candidate genes along the androgen signaling pathway as risk factors for prostate cancer.


Germ-Line Mutation , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Binding Sites , Female , Hormones/metabolism , Humans , Male , Protein Structure, Tertiary/genetics , Receptors, Androgen/metabolism , Repetitive Sequences, Nucleic Acid
11.
J Biol Chem ; 275(52): 40846-55, 2000 Dec 29.
Article En | MEDLINE | ID: mdl-11006269

Prostate-specific antigen (PSA) is expressed primarily by both normal prostate epithelium and the vast majority of prostate cancers. Increases in serum PSA during endocrine therapy are generally considered as evidence for prostate cancer recurrence or progression to androgen independence. The mechanisms by which PSA up-regulation occurs in androgen-refractory prostate cancer cells are unknown. In this study, by using LNCaP and its lineage-derived androgen-independent PSA-producing subline, C4-2, we identified two cis-elements within the 5.8-kilobase pair PSA promoter that are essential for the androgen-independent activity of PSA promoter in prostate cancer cells. First, a previously reported 440-bp androgen-responsive element enhancer core (AREc) was found to be important for the high basal PSA promoter activity in C4-2 cells. Both mutation analysis and supershift experiments demonstrated that androgen receptor (AR) binds to the AREs within the AREc and activate the basal PSA promoter activity in C4-2 cells under androgen-deprived conditions. Second, a 150-bp pN/H region was demonstrated to be a strong AR-independent positive-regulatory element of the PSA promoter in both LNCaP and C4-2 cells. Through DNase I footprinting and linker scan mutagenesis, a 17-bp RI site was identified as the key cis-element within the pN/H region. Data from electrophoretic mobility shift analysis and UV cross-linking experiments further indicated that a 45-kDa (p45) cell-specific transcription factor associates with RI in prostate cancer cells and may be responsible for driving the PSA promoter activity independent of androgen and AR. Furthermore, by juxtaposing AREc and pN/H, we produced a chimeric PSA promoter (supra-PSA) that exhibits 2-3-fold higher activity than the wild type PSA promoter in both LNCaP and C4-2 cells.


Androgens/physiology , Promoter Regions, Genetic , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/metabolism , Base Sequence , Gene Expression Regulation , Humans , Male , Molecular Sequence Data , Receptors, Androgen/physiology , Response Elements , Tumor Cells, Cultured
14.
Clin Cancer Res ; 5(11): 3578-82, 1999 Nov.
Article En | MEDLINE | ID: mdl-10589774

Finasteride is commonly used for the treatment of benign prostatic hyperplasia and has been suggested to prevent prostate cancer development. To gain insight to the molecular effects of finasteride on prostate cancer development, we studied six prostate cancers diagnosed during finasteride treatment for benign prostatic hyperplasia. Comparative genomic hybridization detected genetic alterations in four tumors (1-5 changes/tumor). Xq gains and 6q losses were the most common alterations. The recurrent Xq gains motivated us to study the involvement of the androgen receptor (AR) gene. One tumor with Xq gain had a 3-fold amplification of the AR gene, suggesting that tumor development in finasteride-treated patients may require increased AR copy number and expression, as has previously been shown for prostate cancers recurring during hormonal therapy. Furthermore, in another tumor, an Arg726Leu mutation of the AR gene was found. This mutation was also present in the germ-line DNA of the patient. Arg726Leu mutation has previously been reported to affect the transactivational properties of the AR. In summary, prostate cancers developing during finasteride therapy may have distinct biological properties, such as a low number of chromosomal alterations and frequent involvement of the AR gene. Further studies are needed to explore the role of germ-line AR mutations in these patients.


Chromosome Aberrations , Finasteride/therapeutic use , Prostatic Hyperplasia/drug therapy , Prostatic Hyperplasia/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Chromosome Mapping , Chromosomes, Human, Pair 6 , Enzyme Inhibitors/therapeutic use , Humans , In Situ Hybridization, Fluorescence , Male , Neoplasm Staging , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , X Chromosome
15.
Gene ; 240(2): 261-7, 1999 Nov 29.
Article En | MEDLINE | ID: mdl-10580145

Human prostate-specific transglutaminase (hTG(P)) is a cross-linking enzyme encoded by the TGM4 gene. The TGM4 gene promoter was characterized by deletion mapping and mutational analysis. Promoter constructs, containing the minimal promoter requirements, could efficiently drive transcription in the prostate cancer cell lines PC346C and LNCaP and the hepatic cancer cell line Hep3B. The region between positions -113 and -61 was demonstrated to be essential for core promoter activity. Further analysis revealed the functional importance of an Sp1 binding motif, 5'-ACCCCGCCCC-3', at positions -96 to -87. This sequence is a binding site of the ubiquitous transcription factors Sp1 and Sp3.


Promoter Regions, Genetic/genetics , Prostate/enzymology , Sp1 Transcription Factor/metabolism , Transglutaminases/genetics , Base Sequence , Binding Sites/genetics , Binding, Competitive , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , Luciferases/genetics , Luciferases/metabolism , Male , Molecular Sequence Data , Mutation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Sp3 Transcription Factor , Transcription Factors/metabolism , Transglutaminases/chemistry , Transglutaminases/metabolism , Tumor Cells, Cultured
16.
J Steroid Biochem Mol Biol ; 69(1-6): 307-13, 1999.
Article En | MEDLINE | ID: mdl-10419007

Androgens play a crucial role in several stages of male development and in the maintenance of the male phenotype. Androgens act in their target cells via an interaction with the androgen receptor, resulting in direct regulation of gene expression. The androgen receptor is a phosphoprotein and modulation of the phosphorylation status of the receptor influences ligand-binding and consequently transcription activation of androgen responsive genes. Androgen binding induces a conformational change in the ligand-binding domain, accompanied by additional receptor phosphorylation. Subsequently the liganded androgen receptor interacts with specific androgen response elements in the regulatory regions of androgen target genes, resulting in stimulation of gene expression. Anti-androgens induce a different conformational change of the ligand-binding domain, which does not or only partially result in stimulation of transactivation. Interestingly, different anti-androgens can induce different inactive conformations of the androgen receptor ligand-binding domain. Recent evidence strongly supports a ligand dependent functional interaction between the ligand-binding domain and the NH2-terminal transactivating domain of the androgen receptor. Two regions in the NH2-terminal domain are involved in this interaction, whereas in the ligand-binding domain the AF-2 AD core region is involved.


Receptors, Androgen/metabolism , Androgens/metabolism , Humans , Ligands , Phosphorylation , Protein Conformation , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Transcriptional Activation
17.
Int J Cancer ; 80(3): 439-43, 1999 Jan 29.
Article En | MEDLINE | ID: mdl-9935187

Down-regulation of the cell-surface adhesion molecule CD44 has been suggested to play an important role in tumor progression and metastasis of prostate cancer. CD44 is encoded by a gene that contains a CpG-rich region (CpG island) in its 5' regulatory sequence. We tried to assess whether hypermethylation of this region is the mechanism responsible for CD44 transcriptional inactivation. A panel of prostatic-carcinoma cell lines, Du145, LNCaP, PC3, PC346C and TSU, was analyzed for CD44 mRNA and protein expression. Du145, PC3 and TSU were positive for CD44, whereas in LNCaP and PC346C both CD44 mRNA and protein expression was suppressed. Methylation-sensitive restriction-enzyme analysis of genomic DNA showed that, in contrast to the CD44-positive cell lines, the CD44-negative lines were hypermethylated in the CD44 promoter CpG island. Furthermore, treatment of a PC346C culture with the demethylating agent 5-azacytidine resulted in re-expression of CD44 mRNA. It is concluded that hypermethylation of the CD44 5' promoter region is one of the mechanisms by which CD44 expression is down-regulated in prostatic-carcinoma cell lines.


CpG Islands/physiology , DNA Methylation , DNA, Neoplasm/metabolism , Hyaluronan Receptors/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Down-Regulation , Humans , Hyaluronan Receptors/genetics , Male , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured
18.
Genes Chromosomes Cancer ; 24(2): 119-26, 1999 Feb.
Article En | MEDLINE | ID: mdl-9885978

One of the most frequent genetic abnormalities in prostate cancer is loss of the complete or part of the short arm of chromosome 8, indicating the localization of one or more tumor suppressor genes on this chromosomal arm. Using allelotyping, a frequently deleted region in prostate cancer in a genetic interval of approximately 17 cM between sequence tagged sites D8S87 and D8S133 at chromosome arm 8p12-21 was previously detected. A detailed physical map of this region is now available. Using known and novel polymorphic and nonpolymorphic sequence tagged sites in this interval, a search for homozygous deletions in DNAs from 14 prostate cancer-derived cell lines and xenografts was carried out. In DNA from xenograft PC133, the presence of a small homozygously deleted region of 730-1,320 kb was unambiguously established. At one site, the deletion disrupts the Werner syndrome gene. Data from allelotyping were confirmed and extended by fluorescence in situ hybridization analysis of PC133 chromosome spreads using centromere, YAC, and PAC chromosome 8 probes.


Chromosome Deletion , Chromosomes, Human, Pair 8/genetics , Homozygote , Prostatic Neoplasms/metabolism , Transplantation, Heterologous/methods , Animals , Chromosome Mapping/methods , DNA, Neoplasm/analysis , Genetic Markers/genetics , Humans , In Situ Hybridization, Fluorescence/methods , Male , Mice , Mice, Nude , Physical Chromosome Mapping/methods , Tumor Cells, Cultured/transplantation
19.
Genomics ; 51(3): 434-44, 1998 Aug 01.
Article En | MEDLINE | ID: mdl-9721214

Human prostate-specific transglutaminase (hTGP) is a cross-linking enzyme secreted by the prostate. In this study, we performed dot blot analysis of 50 normal human tissues to demonstrate unambiguously the prostate-specific expression of hTGP. Furthermore, we elucidated the genomic organization of the TGM4 gene, the gene encoding hTGP. The structure of this gene displays striking similarity to that of other transglutaminase (TGase) genes. The TGM4 gene spans approximately 35 kb of genomic DNA and consists of 13 exons and 12 introns. The main transcription initiation site is located 52 bp upstream of the translational start codon. A hTGP splice variant of intron 1 was detected. This splice variant contains an in-frame antisense Alu element insertion. The TGM4 promoter was analyzed by sequencing and transfection experiments. At positions -1276 to -563, the promoter harbors a cyclophilin pseudogene with 94% similarity to the cyclophilin A cDNA. Deletion mapping of the TGM4 promoter in the transiently transfected human prostate cancer cell line PC346C showed comparable activity of 2.1-, 1.5-, and 0.5-kb promoter fragments.


Gene Expression Regulation, Enzymologic/genetics , Promoter Regions, Genetic/genetics , Prostate/enzymology , Transglutaminases/genetics , Amino Acid Sequence , Base Sequence , Exons/genetics , Genes, Reporter/genetics , Humans , Introns/genetics , Male , Molecular Sequence Data , Peptidylprolyl Isomerase/chemistry , Protein Biosynthesis/genetics , Pseudogenes/genetics , RNA Splicing/genetics , RNA, Messenger/metabolism , Sequence Analysis, DNA , Transcription, Genetic/genetics , Transfection/genetics , Transglutaminases/chemistry , Tumor Cells, Cultured
20.
Mol Endocrinol ; 12(8): 1172-83, 1998 Aug.
Article En | MEDLINE | ID: mdl-9717843

Previous studies in yeast and mammalian cells showed a functional interaction between the amino-terminal domain and the carboxy-terminal, ligand-binding domain (LBD) of the human androgen receptor (AR). In the present study, the AR subdomains involved in this in vivo interaction were determined in more detail. Cotransfection experiments in Chinese hamster ovary (CHO) cells and two-hybrid experiments in yeast revealed that two regions in the NH2-terminal domain are involved in the functional interaction with the LBD: an interacting domain at the very NH2 terminus, located between amino acid residues 3 and 36, and a second domain, essential for transactivation, located between residues 370 and 494. Substitution of glutamic acid by glutamine at position 888 (E888Q) in the AF-2 activation domain (AD) core region in the LBD, markedly decreased the interaction with the NH2-terminal domain. This mutation neither influenced hormone binding nor LBD homodimerization, suggesting a role of the AF-2 AD core region in the functional interaction between the NH2-terminal domain and the LBD. The AF-2 AD core region was also involved in the interaction with the coactivator TIF2 (transcriptional intermediary factor 2), as the E888Q mutation decreased the stimulatory effect of TIF2 on AR AF-2 activity. Cotransfection of TIF2 and the AR NH2-terminal domain expression vectors did not result in synergy between both factors in the induction of AR AF-2 activity. TIF2 highly induced AR AF-2 activity on a complex promoter [mouse mammary tumor virus (MMTV)], but it was hardly active on a minimal promoter (GRE-TATA). In contrast, the AR NH2-terminal domain induced AR AF-2 activity on both promoter constructs. These data indicate that both the AR NH2-terminal domain and the coactivator TIF2 functionally interact, either directly or indirectly, with the AF-2 AD core region in the AR-LBD, but the level of transcriptional response induced by TIF2 depends on the promoter context.


Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , CHO Cells , Cricetinae , Dimerization , Humans , Luciferases/genetics , Luciferases/metabolism , Mutation , Nuclear Receptor Coactivator 2 , Promoter Regions, Genetic , Receptors, Androgen/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
...