Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Oncogene ; 31(16): 2090-100, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-21892209

ABSTRACT

Tumor-suppressor genes (TSGs) have been classically defined as genes whose loss of function in tumor cells contributes to the formation and/or maintenance of the tumor phenotype. TSGs containing nonsense mutations may not be expressed because of nonsense-mediated RNA decay (NMD). We combined inhibition of the NMD process, which clears transcripts that contain nonsense mutations, with the application of high-density single-nucleotide polymorphism arrays analysis to discriminate allelic content in order to identify candidate TSGs in five breast cancer cell lines. We identified ARID1A as a target of NMD in the T47D breast cancer cell line, likely as a consequence of a mutation in exon-9, which introduces a premature stop codon at position Q944. ARID1A encodes a human homolog of yeast SWI1, which is an integral member of the hSWI/SNF complex, an ATP-dependent, chromatin-remodeling, multiple-subunit enzyme. Although we did not find any somatic mutations in 11 breast tumors, which show DNA copy-number loss at the 1p36 locus adjacent to ARID1A, we show that low ARID1A RNA or nuclear protein expression is associated with more aggressive breast cancer phenotypes, such as high tumor grade, in two independent cohorts of over 200 human breast cancer cases each. We also found that low ARID1A nuclear expression becomes more prevalent during the later stages of breast tumor progression. Finally, we found that ARID1A re-expression in the T47D cell line results in significant inhibition of colony formation in soft agar. These results suggest that ARID1A may be a candidate TSG in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Genes, Tumor Suppressor , Nuclear Proteins/genetics , Transcription Factors/genetics , Cell Line, Tumor , Chromosomes, Human, Pair 1 , Codon, Nonsense , DNA Copy Number Variations , DNA-Binding Proteins , Female , Humans , RNA/metabolism , Transfection
2.
Oncogenesis ; 1: e27, 2012 Sep 10.
Article in English | MEDLINE | ID: mdl-23552840

ABSTRACT

The frequent loss of heterozygosity of chromosome (Chr) 17 in epithelial ovarian cancer (EOC), particularly high-grade ovarian serous carcinomas (HGOSCs), has been attributed to the disruption of known tumour suppressor genes, such as TP53 (17p13), as well as other genes on this chromosome that alone or in combination have a role in EOC. In a transcriptome analysis of Chr17 genes, we observed significant underexpression of the chemokine CCL2 (17q12) in a small set of HGOSC samples relative to normal ovarian surface epithelial cells and a significant upregulation of CCL2 in the TP53-mutated OV-90 EOC cell line rendered non-tumourigenic as a consequence of genetic manipulation. Here, we report that overexpressing CCL2 in OV-90 resulted in latency of tumour formation at intraperitoneal (i.p.) but not subcutaneous sites in a mouse xenograft model. Overexpressing CCL2 affected cell morphology and exerted modest, but not significant effects on cell viability, colony formation and cell migration. We report significant underexpression of CCL2 by transcriptome analysis (P=0.015) and by immunohistochemistry in 77% of HGOSC samples (n=65). Absent or a very low level of protein expression by immunohistochemistry was also observed in 71% of additional HGOSC samples (n=122). However, CCL2 protein expression did not significantly correlate with overall or disease-free survival. The epithelial cells of normal fallopian tubes, a purported origin of HGOSC, exhibited expression of CCL2 protein by immunohistochemistry. Our results affirm that CCL2 underexpression is a significant feature of HGOSC samples, and that CCL2 overexpression in an EOC cell line model affects tumourigenic potential in the i.p. setting.

3.
Int J Gynecol Cancer ; 18(5): 963-75, 2008.
Article in English | MEDLINE | ID: mdl-18028382

ABSTRACT

Cytogenetic, molecular genetic, and functional analyses have implicated chromosome 17 genes in epithelial ovarian cancer (EOC). To further characterize the contribution of chromosome 17 genes in EOC, the Affymetrix U133A GeneChip was used to perform transcriptome analyses of 15 primary cultures of normal ovarian surface epithelial (NOSE) cells and 17 malignant ovarian tumor (TOV) samples of the serous histopathologic subtype. A two-way comparative analysis of 776 known genes and expressed sequences identified 253 genes that exhibited at least a threefold difference in expression in at least one TOV sample compared to the mean of NOSE samples. Within this data set, 99 of the 253 (39.1%) genes exhibited similar patterns of expression across all tested samples, suggesting a high degree of concordance in the chromosome 17 transcriptome. This observation was supported by hierarchical clustering analysis that segregated the TOV and NOSE samples into two separate groups. There were 77 genes that were differentially expressed in at least 50% of the TOV samples. Five genes (AdoRA(2B)at 17p12, CCL2 at 17q12, ACLY at 17q21.2, WIPI1 at 17q24.2, and SLC16A3 at 17q25.3) were significantly (P < 5.13E-11) differentially expressed at least threefold in all serous TOV samples, and all five genes were underexpressed in these TOV samples as compared to the NOSE samples. Interestingly, several of these differentially expressed genes have been previously associated with response to hypoxia.


Subject(s)
Chromosomes, Human, Pair 17/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Adult , Aged , Female , Humans , Middle Aged , RNA, Messenger/genetics , Tumor Cells, Cultured
4.
Oncogene ; 26(4): 618-32, 2007 Jan 25.
Article in English | MEDLINE | ID: mdl-16909122

ABSTRACT

Multiple chromosome 3p tumor suppressor genes (TSG) have been proposed in the pathogenesis of ovarian cancer based on complex patterns of 3p loss. To attain functional evidence in support of TSGs and identify candidate regions, we applied a chromosome transfer method involving cell fusions of the tumorigenic OV90 human ovarian cancer cell line, monoallelic for 3p and an irradiated mouse cell line containing a human chromosome 3 in order to derive OV90 hybrids containing normal 3p fragments. The resulting hybrids showed complete or incomplete suppression of tumorigenicity in nude mouse xenograft assays, and varied in their ability to form colonies in soft agarose and three-dimensional spheroids in a manner consistent with alteration of their in vivo tumorigenic phenotypes. Expression microarray analysis identified a set of common differentially expressed genes, such as SPARC, DAB2 and VEGF, some of which have been shown implicated in ovarian cancer. Genotyping assays revealed that they harbored normal 3p fragments, some of which overlapped candidate TSG regions (3p25-p26, 3p24 and 3p14-pcen) identified previously in loss of heterozygosity analyses of ovarian cancers. However, only the 3p12-pcen region was acquired in common by all hybrids where expression microarray analysis identified differentially expressed genes. The correlation of 3p12-pcen transfer and tumor suppression with a concerted re-programming of the cellular transcriptome suggest that the putative TSG may have affected key underlying events in ovarian cancer.


Subject(s)
Adenocarcinoma/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 3 , Genes, Tumor Suppressor/physiology , Ovarian Neoplasms/genetics , Animals , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Transfer Techniques , Humans , Mice , Mice, Nude , Xenograft Model Antitumor Assays
5.
Clin Genet ; 70(4): 320-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16965326

ABSTRACT

The BRCAPRO, Couch, Myriad I and II, Ontario Family History Assessment Tool (FHAT), and Manchester models have been used to predict BRCA1 or BRCA2 mutation carrier status of women at high risk for developing the heritable form of breast and ovarian cancers. We have evaluated these models for their accuracy in classifying 224 French Canadian families with at least three cases of breast cancer (diagnosed before the age of 65 years), ovarian cancer, or male breast cancer where mutation status was known for an index affected case used to assess the model. This series includes 44 BRCA1 and 52 BRCA2 mutation-positive families. Using receiver operator characteristics analyses, the C-statistics were found to be 0.81, 0.80, 0.79, and 0.74 for the BRCAPRO, FHAT, Manchester, and Myriad II models, respectively, when incorporating both BRCA1 and BRCA2 mutation carrier predictions. For the BRCAPRO model, 75% scored greater than a 0.43 probability in the mutation-positive group and 75% scored less than 0.50 in the mutation-negative group. Only 38 of 128 (30%) mutation-negative group had a probability greater than 0.43 with the BRCAPRO model. While all models were highly predictive of carrier status, the BRCAPRO model was the most accurate where a cut-off of 10% would have eliminated 60 of 128 (47%) mutation-negative families for genetic testing and only miss 10 of 96 (10%) mutation-positive families. A review of the cancer phenotypes with high BRCAPRO probabilities showed that significantly more metachronous bilateral breast cancer cases occurred in BRCA1/2 mutation carrier families in comparison to mutation-negative families, a feature which is not discriminated in the BRCAPRO model.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Models, Genetic , Ovarian Neoplasms/genetics , Breast Neoplasms/ethnology , Breast Neoplasms, Male/ethnology , Breast Neoplasms, Male/genetics , Canada , Female , Genetic Predisposition to Disease/ethnology , Germ-Line Mutation , Humans , Male , Middle Aged , Ovarian Neoplasms/ethnology , Pedigree , Probability , Quebec , ROC Curve
6.
Br J Cancer ; 94(3): 436-45, 2006 Feb 13.
Article in English | MEDLINE | ID: mdl-16421595

ABSTRACT

In order to elucidate the biological variance between normal ovarian surface epithelial (NOSE) and epithelial ovarian cancer (EOC) cells, and to build a molecular classifier to discover new markers distinguishing these cells, we analysed gene expression patterns of 65 primary cultures of these tissues by oligonucleotide microarray. Unsupervised clustering highlights three subgroups of tumours: low malignant potential tumours, invasive solid tumours and tumour cells derived from ascites. We selected 18 genes with expression profiles that enable the distinction of NOSE from these three groups of EOC with 92% accuracy. Validation using an independent published data set derived from tissues or primary cultures confirmed a high accuracy (87-96%). The distinctive expression pattern of a subset of genes was validated by quantitative reverse transcription-PCR. An ovarian-specific tissue array representing tissues from NOSE and EOC samples of various subtypes and grades was used to further assess the protein expression patterns of two differentially expressed genes (Msln and BMP-2) by immunohistochemistry. This study highlights the relevance of using primary cultures of epithelial ovarian cells as a model system for gene profiling studies and demonstrates that the statistical analysis of gene expression profiling is a useful approach for selecting novel molecular tumour markers.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Ovarian Neoplasms/classification , Ovarian Neoplasms/diagnosis , Biomarkers, Tumor/metabolism , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/analysis , Bone Morphogenetic Proteins/genetics , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/pathology , Female , GPI-Linked Proteins , Genes, Neoplasm , Humans , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Mesothelin , Neoplasm Staging , Ovarian Neoplasms/genetics , Ovary/chemistry , Ovary/cytology , Ovary/pathology , Receptors, Tumor Necrosis Factor, Type I/analysis , Receptors, Tumor Necrosis Factor, Type I/genetics , Sodium-Potassium-Exchanging ATPase/analysis , Sodium-Potassium-Exchanging ATPase/genetics , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured
7.
Cancer Chemother Pharmacol ; 54(6): 497-504, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15258697

ABSTRACT

To address the cellular basis for the response to ovarian cancer treatment, we characterized the chemosensitivity and radiosensitivity of four human epithelial ovarian cancer cell lines that harbor different genetic alterations. The TOV-21G, TOV-81D, OV-90, and TOV-112D cell lines were derived from ovarian tumors (TOV) or ascites (OV) from chemotherapy- and radiotherapy-naive patients and were characterized by their mutation spectrum of BRCA2, TGFbeta-RII, KRAS2, TP53, and CDKN2A. Cells were monitored for survival following exposure at various concentrations to different cytotoxic agents including cisplatin, camptothecin or paclitaxel or to different doses of gamma-irradiation. At the lowest doses, the TGFbeta-RII-mutated and KRAS2-mutated cell line, TOV-21G, and the BRCA2-mutated cell line, TOV-81D, demonstrated a significantly higher sensitivity to cisplatin and gamma-irradiation than the TP53-mutated cell lines, TOV-112D and OV-90. At higher doses, differences between the TP53-mutated lines were observed with TOV-112D being less sensitive to cisplatin than OV-90 that also harbors a CDNK2A mutation. All cell lines were similarly sensitive to high doses of gamma-irradiation. In contrast, sensitivity to camptothecin or paclitaxel was not significantly different between all cell lines, irrespective of the mutation status of BRCA1, BRCA2, TGFbeta-RII, KRAS2, TP53, and CDKN2A. The observed responses to treatment are consistent with the current knowledge concerning BRCA2, TGFbeta-RII, KRAS2, TP53, and/or CDKN2A aberrant function.


Subject(s)
Antineoplastic Agents/pharmacology , Ovarian Neoplasms/genetics , Cell Survival/drug effects , Cell Survival/radiation effects , Female , Genes, BRCA2 , Genes, p16 , Genes, p53 , Humans , Mutation , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Radiation Tolerance , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Tumor Cells, Cultured , ras Proteins
9.
Phys Med Biol ; 47(10): 1623-32, 2002 May 21.
Article in English | MEDLINE | ID: mdl-12069082

ABSTRACT

It has been suggested that the small angle x-ray scattering (SAXS) pattern of human hair can be used to diagnose breast cancer and possibly to identify BRCA1/2 mutation carriers, who are at significantly elevated risk for developing breast cancer. In particular, the presence of a diffuse ring in the SAXS pattern was said to be diagnostic of either breast cancer or an increased risk thereof. To test this hypothesis, we measured SAXS from the pubic hair of 56 subjects with known BRCA1/2 and breast cancer status. We found that there is no clear association between the pattern of SAXS seen in human pubic hair and the risk of breast cancer or the presence of BRCA1/2 mutations. The possible use of SAXS to diagnose cancer remains conjectural, but this and previous studies do not suggest that SAXS can be used as a reliable method of identifying either BRCA1/2 mutation carriers or women who have had breast cancer.


Subject(s)
BRCA2 Protein/genetics , Breast Neoplasms/genetics , Genes, BRCA1 , Hair/chemistry , Mutation , X-Ray Diffraction/methods , Breast Neoplasms/diagnosis , Female , Humans , Scattering, Radiation , X-Rays
10.
Oncogene ; 20(45): 6617-26, 2001 Oct 04.
Article in English | MEDLINE | ID: mdl-11641787

ABSTRACT

We have previously described an ovarian cancer model based on four independent spontaneously immortalized epithelial ovarian cancer cell lines (TOV-21G, TOV-81D, TOV-112D and OV-90) from patients who were never exposed to chemotherapy or radiation therapy. These cell lines are particularly interesting since they retain characteristics of the original epithelial ovarian cancers (EOC) from which they were derived. Here we report the characterization of this model system using high-density DNA microarrays in order to assess gene expression. Expression profiles were generated from total RNAs extracted from the four EOC cell lines. For comparison, expression profiling is also provided for a primary culture of normal ovarian surface epithelium (NOV-31) and a fresh EOC sample (TOV-578G). Comparison of expression profiles revealed patterns of expression that distinguish NOV-31 from that of all tumor derived samples. The expression pattern of TOV-81D, an EOC cell line that was derived from a patient with indolent disease, most closely resembles NOV-31 while profiles of samples derived from patients with more aggressive disease (TOV-21G, OV-90, TOV-112D and TOV-578G) showed more divergent patterns of expression. The microarray analysis (http://genome.mcgill.ca) results confirm the usefulness of an ovarian cancer model based on the characterization of these EOC cell lines.


Subject(s)
Carcinoma/genetics , Ovarian Neoplasms/genetics , Blotting, Northern , Carcinoma/metabolism , Chromosomes , Female , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/metabolism , RNA, Neoplasm/biosynthesis , Reproducibility of Results , Tumor Cells, Cultured
11.
Hum Hered ; 52(2): 116-20, 2001.
Article in English | MEDLINE | ID: mdl-11512557

ABSTRACT

The BRCA2 8765delAG mutation was previously reported in hereditary breast cancer families of French Canadian and Yemenite Jewish descent. Haplotype analysis, using six microsatellite markers that span BRCA2 and two intragenic polymorphisms, was performed on 8765delAG mutation carriers to determine if there was evidence that the mutations were identical by descent. The alleles of the microsatellite markers most closely flanking BRCA2 (D13S1697 and D13S1701) were found to be identical in state in all the mutation carriers. However, the disease-associated allele of one of the intragenic markers differed between the Yemenite Jews and French Canadian families, indicating that the 8765delAG mutation has independent origins in these two geographically and ethnically distinct populations.


Subject(s)
Breast Neoplasms/genetics , Haplotypes , Jews/genetics , Mutation , Neoplasm Proteins/genetics , Transcription Factors/genetics , BRCA2 Protein , Canada , Female , France/ethnology , Gene Deletion , Genetic Carrier Screening , Humans
12.
Hum Hered ; 52(1): 55-8, 2001.
Article in English | MEDLINE | ID: mdl-11359068

ABSTRACT

Mauritius, a small island some 855 km off the east coast of Madagascar, has a multiethnic population of about 1.2 million with a high population density of about 611 per km(2). The recent industrialization of the island seems to have been accompanied, in less than 10 years, by an increase of at least 30% in breast cancer incidence. We have detected the BRCA2 6503delTT mutation in two sisters of the same family of Indian origin but living in Mauritius for at least five generations. This mutation has been found to recur in geographically diverse populations and haplotype analysis has shown a common ancestry. The haplotype of the mutation found in the Mauritian family differs from that found in other populations harbouring the same mutation, suggesting that the BRCA2 6503delTT mutation most likely arose independently.


Subject(s)
Breast Neoplasms/genetics , Chromosomes, Human, Pair 13 , Germ-Line Mutation , Neoplasm Proteins/genetics , Transcription Factors/genetics , BRCA2 Protein , Female , Gene Deletion , Haplotypes , Humans , Male , Mauritius , Middle Aged , Pedigree
13.
Int J Cancer ; 95(3): 189-93, 2001 May 20.
Article in English | MEDLINE | ID: mdl-11307153

ABSTRACT

Recently, founder BRCA1 and BRCA2 mutations were identified in Canadian breast cancer and breast-ovarian cancer families of French ancestry. The presence of a breast cancer case diagnosed at younger than 36 years of age was strongly predictive of the presence of any founder mutation screened. Here we report the occurrence of founder BRCA1 and BRCA2 mutations in a series of 61 French Canadian women with invasive breast cancer diagnosed at age 40 or younger, unselected for family history of breast and ovarian cancer. Germline mutations in BRCA1 (n = 4) and BRCA2 (n = 4) were identified in 8 of 61 (13%) cases. All BRCA1 mutations were found in invasive ductal carcinomas, the most common histologic type of tumor in this series. In contrast, the BRCA2 mutations were found in tumors of various histologic types: two ductal carcinomas, a tumor containing both ductal and lobular histologic types and an invasive lobular carcinoma. Of the 37 women with at least one first-, second- or third-degree relative with breast or ovarian cancer and the 24 women with no history of these cancers, 7 (19%) and 1 (4%), respectively, were mutation carriers. The seven mutation carriers with a family history of cancer had at least one first-, second- or third-degree relative with a breast cancer diagnosis at less than 51 years of age. The identification of founder BRCA1 and BRCA2 mutations in young-onset breast cancer cases unselected for family history can facilitate carrier detection when the expected yield of a comprehensive screen may be low.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Founder Effect , Neoplasm Proteins/genetics , Transcription Factors/genetics , Adult , BRCA2 Protein , Breast Neoplasms/ethnology , Canada/epidemiology , Canada/ethnology , DNA Mutational Analysis , Female , Gene Frequency , Humans , Mutation
16.
Semin Surg Oncol ; 18(4): 281-6, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10805949

ABSTRACT

The medical histories of breast cancer-prone families have been described for over a century. The pattern of breast cancer occurrences in these families is most consistent with an autosomal dominant mode of inheritance. The location of a gene that could explain the pattern of transmission of the breast cancer trait in families averaging early (pre-menopausal) onset of breast cancer was reported in 1990. Since then, two genes have been identified: BRCA1 and BRCA2. Germ-line mutations in these two genes confer susceptibility to breast (female and male) and ovarian cancer, and account for a significant proportion of hereditary breast cancer in two cancer syndromes: site-specific breast cancer and the breast-ovarian cancer syndrome. Other hereditary syndromes that feature breast cancer are Li-Fraumeni syndrome, Cowden disease, and ataxia telangiectasia, whose carriers have been shown to harbor germ-line mutations in TP53, PTEN, and ATM, respectively. There may be other genetic factors that contribute to hereditary breast cancer, since not all families with multiple cases of breast cancer harbor germ-line BRCA1 or BRCA2 mutations. Host factors (such as lifestyle choices) and other genes may modulate risk of breast cancer in mutation carriers.


Subject(s)
Breast Neoplasms, Male/genetics , Breast Neoplasms/genetics , Genes, BRCA1/genetics , Genetic Predisposition to Disease , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Transcription Factors/genetics , BRCA2 Protein , Breast Neoplasms/etiology , Breast Neoplasms, Male/etiology , Female , Germ-Line Mutation , Humans , Life Style , Male , Ovarian Neoplasms/etiology , Syndrome
18.
Cancer Genet Cytogenet ; 118(1): 65-8, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10731594

ABSTRACT

Simple numerical chromosome aberrations have been observed in tumorigenesis and may point to indicative initiating or early events in tumorigenesis. We have identified two cases of ovarian carcinomas with trisomy of chromosome 10 using conventional GTG-banding and fluorescence in situ hybridization. This is, to our knowledge, the first report of trisomy 10 as a simple karyotypic abnormality observed in ovarian carcinoma. These results suggest that further studies investigating whether chromosome 10 genes are associated with the pathogenesis of some ovarian tumors are warranted.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Mucinous/genetics , Chromosomes, Human, Pair 10/genetics , Ovarian Neoplasms/genetics , Trisomy/genetics , Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Mucinous/pathology , Adult , Chromosome Banding , Chromosome Painting , Female , Humans , Karyotyping , Middle Aged , Ovarian Neoplasms/pathology , Tumor Cells, Cultured
19.
Oncogene ; 19(11): 1466-72, 2000 Mar 09.
Article in English | MEDLINE | ID: mdl-10723138

ABSTRACT

Allelic deletions of multiple chromosome 17q loci in sporadic ovarian cancer of epithelial origin suggest that inactivation of tumor suppressor gene(s) in these regions may be important for ovarian tumorigenesis. To further define the pattern of allelic imbalance in epithelial ovarian tumors of different histologies, a PCR-based assay was used to assess loss of heterozygosity (LOH) of polymorphic markers representative of TP53, BRCA1, NME1 and GH1, and region 17q23-25. LOH was observed for at least one marker in 68% of malignant tumors (n=60) and in 18% tumors of borderline malignancy (n=11), but not in benign tumors (n=5). The highest frequency of LOH in malignant tumors (64%) was observed with D17S801 on 17q25. Ten of 39 malignant ovarian tumors displaying LOH of at least one 17q marker, displayed a LOH pattern enabling the determination of a minimal region of overlapping deletion defined by D17S795 and D17S801. One borderline tumor also displayed an interstitial LOH pattern that overlapped this 17q25 minimal region of deletion. The histologies of malignant tumors displaying a pattern indicative of interstitial 17q deletions were of the endometrioid, clear cell and mucinous epithelial types. As the minimal region of overlap defined by these tumors overlap regions deleted in malignant tumors of all histologic types, and in a tumor of borderline malignancy, the 17q25-tumor suppressor may be implicated in the development of all types of epithelial ovarian tumors.


Subject(s)
Alleles , Carcinoma/genetics , Chromosomes, Human, Pair 17/genetics , Ovarian Neoplasms/genetics , Carcinoma/classification , Carcinoma/pathology , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/pathology , Cell Differentiation/genetics , Cystadenocarcinoma, Mucinous/genetics , Cystadenocarcinoma, Mucinous/pathology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Female , Genetic Markers , Humans , Loss of Heterozygosity , Neoplasm Staging , Ovarian Neoplasms/classification , Ovarian Neoplasms/pathology , Tumor Cells, Cultured
20.
Prostate Cancer Prostatic Dis ; 3(4): 241-247, 2000 Dec.
Article in English | MEDLINE | ID: mdl-12497071

ABSTRACT

There is evidence for genetic predisposition to prostate cancer. However, prostate cancer genes have been more difficult to find than genes for some of the other common cancers, such as breast and colon cancer. The reasons for this are discussed in this article and it is now becoming clear that prostate cancer is probably due to multiple genes, many of which are moderate or low penetrance. The advances in the Human Genome Project and technology, especially that of robotics, will help to overcome these problems. Prostate Cancer and Prostatic Diseases (2000) 3, 241-247

SELECTION OF CITATIONS
SEARCH DETAIL
...