Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Biocell ; 32(3): 237-243, Dec. 2008. ilus, tab
Article in English | LILACS | ID: lil-541105

ABSTRACT

Gastric cancer is one of the most common malignancies. DNA methylation is implicated in DNA mismatch repair genes deficiency. In the present study, we evaluated the methylation status of MLH1, MSH2, MSH6 and PMS2 in 20 diffuse- and 26 intestinal-type gastric cancer samples and 20 normal gastric mucosal of gastric cancer patients from Northern Brazil. We found that none of the nonneoplastic samples showed methylation of any gene promoter and 50% of gastric cancer samples showed at least one methylated gene promoter. Methylation frequencies of MLH1, MSH2, MSH6 and PMS2 promoter were 21.74%, 17.39%, 0% and 28.26% respectively in gastric cancer samples. MLH1 and PMS2 methylation were associated with neoplastic samples compared to nonneoplastic ones. PMS2 methylation was associated with diffuse- and intestinal-type cancer compared with normal controls. Intestinal-type cancer showed significant association with MLH1 methylation. Diffuse-type cancer was significantly associated with MSH2 methylation. Our findings show differential gene methylation in tumoral tissue, which allows us to conclude that methylation is associated with gastric carcinogenesis. Methylation of mismatch repair genes was associated with gastric carcinogenesis and may be a helpful tool for diagnosis, prognosis and therapies. However, MSH6 does not seem to be regulated by methylation in our samples.


Subject(s)
Humans , Male , Adult , Female , Middle Aged , DNA Methylation , Stomach Neoplasms/genetics , DNA Mismatch Repair , Sequence Analysis, DNA , Brazil , DNA Repair Enzymes/genetics , Promoter Regions, Genetic
2.
Biocell ; 32(3): 237-243, Dec. 2008. ilus, tab
Article in English | BINACIS | ID: bin-127201

ABSTRACT

Gastric cancer is one of the most common malignancies. DNA methylation is implicated in DNA mismatch repair genes deficiency. In the present study, we evaluated the methylation status of MLH1, MSH2, MSH6 and PMS2 in 20 diffuse- and 26 intestinal-type gastric cancer samples and 20 normal gastric mucosal of gastric cancer patients from Northern Brazil. We found that none of the nonneoplastic samples showed methylation of any gene promoter and 50% of gastric cancer samples showed at least one methylated gene promoter. Methylation frequencies of MLH1, MSH2, MSH6 and PMS2 promoter were 21.74%, 17.39%, 0% and 28.26% respectively in gastric cancer samples. MLH1 and PMS2 methylation were associated with neoplastic samples compared to nonneoplastic ones. PMS2 methylation was associated with diffuse- and intestinal-type cancer compared with normal controls. Intestinal-type cancer showed significant association with MLH1 methylation. Diffuse-type cancer was significantly associated with MSH2 methylation. Our findings show differential gene methylation in tumoral tissue, which allows us to conclude that methylation is associated with gastric carcinogenesis. Methylation of mismatch repair genes was associated with gastric carcinogenesis and may be a helpful tool for diagnosis, prognosis and therapies. However, MSH6 does not seem to be regulated by methylation in our samples.(AU)


Subject(s)
Humans , Male , Adult , Female , Middle Aged , Aged , DNA Methylation , Stomach Neoplasms/genetics , DNA Mismatch Repair , Sequence Analysis, DNA , Brazil , DNA Repair Enzymes/genetics , Promoter Regions, Genetic
3.
Braz J Med Biol Res ; 41(6): 539-43, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18622497

ABSTRACT

Gastric cancer is the forth most frequent malignancy and the second most common cause of cancer death worldwide. DNA methylation is the most studied epigenetic alteration, occurring through a methyl radical addition to the cytosine base adjacent to guanine. Many tumor genes are inactivated by DNA methylation in gastric cancer. We evaluated the DNA methylation status of ANAPC1, CDKN2A and TP53 by methylation-specific PCR in 20 diffuse- and 26 intestinal-type gastric cancer samples and 20 normal gastric mucosa in individuals from Northern Brazil. All gastric cancer samples were advanced stage adenocarcinomas. Gastric samples were surgically obtained at the João de Barros Barreto University Hospital, State of Pará, and were stored at -80 degrees C before DNA extraction. Patients had never been submitted to chemotherapy or radiotherapy, nor did they have any other diagnosed cancer. None of the gastric cancer samples presented methylated DNA sequences for ANAPC1 and TP53. CDKN2A methylation was not detected in any normal gastric mucosa; however, the CDKN2A promoter was methylated in 30.4% of gastric cancer samples, with 35% methylation in diffuse-type and 26.9% in intestinal-type cancers. CDKN2A methylation was associated with the carcinogenesis process for ~30% diffuse-type and intestinal-type compared to non-neoplastic samples. Thus, ANAPC1 and TP53 methylation was probably not implicated in gastric carcinogenesis in our samples. CDKN2A can be implicated in the carcinogenesis process of only a subset of gastric neoplasias.


Subject(s)
Adenocarcinoma/genetics , DNA Methylation/genetics , Genes, p16 , Genes, p53 , Stomach Neoplasms/genetics , Ubiquitin-Protein Ligase Complexes/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Anaphase-Promoting Complex-Cyclosome , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome , Case-Control Studies , Female , Humans , Male , Middle Aged , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
4.
Braz. j. med. biol. res ; 41(6): 539-543, June 2008. ilus, tab
Article in English | LILACS | ID: lil-485848

ABSTRACT

Gastric cancer is the forth most frequent malignancy and the second most common cause of cancer death worldwide. DNA methylation is the most studied epigenetic alteration, occurring through a methyl radical addition to the cytosine base adjacent to guanine. Many tumor genes are inactivated by DNA methylation in gastric cancer. We evaluated the DNA methylation status of ANAPC1, CDKN2A and TP53 by methylation-specific PCR in 20 diffuse- and 26 intestinal-type gastric cancer samples and 20 normal gastric mucosa in individuals from Northern Brazil. All gastric cancer samples were advanced stage adenocarcinomas. Gastric samples were surgically obtained at the João de Barros Barreto University Hospital, State of Pará, and were stored at -80°C before DNA extraction. Patients had never been submitted to chemotherapy or radiotherapy, nor did they have any other diagnosed cancer. None of the gastric cancer samples presented methylated DNA sequences for ANAPC1 and TP53. CDKN2A methylation was not detected in any normal gastric mucosa; however, the CDKN2A promoter was methylated in 30.4 percent of gastric cancer samples, with 35 percent methylation in diffuse-type and 26.9 percent in intestinal-type cancers. CDKN2A methylation was associated with the carcinogenesis process for ~30 percent diffuse-type and intestinal-type compared to non-neoplastic samples. Thus, ANAPC1 and TP53 methylation was probably not implicated in gastric carcinogenesis in our samples. CDKN2A can be implicated in the carcinogenesis process of only a subset of gastric neoplasias.


Subject(s)
Adult , Aged , Female , Humans , Male , Middle Aged , Adenocarcinoma/genetics , DNA Methylation/genetics , Stomach Neoplasms/genetics , Ubiquitin-Protein Ligase Complexes/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Case-Control Studies , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
5.
MAPFRE med ; 17(2): 135-143, feb. 2006. tab
Article in Es | IBECS | ID: ibc-051207

ABSTRACT

El desarrollo neoplásico implica la acumulación secuencial de alteraciones afectando a genes (oncogenes, genes supresores de tumor, genes reparadores) claves en el control de la proliferación y ciclo celular, factores de crecimiento, apoptosis, reparación de ADN, inhibidores de angiogénesis, etc. Con frecuencia estos genes son alterados merced a procesos genéticos: mutaciones, alteraciones de estructura, que resultan en proteínasinactivas o funcionalmente deficientes. No obstante, dichos genes son susceptibles de una expresión anómala como resultado de alteraciones epigenéticas, principalmente metilación de islas CpG de los promotores génicos. Nuestro estudio en 271muestras de tumores del Sistema Nervioso de alto grado demalignidad (gliomas, meduloblastomas, neuroblastomas, metástasis en cerebro) ha demostrado tasas significativas de dicha variación epigenética afectando a genes como RB1, p16INK4a, p14ARF, (controladores del ciclo celular), DAP-kinasa, Caspasa 8 (controladores de apoptosis y metástais), MGMT(reparación ADN), THBS1 (inhibición de angiogénesis), TP53,TP73, etc. Nuestros hallazgos permiten indicar que la hipermetilación de determinados genes puede representar un factor predictivo de la agresividad biológica de los tumores:– MGMT, en gliomas (sensibilidad a quimioterapia).– p14ARF y RB1 en astrocitomas anaplásicos y glioblastomas secundarios.– p16INK4a en oligodendrogliomas.– Caspasa 8 en neuroblastomas y meduloblastomas.– DAP-kinasa en tumores metastásicos en cerebro. Estos hallazgos sugieren que el análisis epigenético de genes específicos puede contribuir a la identificación de aquellos casos más agresivos, susceptibles de seguimientos clínicos y terapias específicas


Tumour development is a multi-step process implying the sequential accumulation of molecular genetic andepigenetic alterations. These abnormalities primarily involve oncogenes, tumour suppressor genes, DNA repair genes, etc. In addition to the classical genetic mechanisms (mutation, deletion, etc) contributing to gene dysfunction, epigenetic changes, mainly aberrant CpG island promoter methylation contributes to tumour related gene silencing. Our study in 271 samples derived from malignant neurogenic neoplams (gliomas, medulloblastomas, neuroblastomas and metastatictumours) demonstrated non-random methylation rates involving cell-cycle control genes: RB1, p16INK4a,p14ARF, as well as DAP-Kinase, Caspase 8 (apoptosis and metastatic inhibitors), MGMT (a DNA-repair gene),or THBS1 (an angiogenesis inhibitor gene). Our finding salso suggested that aberrant methylation of specific genes may represent a predictive factor for biological aggressiveness of these malignant tumours:– MGMT, in gliomas (response to alkylating agents).– p14ARF and RB1 in anaplastic astrocytomas and secondaty glioblastoma.– p16INK4a in oligodendrogliomas.– Caspase 8 in neuroblastomas and medulloblastomas.– DAP-kinasa in brain metastases. Accordingly, epigenetic analysis may contribute in the screening of those aggressive samples susceptible of special clinical follow-up and specific therapies


Subject(s)
Humans , Biomarkers, Tumor/isolation & purification , DNA Methylation , Nervous System Neoplasms/diagnosis , CpG Islands , Chromosome Deletion , Cell Cycle/genetics , Genes, Retinoblastoma
6.
Histopathology ; 47(6): 631-7, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16324202

ABSTRACT

AIMS: Medulloblastoma (MB), a kind of infratentorial primitive neuroectodermal tumour (PNET), is the most frequent malignant brain tumour in childhood. In contrast, supratentorial PNET (sPNET) are very infrequent tumours, but they are histologically similar to MB, although they present a worse clinical outcome. We investigated the differences in genetic abnormalities between sPNET and MB. METHODS AND RESULTS: We analysed 20 central PNET (14 MB and six sPNET) by conventional comparative genomic hybridization (CGH) in order to determine whether a different genetic profile for each tumour exists. Isochromosome 17q was detected in four of the 14 MB cases, but not in any sPNET. Gains at 17q and 7 happened more frequently in MB, and those at 1q in sPNET. Losses at chromosome 10 were detected only in MB, while losses at 16p and 19p happened more frequently in sPNET. A new amplification site, on 4q12, was detected in two MB. CONCLUSIONS: Central PNET are a heterogeneous group of tumours from the genetic point of view. The present and previous data, together with further results from larger series, might contribute to the establishment of specific treatments for supratentorial and infratentorial PNET.


Subject(s)
Brain Neoplasms/genetics , Genetic Heterogeneity , Infratentorial Neoplasms/genetics , Neuroectodermal Tumors, Primitive/genetics , Supratentorial Neoplasms/genetics , Brain Neoplasms/pathology , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 10 , Chromosomes, Human, Pair 16 , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 4 , Chromosomes, Human, Pair 7 , Humans , Infratentorial Neoplasms/pathology , Medulloblastoma/genetics , Medulloblastoma/pathology , Neuroectodermal Tumors, Primitive/pathology , Nucleic Acid Hybridization , Supratentorial Neoplasms/pathology
7.
Neuropathol Appl Neurobiol ; 31(4): 384-94, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16008822

ABSTRACT

The epidermal growth factor receptor (EGFR) is a transmembrane glycoprotein with tyrosine kinase activity. This report investigates the presence of mutations, amplification and/or over-expression of the EGFR gene in 86 glial tumours including 44 glioblastomas, 21 anaplastic astrocytomas, and 21 WHO grade II astrocytomas, using polymerase chain reaction/single-strand conformation polymorphism, semiquantitative reverse-transcription-polymerase chain reaction (RT-PCR) and Southern Blot techniques. Gene amplification values were found in 34 tumours. Amplification levels were not uniform, as the transmembrane region presented lower amplification rates than extra- and intracellular domains. For the 19 samples with sufficient available tumour tissue we found over-expression in 11, and no EGFR mRNA expression in three. Ten cases showed deletion transcripts, and EGFR VIII was identified in all of these cases. One of the cases with EGFR vIII also presented a truncated form, C-958, while another showed an in frame tandem duplication of exons 18--25. We found 14 cases with sequence/structure gene alterations, including seven on which genomic novel DNA changes were identified: a missense mutation (1052C > T/Ala265Val), an insertion (InsCCC2498/Ins Pro748), three intronic changes (E6+72delG, E22--14C>G and E18--109T>C), a new polymorphic variant E12+ 22A > T, and one case that presented a 190 bp insertion, that was produced by the intron-7-exon-8 duplication and generated a truncated EGFR with intact exons 1--8 followed by an additional amino acidic sequence: Val-Ile-Met-Trp. These findings corroborate that EGFR is non-randomly involved in malignant glioma development and that different mutant forms participate in aberrant activation of tyrosine kinase pathways.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Epidermal Growth Factor/genetics , Gene Amplification , Base Sequence , Blotting, Southern , DNA Mutational Analysis , Humans , Molecular Sequence Data , Mutation , Polymorphism, Single-Stranded Conformational , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
8.
Rev Neurol ; 37(10): 912-6, 2003.
Article in Spanish | MEDLINE | ID: mdl-14634918

ABSTRACT

INTRODUCTION: The influence of surgery, radiotherapy and/or chemotherapy on the outcome on the results in patients with malignant gliomas is controversial. PATIENTS AND METHODS: We studied 44 patients (26, women; 18 men; age 38 72 years) diagnosed with grades III and IV astrocytoma who had been operated and then received adjuvant radiotherapy and either BCNU or temozolamyde chemotherapy. Survival time and adverse effects of the chemotherapy were analysed. CONCLUSION: Aggressive surgery associated with radiotherapy and temozolamyde chemotherapy prolonged survival in our patients with malignant astrocytomas.


Subject(s)
Astrocytoma/therapy , Brain Neoplasms/therapy , Adult , Aged , Antineoplastic Agents/adverse effects , Astrocytoma/mortality , Brain Neoplasms/mortality , Combined Modality Therapy , Female , Humans , Male , Middle Aged , Survival Rate
9.
Eur J Cancer ; 39(10): 1478-85, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12826052

ABSTRACT

Aberrant hypermethylation occurs in tumour cell CpG islands and is an important pathway for the repression of gene transcription in cancers. We investigated aberrant hypermethylation of 11 genes by methylation-specific polymerase chain reaction (PCR), after treatment of the DNA with bisulphite, and correlated the findings with MYCN amplification and allelic status at 1p in a series of 44 neuroblastic tumours. This tumour series includes five ganglioneuromas (G), one ganglioneuroblastoma (GN) and 38 neuroblastomas (six stage 1 tumours; five stage 2 tumours; six stage 3 cases; 19 stage 4 tumours, and two stage 4S cases). Aberrant methylation of at least one of the 11 genes studied was detected in 95% (42 of 44) of the cases. The frequencies of aberrant methylation were: 64% for thrombospondin-1 (THBS1); 30% for tissue inhibitor of metalloproteinase 3 (TIMP-3); 27% for O6-methylguanine-DNA methyltransferase (MGMT); 25% for p73; 18% for RB1; 14% for death-associated protein kinase (DAPK), p14ARF, p16INK4a and caspase 8, and 0% for TP53 and glutathione S-transferase P1 (GSTP1). No aberrant methylation was observed in four control normal tissue samples (brain and adrenal medulla). MYCN amplification was found in 11 cases (all stage 4 neuroblastomas), whereas allelic loss at 1p was identified in 16 samples (13 stage 4 and two stage 3 neuroblastomas, and one ganglioneuroma). All but one case with caspase 8 methylation also displayed MYCN amplification. Our results suggest that promoter hypermethylation is a frequent epigenetic event in the tumorigenesis of neuroblastic tumours, but no specific pattern of hypermethylated genes could be demonstrated.


Subject(s)
DNA Methylation , Genes, myc/genetics , Neuroblastoma/genetics , Child , Child, Preschool , Female , Gene Amplification , Humans , Infant , Loss of Heterozygosity , Male , Polymerase Chain Reaction/methods
10.
Br J Cancer ; 88(1): 109-14, 2003 Jan 13.
Article in English | MEDLINE | ID: mdl-12556968

ABSTRACT

A series of 136 nervous system tumours were studied to determine the methylation status of the CpG island contained within the promoter region of the RB1 gene, as well as mutation analysis of the essential promoter region and exons 20-24 (and surrounding intronic regions) coding for the protein-binding pocket domain. Methylation of the RB1 CpG island was detected in 26 samples corresponding to nine glioblastomas, three anaplastic astrocytomas, one mixed oligo-astrocytoma, one ependymoma, two medulloblastomas, two primary central nervous system lymphomas, two neurofibrosarcomas, and six brain metastasis from solid tumours. No inactivating mutations were found within the RB1 promoter region, whereas one glioblastoma and one oligodendroglioma displayed similar sequence variations consisting of 12 and 8 base pair deletions at intron 21. These results suggest that RB1 CpG island hypermethylation is a common epigenetic event that is associated with the development of malignant nervous system tumours.


Subject(s)
CpG Islands , DNA Methylation , Nervous System Neoplasms/genetics , Promoter Regions, Genetic/genetics , Retinoblastoma Protein/genetics , DNA Mutational Analysis , Humans , Mutation
11.
Cancer Genet Cytogenet ; 129(1): 88-91, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11520574

ABSTRACT

The p73 gene has been mapped to 1p36.33, a chromosome region that is frequently deleted in a wide variety of neoplasms including meningiomas. The protein encoded by p73 shows structural and functional similarities to p53 and may thus represent a candidate tumor suppressor gene. To determine whether p73 is involved in the development of meningiomas, we examined 30 meningioma samples with proven 1p deletion for mutations of p73. Sequence analysis of the entire coding region of the p73 gene revealed previously reported polymorphisms in eight cases. A tumor-specific missense mutation as a result of an A-to-G transition with an Asn204Ser change was found in one meningioma that nevertheless retained the normal allele. These results suggest that if p73 plays a role in meningioma carcinogenesis, it must be in a manner different from the Knudson two-hit model.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1 , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Meningeal Neoplasms/genetics , Meningioma/genetics , Nuclear Proteins/genetics , Humans , Tumor Protein p73 , Tumor Suppressor Proteins
12.
Int J Oncol ; 19(3): 609-12, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11494043

ABSTRACT

In subgroups of astrocytic neoplasms, including glioblastoma (GBM), mutations of the p53 tumour suppressor gene lead to loss of growth-suppressive properties. A p53-related gene termed p73 has recently been identified; its gene product shows structural and functional similarities to p53. After being mapped to chromosome region 1p36, p73 was proposed to act as a tumour suppressor gene, as this region is frequently deleted in a variety of human cancers, including astrocytic tumours. To determine whether p73 is involved in astrocytoma/GBM development, we analysed 10 pilocytic astrocytomas, 15 WHO grade II astrocytomas, 15 WHO grade III anaplastic astrocytomas, and 20 GBM for p73 gene alterations. In parallel, we used six polymorphic markers to determine the allelic status of region 1p36 in this tumour series. Although loss of heterozygosity was evidenced in 12 of 60 cases (20% of samples), PCR-SSCP and direct sequencing failed to detect any gene mutation in the entire coding region and intronic sequences of p73. Eight tumours displayed five distinct polymorphic nucleotide changes, also present in the corresponding normal DNA. These variations consisted of T-->C variation, with no change in Thr173; C-->T transition, with no change in His197; exon 9 simultaneous double change C-->T and T-->C , with no variations in Ala336 and His349, respectively, and C-->T change at exon 9/-24 position of intron 8. These results suggest that, in astrocytic gliomas, p73 may not play a major role as a tumour suppressor, but the relatively high incidence of LOH confirms the presence at 1p36 of an as yet unidentified gene of this category, with a key function in astrocytoma/GBM progression.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Chromosomes, Human, Pair 1/genetics , DNA-Binding Proteins/genetics , Glioblastoma/genetics , Mutation , Nuclear Proteins/genetics , Astrocytoma/surgery , Base Sequence , Brain Neoplasms/surgery , DNA Mutational Analysis , DNA, Neoplasm/analysis , DNA-Binding Proteins/metabolism , Genes, Tumor Suppressor/genetics , Glioblastoma/surgery , Humans , Loss of Heterozygosity , Molecular Sequence Data , Nuclear Proteins/metabolism , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Tumor Protein p73 , Tumor Suppressor Proteins
13.
Br J Cancer ; 85(2): 204-8, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11461077

ABSTRACT

Loss of heterozygosity (LOH) involving the distal chromosome 1 p36 region occurs frequently in nonastrocytic brain tumours, but the tumour suppressor gene targeted by this deletion is unknown. p73 is a novel gene that has high sequence homology and similar gene structure to the p53 gene; it has been mapped to 1 p36, and may thus represent a candidate for this tumour suppressor gene. To determine whether p73 is involved in nonastrocytic brain tumour development, we analysed 65 tumour samples including 26 oligodendrogliomas, 4 ependymomas, 5 medulloblastomas, 10 meningiomas, 2 meningeal haemangiopericytomas, 2 neurofibrosarcomas, 3 primary lymphomas, 8 schwannomas and 5 metastatic tumours to the brain, for p73 alterations. Characterization of allelic loss at 1 p36-p35 showed LOH in about 50% of cases, primarily involving oligodendroglial tumours (22 of 26 cases analysed; 85%) and meningiomas (4 of 10; 40%). PCR-SSCP and direct DNA sequencing of exons 2 to 14 of p73 revealed a missense mutation in one primary lymphoma: a G-to-A transition, with Glu291Lys change. 8 additional cases displayed no tumour-specific alterations, as 3 distinct polymorphic changes were identified: a double polymorphic change of exon 5 was found in one ependymoma and both samples derived from an oligodendroglioma, as follows: a G-to-A transition with no change in Pro 146, and a C-to-T variation with no change in Asn 204: a delG at exon 3/+12 position was identified in 4 samples corresponding to 2 oligodendrogliomas, 1 ependymoma and 1 meningioma, and a C-to-T change at exon 2/+10 position was present in a metastatic tumour. Although both LOH at 1 p36 and p73 sequence changes were evidenced in 4 cases, it is difficult to establish a causal role of the p73 variations and nonastrocytic brain tumours development.


Subject(s)
Brain Neoplasms/genetics , DNA-Binding Proteins/genetics , Mutation , Nuclear Proteins/genetics , Brain Neoplasms/classification , Brain Neoplasms/pathology , Chromosomes, Human, Pair 1 , Genes, Tumor Suppressor , Humans , Loss of Heterozygosity , Polymorphism, Single-Stranded Conformational , Tumor Protein p73 , Tumor Suppressor Proteins
14.
Cancer Genet Cytogenet ; 124(1): 76-9, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11165326

ABSTRACT

Cytogenetic studies were conducted on 30 pituitary adenomas, using both direct and/or short-term in vitro culture methods. An apparently normal chromosome complement was found in 14 tumors; 5 adenomas were characterized by hyperdiploid or near-triploid modal chromosome numbers. Recurrent numerical deviations were identified in 12 samples, which primarily involved gains of chromosomes 4, 7, 8, 9, 12, and 20 by gains, and losses of chromosomes 10, 14, 19, and 22. Four adenomas were shown to have structural chromosome rearrangements with no apparent recurrent pattern of involvement.


Subject(s)
Adenoma/genetics , Chromosome Aberrations/genetics , Pituitary Neoplasms/genetics , Adolescent , Adult , Aged , Chromosome Deletion , Chromosome Disorders , Female , Humans , Karyotyping , Male , Middle Aged , Ploidies
15.
Cancer Genet Cytogenet ; 120(1): 30-6, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10913674

ABSTRACT

Loss of heterozygosity (LOH) for loci on chromosome arm 1p is a relatively common event in human meningioma, and this anomaly has been proposed to be associated with the development of grade II or grade III forms (atypical and anaplastic meningiomas). Nevertheless, the limited data available do not allow the establishment of the frequency and the extent of the affected 1p regions. To determine the status of chromosome 1p in meningiomas, we have performed a comprehensive analysis of LOH on 1p in 100 meningiomas using a high density of 1p-marker loci. Allelic loss was found in 35% of tumors, most corresponding to nontypical meningiomas that also displayed losses for loci on chromosome 22. Although some tumors displayed complex rearrangements leading to distinct 1p deletions, the patterns of loss indicated two main target regions: 1p36 and 1p34-p32, which represent the most frequently involved regions, whereas 1p22 and 1p21.1-1p13 regions appeared deleted in some tumors. These results suggest that there may be several putative tumor suppressor genes on 1p, the inactivation of which may be important in the pathogenesis of meningiomas, as well as in other tumor types.


Subject(s)
Brain Neoplasms/genetics , Chromosomes, Human, Pair 1 , Loss of Heterozygosity , Meningioma/genetics , Humans , Microsatellite Repeats
16.
Cancer Genet Cytogenet ; 116(2): 142-7, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10640146

ABSTRACT

The hRAD54 protein belongs to a superfamily of DNA helicases, and mutations in genes with DNA helicase function have been found to be responsible for cancer-prone syndromes (xeroderma pigmentosum, Bloom syndrome, Werner syndrome). hRAD54 thus could be a candidate modifier gene in tumors characterized by allelic imbalance at 1p32, the chromosome region in which this gene is located. Using a panel of 38 1p and five 1q markers, we therefore performed deletion-mapping analysis on a series of 35 oligodendrogliomas, which were also studied for mutations in the hRAD54 gene. Deletions of the short arm of chromosome 1 were evidenced in 26 tumors, mostly involving 1p36-1p13; all thus displayed loss of the 1p32 region. We used PCR/SSCP to examine all 18 exons of the hRAD54 gene for mutations in 25 tumors, but the mobility shifts detected corresponded to previously identified polymorphic changes: T-to-C transition at nucleotide 2865 (with no amino acid change) and at nucleotide 3008, at the 3' untranslated region. We conclude that hRAD54 gene alterations are not required for malignant transformation of oligodendrogliomas.


Subject(s)
Chromosome Mapping , Chromosomes, Human, Pair 1/genetics , DNA Helicases/genetics , DNA, Neoplasm/genetics , Nuclear Proteins/genetics , Oligodendroglioma/genetics , Chromosome Deletion , DNA Mutational Analysis , DNA-Binding Proteins , Gene Deletion , Humans , Loss of Heterozygosity , Polymorphism, Single-Stranded Conformational
17.
Mol Carcinog ; 24(4): 300-4, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10326867

ABSTRACT

The hRAD54 gene is related to a family of genes involved in DNA recombination and repair and encodes a protein with DNA helicase activity. hRAD54 has been mapped to 1p32, a region frequently involved in deletions in a variety of tumor types, including atypical and anaplastic meningiomas. To determine whether alterations of hRAD54 are a common event in meningeal tumors, by means of polymerase chain reaction-single-stranded conformation analysis we examined 29 tumor samples characterized by 1p deletions for hRAD54 mutations. Although 18 tumors displayed allelic loss at the gene region (1p32) as determined by microsatellite marker analysis, the sole coding-sequence alteration detected corresponded to a T-->C transition, with no amino-acid change. The genotype distribution was 10.34% TT, 44.8% TC, and 44.8% CC, whereas in the normal controls it was 3.77% TT, 13.2% TC, and 83.01% CC, and most meningiomas with 1 p32 deletion retained allele C. Another polymorphism due to a T-->C change was evidenced at nt 3008, in the 3' untranslated region. This change was evidenced in all cases we sequenced. These results appear to exclude the involvement of the hRAD54 gene in the pathogenesis of the nontypical meningiomas, although a detrimental effect of the hRAD54 polymorphisms cannot be ruled out.


Subject(s)
Chromosomes, Human, Pair 1/genetics , DNA, Neoplasm/genetics , Gene Deletion , Meningeal Neoplasms/genetics , Meningioma/genetics , Mutation , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , 3' Untranslated Regions/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Chromosomes, Human, Pair 1/ultrastructure , DNA Helicases , DNA Mutational Analysis , DNA Repair/genetics , DNA-Binding Proteins , Exons/genetics , Female , Genotype , Humans , Loss of Heterozygosity , Male , Microsatellite Repeats , Middle Aged , Point Mutation , Polymorphism, Genetic , Polymorphism, Single-Stranded Conformational
18.
Oncogene ; 18(13): 2231-9, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10327069

ABSTRACT

Formation of meningiomas and their progression to malignancy may be a multi-step process, implying accumulation of genetic mutations at specific loci. To determine the relationship between early NF2 gene inactivation and the molecular mechanisms that may contribute to meningioma tumor progression, we have performed deletion mapping analysis at chromosomes 1, 14 and 22 in a series of 81 sporadic meningiomas (54 grade I (typical), 25 grade II (atypical) and two grade III (anaplastic)), which were also studied for NF2 gene mutations. Single-strand conformational polymorphism analysis was used to identify 11 mutations in five of the eight exons of the NF2 gene studied. All 11 tumors displayed loss of heterozygosity (LOH) for chromosome 22 markers; this anomaly was also detected in 33 additional tumors. Twenty-nine and 23 cases were characterized by LOH at 1p and 14q, respectively, mostly corresponding to aggressive tumors that also generally displayed LOH 22. All three alterations were detected in association in seven grade II and two grade III meningiomas, corroborating the hypothesis that the formation of aggressive meningiomas follows a multi-step tumor progression model.


Subject(s)
Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 22/genetics , Genes, Neurofibromatosis 2 , Meningeal Neoplasms/genetics , Meningioma/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Cell Transformation, Neoplastic/genetics , DNA Mutational Analysis , DNA, Neoplasm/genetics , Disease Progression , Female , Genotype , Humans , Loss of Heterozygosity , Male , Meningeal Neoplasms/pathology , Meningioma/pathology , Microsatellite Repeats , Middle Aged , Polymorphism, Single-Stranded Conformational , Sequence Deletion
19.
Int J Mol Med ; 1(5): 889-92, 1998 May.
Article in English | MEDLINE | ID: mdl-9852312

ABSTRACT

Schwannomas are common benign tumours of schwann cell origin, frequently found in patients with neurofibromatosis type 2 (NF2). Inactivation of the NF2 tumour suppressor gene appears to be a molecular event responsible for the development of up to 60% of cases, but no data are available on other superimposed secondary or alternative molecular abnormalities in those schwannomas lacking NF2 gene inactivation. We analysed 23 sporadic schwannomas for mutations in the NF2 gene and for the allelic status at 1p, 14q and 22q, as alterations of these genomic regions appear to be related to tumour progression in meningiomas, another NF2-associated neoplasm. Nine samples displayed allelic losses for markers on chromosome 22, and deletions at 1p were detected in two. No case showed losses for 14q. Three tumours displayed NF2 gene mutations, at exons 2, 7 and 12. Our results confirm that inactivation of the NF2 gene is a primary event in schwannoma development, and provide data suggesting that allelic loss at 1p may contribute to the pathogenesis of a small subgroup of this histological tumour type.


Subject(s)
Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 22 , Genes, Neurofibromatosis 2/genetics , Neurilemmoma/genetics , Alleles , DNA Mutational Analysis , Genetic Markers , Humans , Loss of Heterozygosity/genetics , Mutation , Sequence Analysis, DNA
20.
Int J Oncol ; 12(4): 935-8, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9499458

ABSTRACT

Six novel mutations were identified in the NF2 tumor suppressor gene in a panel of meningiomas and neurinomas. Screening was performed using a combination of single-strand conformation polymorphism and heteroduplex analyses on polymerase chain reaction-amplified DNA from tumors and matched peripheral blood lymphocytes. Mutations involved exons 2, 7, 11 and 12, and corresponded to three frameshift, one nonsense, one missense and one polymorphism.


Subject(s)
Genes, Tumor Suppressor , Membrane Proteins/genetics , Mutation , Chromosome Aberrations , Humans , Neurofibromin 2 , Polymorphism, Single-Stranded Conformational
SELECTION OF CITATIONS
SEARCH DETAIL
...