Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Genet Mol Res ; 11(1): 332-9, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22370935

RESUMO

The gene XRCC3 (X-ray cross complementing group 3) has the task of repairing damage that occurs when there is recombination between homologous chromosomes. Repair of recombination between homologous chromosomes plays an important role in maintaining genome integrity, although it is known that double-strand breaks are the main inducers of chromosomal aberrations. Changes in the XRCC3 protein lead to an increase in errors in chromosome segregation due to defects in centrosomes, resulting in aneuploidy and other chromosomal aberrations, such as small increases in telomeres. We examined XRCC3 Thr241Met polymorphism using PCR-RFLP in 80 astrocytoma and glioblastoma samples. The individuals of the control group (N = 100) were selected from the general population of the São Paulo State. Odds ratio and 95%CI were calculated using a logistic regression model. Patients who had the allele Met of the XRCC3 Thr241Met polymorphism had a significantly increased risk of tumor development (odds ratio = 3.13; 95% confidence interval = 1.50-6.50). There were no significant differences in overall survival of patients. We suggest that XRCC3 Thr241Met polymorphism is involved in susceptibility for developing astrocytomas and glioblastomas.


Assuntos
Astrocitoma/genética , Proteínas de Ligação a DNA/genética , Glioblastoma/genética , Adolescente , Adulto , Idoso , Alelos , Centrossomo/patologia , Criança , Pré-Escolar , Aberrações Cromossômicas , Segregação de Cromossomos/genética , Reparo do DNA , Feminino , Predisposição Genética para Doença , Variação Genética , Genótipo , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Adulto Jovem
2.
Genet Mol Res ; 10(2): 1120-9, 2011 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-21710464

RESUMO

XRCC genes (X-ray cross-complementing group) were discovered mainly for their roles in protecting mammalian cells against damage caused by ionizing radiation. Studies determined that these genes are important in the genetic stability of DNA. Although the loss of some of these genes does not necessarily confer high levels of sensitivity to radiation, they have been found to represent important components of various pathways of DNA repair. To ensure the integrity of the genome, a complex system of DNA repair was developed. Base excision repair is the first defense mechanism of cells against DNA damage and a major event in preventing mutagenesis. Repair genes may play an important role in maintaining genomic stability through different pathways that are mediated by base excision. In the present study, we examined XRCC1Arg194Trp and XRCC1Arg399Gln polymorphism using PCR-RFLP in 80 astrocytoma and glioblastoma samples. Patients who had the allele Trp of the XRCC1Arg194Trp polymorphism had an increased risk of tumor development (OR = 8.80; confidence interval at 95% (95%CI) = 4.37-17.70; P < 0.001), as did the allele Gln of XRCC1Arg399Gln (OR = 1.01; 95%CI = 0.53-1.93; P = 0.971). Comparison of overall survival of patients did not show significant differences. We suggest that XRCC1Arg194Trp and XRCC1Arg399Gln polymorphisms are involved in susceptibility for developing astrocytomas and glioblastomas.


Assuntos
Neoplasias Encefálicas/genética , Proteínas de Ligação a DNA/genética , Glioma/genética , Arginina/química , Primers do DNA , Proteínas de Ligação a DNA/química , Glicina/química , Humanos , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único , Triptofano/química , Proteína 1 Complementadora Cruzada de Reparo de Raio-X
3.
Genet Mol Res ; 9(4): 2328-34, 2010 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-21128213

RESUMO

Glutathione S-transferases (GSTs) constitute a superfamily of ubiquitous multifunctional enzymes that are involved in the cellular detoxification of a large number of endogenous and exogenous chemical agents that have electrophilic functional groups. People who have deficiencies in this family of genes are at increased risk of developing some types of tumors. We examined GSTP1 Ile105Val polymorphism using PCR-RFLP in 80 astrocytoma and glioblastoma samples. Patients who had the Val allele of the GSTP1 Ile105Val polymorphism had an increased risk of tumor development (odds ratio = 8.60; 95% confidence interval = 4.74-17.87; P < 0.001). Overall survival of patients did not differ significantly. We suggest that GSTP1 Ile105Val polymorphisms are involved in susceptibility to developing astrocytomas and glioblastomas.


Assuntos
Astrocitoma/genética , Neoplasias Encefálicas/genética , Glioblastoma/genética , Glutationa Transferase/genética , Isoleucina/genética , Polimorfismo de Nucleotídeo Único , Valina/genética , Adolescente , Adulto , Idoso , Astrocitoma/enzimologia , Sequência de Bases , Neoplasias Encefálicas/enzimologia , Estudos de Casos e Controles , Primers do DNA , Feminino , Glioblastoma/enzimologia , Glutationa Transferase/química , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Adulto Jovem
4.
Genet Mol Res ; 8(4): 1257-63, 2009 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-19876867

RESUMO

Disruption or loss of tumor suppressor gene TP53 is implicated in the development or progression of almost all different types of human malignancies. Other members of the p53 family have been identified. One member, p73, not only shares a high degree of similarity with p53 in its primary sequence, but also has similar functions. Like p53, p73 can bind to DNA and activate transcription. Using PCR-SSCP and gene sequencing, we analyzed the TP53 and TP73 genes in a case of a grade III anaplastic astrocytoma that progressed to glioblastoma. We found a deletion of AAG at position 595-597 of TP53 (exon 6), resulting in the deletion of Glu 199 in the protein and a genomic polymorphism of TP73, identified as an A-to-G change, at position E8/+15 at intron 8 (IVS8-15A>G). The mutation found at exon 6 of the gene TP53 could be associated with the rapid tumoral progression found in this case, since the mutated p53 may inactivate the wild-type p53 and the p73alpha protein, which was conserved here, leading to an increase in cellular instability.


Assuntos
Astrocitoma/genética , Neoplasias Encefálicas/genética , Proteínas de Ligação a DNA/genética , Proteínas Nucleares/genética , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética , Adulto , Sequência de Bases , Primers do DNA , Humanos , Masculino , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Proteína Tumoral p73
5.
Genet Mol Res ; 8(1): 8-18, 2009 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-19224462

RESUMO

The p53 tumor suppressor gene is the most frequently mutated gene in human cancer; this gene is mutated in up to 50% of human tumors. It has a critical role in the cell cycle, apoptosis and cell senescence, and it participates in many crucial physiological and pathological processes. Polymorphisms of p53 have been suggested to be associated with genetically determined susceptibility in various types of cancer. Another process involved with the development and progression of tumors is DNA hypermethylation. Aberrant methylation of the promoter is an alternative epigenetic change in genetic mechanisms, leading to tumor suppressor gene inactivation. In the present study, we examined the TP53 Arg72Pro and Pro47Ser polymorphisms using PCR-RFLP and the pattern of methylation of the p53 gene by methylation-specific PCR in 90 extra-axial brain tumor samples. Patients who had the allele Pro of the TP53 Arg72Pro polymorphism had an increased risk of tumor development (odds ratio, OR = 3.23; confidence interval at 95%, 95%CI = 1.71-6.08; P = 0.003), as did the allele Ser of TP53 Pro47Ser polymorphism (OR = 1.28; 95%CI = 0.03-2.10; P = 0.01). Comparison of overall survival of patients did not show significant differences. In the analysis of DNA methylation, we observed that 37.5% of meningiomas, 30% of schwannomas and 52.6% of metastases were hypermethylated, suggesting that methylation is important for tumor progression. We suggest that TP53 Pro47Ser and Arg72Pro polymorphisms and DNA hypermethylation are involved in susceptibility for developing extra-axial brain tumors.


Assuntos
Neoplasias Encefálicas/genética , Metilação de DNA/genética , Genes p53/genética , Meningioma/genética , Estudos de Casos e Controles , Códon , Predisposição Genética para Doença , Humanos , Neurilemoma/genética , Reação em Cadeia da Polimerase , Polimorfismo Genético
6.
Genet Mol Res ; 7(2): 451-9, 2008 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-18551412

RESUMO

Cancer is one of the most common and severe problems in clinical medicine, and nervous system tumors represent about 2% of the types of cancer. The central role of the nervous system in the maintenance of vital activities and the functional consequences of the loss of neurons can explain how severe brain cancers are. The cell cycle is a highly complex process, with a wide number of regulatory proteins involved, and such proteins can suffer alterations that transform normal cells into malignant ones. The INK4 family members (CDK inhibitors) are the cell cycle regulators that block the progression of the cycle through the R point, causing an arrest in G1 stage. The p14ARF (alternative reading frame) gene is a tumor suppressor that inhibits p53 degradation during the progression of the cell cycle. The PTEN gene is related to the induction of growth suppression through cell cycle arrest, to apoptosis and to the inhibition of cell adhesion and migration. The purpose of the present study was to assess the mutational state of the genes p14ARF, p15INK4b, p16INK4a, and PTEN in 64 human nervous system tumor samples. Homozygous deletions were found in exon 2 of the p15INK4b gene and exon 3 of the p16INK4a gene in two schwannomas. Three samples showed a guanine deletion (63 codon) which led to a loss of heterozygosity in the p15 gene, and no alterations could be seen in the PTEN gene. Although the group of patients was heterogeneous, our results are in accordance with other different studies that indicate that homozygous deletion and loss of heterozygosity in the INK4 family members are frequently observed in nervous system tumors.


Assuntos
Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Neoplasias do Sistema Nervoso/genética , PTEN Fosfo-Hidrolase/genética , Proteína Supressora de Tumor p14ARF/genética , Análise Mutacional de DNA/métodos , Deleção de Genes , Homozigoto , Humanos , Perda de Heterozigosidade , Neoplasias do Sistema Nervoso/patologia , Reação em Cadeia da Polimerase
7.
Genet Mol Res ; 7(1): 207-16, 2008 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-18393224

RESUMO

The TP53 tumor suppressor gene codifies a protein responsible for preventing cells with genetic damage from growing and dividing by blocking cell growth or apoptosis pathways. A common single nucleotide polymorphism (SNP) in TP53 codon 72 (Arg72Pro) induces a 15-fold decrease of apoptosis-inducing ability and has been associated with susceptibility to human cancers. Recently, another TP53 SNP at codon 47 (Pro47Ser) was reported to have a low apoptosis-inducing ability; however, there are no association studies between this SNP and cancer. Aiming to study the role of TP53 Pro47Ser and Arg72Pro on glioma susceptibility and oncologic prognosis of patients, we investigated the genotype distribution of these SNPs in 94 gliomas (81 astrocytomas, 8 ependymomas and 5 oligodendrogliomas) and in 100 healthy subjects by the polymerase chain reaction-restriction fragment length polymorphism approach. Chi-square and Fisher exact test comparisons for genotype distributions and allele frequencies did not reveal any significant difference between patients and control groups. Overall and disease-free survivals were calculated by the Kaplan-Meier method, and the log-rank test was used for comparisons, but no significant statistical difference was observed between the two groups. Our data suggest that TP53 Pro47Ser and Arg72Pro SNPs are not involved either in susceptibility to developing gliomas or in patient survival, at least in the Brazilian population.


Assuntos
Glioma/genética , Polimorfismo de Nucleotídeo Único , Proteína Supressora de Tumor p53/genética , Adolescente , Adulto , Idoso , Apoptose/genética , Brasil , Estudos de Casos e Controles , Criança , Pré-Escolar , Feminino , Frequência do Gene , Genes p53 , Predisposição Genética para Doença , Genótipo , Glioma/etiologia , Glioma/mortalidade , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Prognóstico , Análise de Sobrevida
8.
Genet. mol. res. (Online) ; 7(1): 207-216, Jan. 2008. ilus, tab
Artigo em Inglês | LILACS | ID: lil-553787

RESUMO

The TP53 tumor suppressor gene codifies a protein responsible for preventing cells with genetic damage from growing and dividing by blocking cell growth or apoptosis pathways. A common single nucleotide polymorphism (SNP) in TP53 codon 72 (Arg72Pro) induces a 15-fold decrease of apoptosis-inducing ability and has been associated with susceptibility to human cancers. Recently, another TP53 SNP at codon 47 (Pro47Ser) was reported to have a low apoptosis-inducing ability; however, there are no association studies between this SNP and cancer. Aiming to study the role of TP53 Pro47Ser and Arg72Pro on glioma susceptibility and oncologic prognosis of patients, we investigated the genotype distribution of these SNPs in 94 gliomas (81 astrocytomas, 8 ependymomas and 5 oligodendrogliomas) and in 100 healthy subjects by the polymerase chain reaction-restriction fragment length polymorphism approach. Chi-square and Fisher exact test comparisons for genotype distributions and allele frequencies did not reveal any significant difference between patients and control groups. Overall and disease-free survivals were calculated by the Kaplan-Meier method, and the log-rank test was used for comparisons, but no significant statistical difference was observed between the two groups. Our data suggest that TP53 Pro47Ser and Arg72Pro SNPs are not involved either in susceptibility to developing gliomas or in patient survival, at least in the Brazilian population.


Assuntos
Humanos , Masculino , Feminino , Pré-Escolar , Criança , Adolescente , Adulto , Pessoa de Meia-Idade , Glioma/genética , Polimorfismo de Nucleotídeo Único , /genética , Apoptose/genética , Brasil , Estudos de Casos e Controles , Frequência do Gene , Predisposição Genética para Doença , Genótipo , Glioma/etiologia , Glioma/mortalidade , Prognóstico , Análise de Sobrevida
9.
Genet. mol. res. (Online) ; 7(2): 451-459, 2008. tab, ilus
Artigo em Inglês | LILACS | ID: lil-640993

RESUMO

The cancer is one of the most common and severe problems in clinical medicine, and nervous system tumors represent about 2% of the types of cancer. The central role of the nervous system in the maintenance of vital activities and the functional consequences of the loss of neurons can explain how severe brain cancers are. The cell cycle is a highly complex process, with a wide number of regulatory proteins involved, and such proteins can suffer alterations that transform normal cells into malignant ones. The INK4 family members (CDK inhibitors) are the cell cycle regulators that block the progression of the cycle through the R point, causing an arrest in G1 stage. The p14ARF (alternative reading frame) gene is a tumor suppressor that inhibits p53 degradation during the progression of the cell cycle. The PTEN gene is related to the induction of growth suppression through cell cycle arrest, to apoptosis and to the inhibition of cell adhesion and migration. The purpose of the present study was to assess the mutational state of the genes p14ARF, p15INK4b, p16INK4a, and PTEN in 64 human nervous system tumor samples. Homozygous deletions were found in exon 2 of the p15INK4b gene and exon 3 of the p16INK4a gene in two schwannomas. Three samples showed a guanine deletion (63 codon) which led to a loss of heterozygosity in the p15 gene, and no alterations could be seen in the PTEN gene. Although the group of patients was heterogeneous, our results are in accordance with other different studies that indicate that homozygous deletion and loss of heterozygosity in the INK4 family members are frequently observed in nervous system tumors.


Assuntos
Humanos , /genética , /genética , Neoplasias do Sistema Nervoso/genética , /genética , Análise Mutacional de DNA/métodos , Deleção de Genes , Homozigoto , Perda de Heterozigosidade , Neoplasias do Sistema Nervoso/patologia , Reação em Cadeia da Polimerase , PTEN Fosfo-Hidrolase
10.
Genet Mol Res ; 6(4): 1019-25, 2007 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-18273794

RESUMO

Gliomas are the most common tumors of the central nervous system. In spite of the marked advances in the characterization of the molecular pathogenesis of gliomas, these tumors remain incurable and, in most of the cases, resistant to treatments, due to their molecular heterogeneity. Gene PAX6, which encodes a transcription factor that plays an important role in the development of the central nervous system, was recently recognized as a tumor suppressor in gliomas. The objective of the present study was to analyze the mutational status of the coding and regulating regions of PAX6 in 94 gliomas: 81 astrocytomas (11 grade I, 23 grade II, 8 grade III, and 39 grade IV glioblastomas), 5 oligodendrogliomas (3 grade II, and 2 grade III), and 8 ependymomas (5 grade II, and 3 grade III). Two regulating regions (SX250 and EIE) and the 11 coding regions (exons 4-13, plus exon 5a resulting from alternative splicing) of gene PAX6 were analyzed and no mutation was found. Therefore, we conclude that the tumor suppressor role of PAX6, reported in previous studies on gliomas, is not due to mutation in its coding and regulating regions, suggesting the involvement of epigenetic mechanisms in the silencing of PAX6 in these tumors.


Assuntos
Neoplasias do Sistema Nervoso Central/genética , Proteínas do Olho/genética , Glioma/genética , Proteínas de Homeodomínio/genética , Mutação , Fatores de Transcrição Box Pareados/genética , Proteínas Repressoras/genética , Adolescente , Adulto , Idoso , Astrocitoma/genética , Sequência de Bases , Criança , Pré-Escolar , Análise Mutacional de DNA , Primers do DNA/genética , DNA de Neoplasias/genética , Ependimoma/genética , Epigênese Genética , Feminino , Inativação Gênica , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Oligodendroglioma/genética , Fator de Transcrição PAX6 , Reação em Cadeia da Polimerase
11.
Genet. mol. res. (Online) ; 6(4): 1019-1025, 2007. tab
Artigo em Inglês | LILACS | ID: lil-520048

RESUMO

Gliomas are the most common tumors of the central nervous system. In spite of the marked advances in the characterization of the molecular pathogenesis of gliomas, these tumors remain incurable and, in most of the cases, resistant to treatments, due to their molecular heterogeneity. Gene PAX6, which encodes a transcription factor that plays an important role in the development of the central nervous system, was recently recognized as a tumor suppressor in gliomas. The objective of the present study was to analyze the mutational status of the coding and regulating regions of PAX6 in 94 gliomas: 81 astrocytomas (11 grade I, 23 grade II, 8 grade III, and 39 grade IV glioblastomas), 5 oligodendrogliomas (3 grade II, and 2 grade III), and 8 ependymomas (5 grade II, and 3 grade III). Two regulating regions (SX250 and EIE) and the 11 coding regions (exons 4-13, plus exon 5a resulting from alternative splicing) of gene PAX6 were analyzed and no mutation was found. Therefore, we conclude that the tumor suppressor role of PAX6, reported in previous studies on gliomas, is not due to mutation in its coding and regulating regions, suggesting the involvement of epigenetic mechanisms in the silencing of PAX6 in these tumors.


Assuntos
Humanos , Masculino , Feminino , Pré-Escolar , Criança , Adolescente , Adulto , Pessoa de Meia-Idade , DNA de Neoplasias/genética , Glioma/genética , Mutação , Neoplasias do Sistema Nervoso Central/genética , Proteínas Repressoras/genética , Proteínas de Homeodomínio/genética , Proteínas do Olho/genética , Astrocitoma , Sequência de Bases , Análise Mutacional de DNA , Epigênese Genética , Ependimoma/genética , Fatores de Transcrição Box Pareados/genética , Inativação Gênica , Oligodendroglioma/genética , Reação em Cadeia da Polimerase , Primers do DNA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...