Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Cancer Genet Cytogenet ; 164(1): 39-43, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16364761

RESUMEN

We have studied EGFR gene amplification and allelic status of chromosome 7 in 68 tumors consisting of 34 WHO grade IV glioblastomas (26 primary and 8 secondary), 14 WHO grade III anaplastic astrocytomas, and 20 WHO grade II astrocytomas, by polymerase chain reaction-single-strand conformation polymorphism (PCR-SSCP), quantitative PCR, and microsatellite analysis. EGFR gene amplification was present in 27 of these tumors (40%), and we identified allelic losses at 7p11 approximately p14 in 38 of the 68 cases (56%), including 17 tumors displaying loss for EGFR intragenic markers. The positive correlation (P < 0.05, chi(2)) between tumors with EGFR intragenic loss and EGFR gene amplification, frequently displaying the EGFR vIII form, suggests that EGFR gene rearrangement leading to intragenic loss is a molecular event that participates in the amplification process of this gene.


Asunto(s)
Astrocitoma/genética , Amplificación de Genes , Genes erbB-1 , Pérdida de Heterocigocidad , Humanos
2.
Oncol Rep ; 15(2): 443-8, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16391867

RESUMEN

The epigenetic changes in pituitary adenomas were identified by evaluating the methylation status of nine genes (RB1, p14(ARF), p16(INK4a), p73, TIMP-3, MGMT, DAPK, THBS1 and caspase-8) in a series of 35 tumours using methylation-specific PCR analysis plus sequencing. The series included non-functional adenomas (n=23), prolactinomas (n=6), prolactinoma plus thyroid-stimulating hormone adenoma (n=1), growth hormone adenomas (n=4), and adrenocorticotropic adenoma (n=1). All of the tumours had methylation of at least one of these genes and 40% of samples (14 of 35) displayed concurrent methylation of at least three genes. The frequencies of aberrant methylation were: 20% for RB1, 17% for p14(ARF), 34% for p16(INK4a), 29% for p73, 11% for TIMP-3, 23% for MGMT, 6% for DAPK, 43% for THBS1 and 54% for caspase-8. No aberrant methylation was observed in two non-malignant pituitary samples from healthy controls. Although some differences in the frequency of gene methylation between functional and non-functional adenomas were detected, these differences did not reach statistical significance. Our results suggest that promoter methylation is a frequent event in pituitary adenoma tumourigenesis, a process in which inactivation of apoptosis-related genes (DAPK, caspase-8) might play a key role.


Asunto(s)
Adenoma/genética , Caspasas/metabolismo , Islas de CpG/genética , Metilación de ADN , Neoplasias Hipofisarias/genética , Regiones Promotoras Genéticas/genética , Adulto , Factores de Edad , Anciano , Caspasa 8 , Epigénesis Genética , Femenino , Genes Relacionados con las Neoplasias/genética , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Factores Sexuales
3.
Otol Neurotol ; 27(8): 1180-5, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16983315

RESUMEN

HYPOTHESIS: The purpose of this study was to examine the DNA methylation profile of several genes in a series of vestibular schwannomas, and to analyze its relationship with clinical and radiological features. BACKGROUND: Aberrant methylation of promoter regions is a major mechanism for silencing of tumor suppressor genes in several tumors. There is limited information about methylation status in vestibular schwannoma, with no clinical or radiological implications described to date. METHODS: The methylation status of 16 tumor-related genes including RASSF1A, RAR-B, VHL, PTEN, HMLH1, RB1, TP16, CASP8, ER, TIMP3, MGMT, DAPK, TP73, GSTP1, TP14, and THBS1 was examined in a series of 22 vestibular schwannomas.The bisulfite modification of genomic DNA was performed. Clinical and radiological features were compared with the methylation results. RESULTS: Methylation values from 9% to 27% were found in 12 of 16 genes tested, including RASSF1A, VHL, PTEN, TP16, CASP8, TIMP3, MGMT, DAPK, THBS1, HMLH1, TP73, and GSTP1. A significant association was found between CASP8 and RASSF1A methylation. Methylation of CASP8 was associated with the patient's age and the tumor size. Methylation of TP73 was associated with hearing loss. RASSF1A methylation was inversely correlated with the clinical growth index. CONCLUSION: Aberrant methylation of tumor-related genes may play a role in the development of vestibular schwannomas. Our results may provide useful clues to the development of prognostic assays for these tumors.


Asunto(s)
Metilación de ADN , Neuroma Acústico/genética , Adolescente , Adulto , Anciano , Umbral Auditivo/fisiología , Femenino , Genes Supresores de Tumor/fisiología , Pérdida Auditiva/genética , Humanos , Masculino , Persona de Mediana Edad , Neuroma Acústico/diagnóstico por imagen , Neuroma Acústico/fisiopatología , Regiones Promotoras Genéticas/genética , Radiografía
4.
Am J Clin Pathol ; 123(6): 900-6, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15899783

RESUMEN

Proto-oncogene amplification is an important alteration that is present in about 45% to 50% of high-grade human gliomas. We studied this mechanism in 8 genes (cyclin-dependent kinase-4 [CDK4], MDM2, MDM4, renin-angiotensin system-1, ELF3, GAC1, human epidermal growth factor receptor-2, and platelet-derived growth factor receptor-A gene) in a series of 40 oligodendrogliomas (World Health Organization (WHO) grade II, 21; WHO grade III, 13; and WHO grade II-III oligoastrocytomas, 6) using real-time quantitative polymerase chain reaction. Amplification of at least 1 of these genes was detected in 58% of samples (23/40). By histopathologic grade, 67% of grade II oligodendrogliomas (14/21), 46% of grade III anaplastic oligodendrogliomas (6/13), and 50% of mixed oligoastrocytomas (3/6) were positive for amplification of at least 1 gene. CDK4, MDM2, and GAC1 were the most frequently involved genes (12/40 [30%], 12/40 [30%], and 13/40 [33%], respectively). Our findings demonstrate gene amplification in low-grade samples indicating that it is an important alteration in the early steps of oligodendroglioma development and, therefore, might be considered a molecular mechanism leading to malignant progression toward anaplastic forms.


Asunto(s)
Neoplasias Encefálicas/genética , Amplificación de Genes , Dosificación de Gen , Oligodendroglioma/genética , Proto-Oncogenes/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Biomarcadores de Tumor/análisis , Humanos , Proto-Oncogenes Mas
5.
Diagn Mol Pathol ; 14(4): 224-9, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16319692

RESUMEN

We have studied gene amplification of genes located in 1q32 (GAC1, ELF3, MDM4, and ren1), 4q11 (PDGFR-alpha), and in 12q13-14 (MDM2 and CDK4) using quantitative real-time PCR in a group of 86 tumors consisting of 44 WHO grade IV glioblastomas (GBM) (34 primary and 10 secondary tumors), 21 WHO grade III anaplastic astrocytomas (AA), and 21 WHO grade II astrocytomas (AII). Gene amplification was present in 56 of the 86 samples (65%) in at least 1 gene in our series. GAC1 (51%) and MDM4 (27%) were the most frequently amplified genes within the 1q32 amplicon, and their higher amplification frequency was statistically significant (P<0.05, chi) in the low-grade astrocytomas. Concordant co-amplification was determined for ELF3 and ren1 or ren1 and MDM4 in the grade III-IV tumors. MDM2 amplification was significantly more frequent in primary GBM (16%) than was in secondary GBM (0%). The present study shows that gene amplification in the studied regions is already present in low-grade astrocytic tumors and that amplification of some genes may represent another molecular marker to differentiate primary from secondary GBM.


Asunto(s)
Astrocitoma/genética , Amplificación de Genes , Dosificación de Gen , Proto-Oncogenes/genética , Biomarcadores de Tumor/análisis , Humanos , Reacción en Cadena de la Polimerasa
6.
Cancer Genet Cytogenet ; 160(2): 169-73, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15993274

RESUMEN

Allelic losses of chromosome 22 found in oligodendrogliomas suggest that at least one tumor suppressor gene on chromosome 22 is inactivated during the multistep process of tumorigenesis in this glial tumor. INI1hSNF5 (HUGO symbol: SMARCB1), located at 22q11, encodes a component of the ATP-dependent chromatin remodeling hSWI-SNF complex; it is a tumor suppressor gene that is mutated in several malignant tumors. The PARVG gene, located at 22q13, has been found to exhibit reduced expression in some cancer lines. Both genes are thus candidate tumor suppressors, potentially involved in the pathogenesis of gliomas. We performed mutation analyses of INI1hSNF5 and PARVG in a series of 40 oligodendrogliomas, but only sequence polymorphic variations were identified. Accordingly, INI1hSNF5 and PARVG do not seem to be the tumor suppressor genes involved in oligodendroglioma development and progression.


Asunto(s)
Actinina/genética , Proteínas de Unión al ADN/genética , Oligodendroglioma/genética , Mutación Puntual/genética , Secuencia de Bases , Proteínas Cromosómicas no Histona , Análisis Mutacional de ADN , Progresión de la Enfermedad , Exones/genética , Genes Supresores de Tumor , Humanos , Intrones/genética , Polimorfismo Conformacional Retorcido-Simple , Proteína SMARCB1 , Factores de Transcripción
7.
Oncol Rep ; 13(3): 539-42, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15711769

RESUMEN

Loss of 1p heterozygosity is one of the most characteristic events in oligodendrogliomas. Several genes located in this region have been previously studied to find the target gene implicated in the development of this tumor without success. Patched-2, RIZ1 and KIF1B are novel oncosuppressor genes located at 1p and involved in different kinds of tumors. We have studied these genes and p18(ink4c) using PCR/SSCP methods to detect sequence variations in a series of 40 oligodendrogliomas in which the allelic status at 1p was analyzed. Polymorphisms or no sequence changes were detected in all four genes analyzed. None of the genes analyzed seem to be the target-gene mapped at 1p involved by mutation in oligodendroglioma development.


Asunto(s)
Neoplasias Encefálicas/genética , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Cinesinas/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Oligodendroglioma/genética , Polimorfismo Genético , Proteína de Retinoblastoma/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Neoplasias Encefálicas/fisiopatología , Transformación Celular Neoplásica , Inhibidor p18 de las Quinasas Dependientes de la Ciclina , Análisis Mutacional de ADN , N-Metiltransferasa de Histona-Lisina , Humanos , Pérdida de Heterocigocidad , Oligodendroglioma/fisiopatología , Receptores Patched , Receptor Patched-2 , Reacción en Cadena de la Polimerasa , Polimorfismo Conformacional Retorcido-Simple , Inhibidores de Proteínas Quinasas , Receptores de Superficie Celular
8.
Int J Mol Med ; 16(4): 771-4, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16142420

RESUMEN

The DAL-1/41B gene (differentially expressed in adenocarcinoma of the lung), located in the chromosome 18p11.3 region, belongs to the protein family 4.1 (membrane-associated proteins), which includes the product of the NF2 gene (merlin), and the proteins, ezrin, radixin, and moesin. DAL-1/4.1B is normally expressed at high levels in the brain, with lower levels in the kidney, intestine, and testis. DAL-1/4.1B is known to suppress growth in meningiomas and can be lost in about 60% of sporadic meningiomas as an early event in tumorigenesis; it is a critical growth regulator in the pathogenesis of neoplastic transformation. The similarity between the DAL-1/4.1B protein and merlin, with their high levels of expression in the brain and their recurrent loss in meningiomas, and the lack of previous DAL-1/4.1B mutational analysis reports initiated this mutational study of DAL-1/4.1B in a series of 83 meningiomas. We found the following sequence variations; Ala555Thr (G1663A in exon 13) and Thr950Lys (C2849A in exon 19) in two cases each, and one case with a 5pb deletion (del taaaa) in intron 18. A polymorphism in exon 14 (C2112T/Thr704Thr, also known as C2166T) was also identified; the tumoral allelic constitutions were heterozygous C/T in 15, homo- or hemizygous C in 67 and hemizygous T in one tumour. The low mutational frequency in our study discounts sequence variations in DAL-1/4.1B as the main mechanism underlying participation of this gene in the neoplastic transformation of meningiomas, and suggests that other inactivating mechanisms, such as epigenetic changes, may participate in DAL1/4.1B silencing.


Asunto(s)
Proteínas de la Membrana/genética , Neoplasias Meníngeas/genética , Meningioma/genética , Mutación , Proteínas Supresoras de Tumor/genética , Alelos , Secuencia de Bases , Análisis Mutacional de ADN , ADN de Neoplasias/química , ADN de Neoplasias/genética , Frecuencia de los Genes , Variación Genética , Genotipo , Humanos , Proteínas de Microfilamentos , Mutación Missense , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Polimorfismo Conformacional Retorcido-Simple , Eliminación de Secuencia
9.
Clin Cancer Res ; 9(15): 5601-6, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-14654541

RESUMEN

PURPOSE: The purpose of this research was to examine the DNA methylation profile of schwannomas. EXPERIMENTAL DESIGN: We examined the DNA methylation status of 12 tumor-related genes (NF2, RB1, p14(ARF), p16(INK4a), p73, TIMP-3, MGMT, DAPK, THBS1, caspase-8, TP53, and GSTP1) in 44 sporadic and/or NF2-associated schwannomas using methylation-specific PCR. RESULTS: The most frequently methylated genes were THBS1 (36%), p73 (27%), MGMT (20%), NF2 (18%), and TIMP-3 (18%). The RB1/p16INK4a gene pair displayed aberrant methylayed alleles in 15% of cases, whereas methylation was relatively rare in the other genes (<5%). Methylation was tumor specific because it was absent in two nonneoplastic nerve sheath samples and two nonneoplastic brain samples studied as controls. CONCLUSIONS: Our findings indicate that aberrant methylation seems to be a mechanism for NF2 gene inactivation, considered an early step in schwannoma tumorigenesis, and as well, aberrant hypermethylation of other tumor-related genes might represent secondary events that also contribute to the development of these tumors.


Asunto(s)
Metilación de ADN , Fosfatos de Dinucleósidos/metabolismo , Neurilemoma/genética , Neurofibromatosis 2/genética , Adulto , Anciano , Femenino , Genes Relacionados con las Neoplasias/genética , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa
10.
Int J Oncol ; 22(3): 601-8, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12579314

RESUMEN

Promoter hypermethylation represents a primary mechanism in the inactivation of tumor suppressor genes during tumorigenesis. To determine the frequency and timing of hypermethylation during carcinogenesis of astrocytic tumors, we analysed promoter methylation status of ten tumor-associated genes (MGMT, GSTP1, DAPK, p14ARF, THBS1, TIMP-3, p73, p16INK4A, RB1 and TP53) in a series of 88 astrocytic gliomas, including 24 diffuse astrocytomas; 21 anaplastic astrocytomas, and 43 glioblastomas (33 primary and 10 secondary), as well as two non-neoplastic brain samples, by methylation-specific PCR. Aberrant CpG island methylation was detected in all ten genes analysed, and all but one sample displayed anomalies in at least one gene. The methylation index (number methylated genes/total genes analysed) was 0.3, 0.38, 0.33 and 0.29 for diffuse astrocytomas, anaplastic astrocytomas and secondary and primary glioblastomas, respectively. Some differences may be established regarding the methylation profiles of specific genes and tumor types: MGMT, THBS1, TIMP-3, and p16INK4A appear hypermethylated in low-grade tumors (at least in 45% of cases), whereas GSTP1, DAPK, and p14ARF are mostly changed in 15-50% of the higher grade forms versus <10% in low-grade tumors. Some variation also exists regarding the methylation values for p73 and RB1 (10-40% of cases) among all groups. TP53 presented hypermethylation rates <10% in all tumor subtypes. Our findings thus suggest that methylation represents a common mechanism that contributes to inactivating cancer-related genes in astrocytic neoplasms. This epigenetic change is, in general, an early event in the development of astrocytic neoplasms but this gene silencing mechanism may also appear as a late event involving some loci.


Asunto(s)
Astrocitoma/metabolismo , Neoplasias Encefálicas/metabolismo , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Glioblastoma/metabolismo , Regiones Promotoras Genéticas/genética , Adulto , Anciano , Astrocitoma/patología , Neoplasias Encefálicas/patología , Islas de CpG , ADN de Neoplasias/genética , Progresión de la Enfermedad , Femenino , Glioblastoma/patología , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Reacción en Cadena de la Polimerasa
11.
Int J Oncol ; 25(5): 1489-94, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15492843

RESUMEN

Primarily involved in cell proliferation and differentiation processes, the plasma membrane-bound ErbB tyrosine kinase receptor family is formed by four members: erbB1/EGFR, erbB2/HER2/Neu, erbB3/HER3 and erbB4/HER4. Calmodulin (CaM) is a Ca2+-binding protein involved in the regulation of multiple intracellular processes that binds directly to EGFR in the presence of Ca2+, inhibiting its tyrosine kinase activity. Two main regions in the receptor have been implicated in this relationship: the calmodulin-binding domain (CaM-BD) and the calmodulin-like domain (CaM-LD); their sequences are highly conserved in other members of this family of receptors. The presence of mutations, amplification and/or overexpression and genomic rearrangement of these domains was investigated for all four erbB family genes in a series of 89 glial tumors, including 44 WHO grade IV glioblastomas, 21 WHO grade III anaplastic astrocytomas, and 24 WHO grade II astrocytomas. Gene alterations were only found in the regions of interest in EGFR. One glioblastoma showed an in frame tandem duplication of the intracellular region including CaM-LD (exons 18-25). CaM-BD gene overdose was evidenced in 18 tumors that showed EGFR amplification in other domains. Over-expression of CaM-BD and CaM-LD was detected in 6 and 17 cases, respectively, of the 19 tumors in which this study was performed. The other three genes coding for the ErbB receptors did not present point mutations, or rearrangements, and only a very low amplification rate was found for erbB2 (1 case) and erbB3 (4 cases). No overexpression of erbB2, erbB3 or erbB4 was detected. These findings suggest that EGFR is the main erbB gene family member non-randomly involved in malignant glioma development, and that the two domains under study, due to their high conservation and wide separation in the EGFR sequence, are good marker regions for evaluating EGFR/erbB1 gene amplification, as well as for analysing the presence of transcripts corresponding to truncated cytosolic forms of the receptor in these tumors.


Asunto(s)
Astrocitoma/genética , Neoplasias Encefálicas/genética , Proteínas de Unión a Calmodulina/genética , Amplificación de Genes , Genes erbB/genética , Glioblastoma/genética , Astrocitoma/patología , Biopsia , Neoplasias Encefálicas/patología , Proteínas de Unión a Calmodulina/farmacología , Transformación Celular Neoplásica , Análisis Mutacional de ADN , Reordenamiento Génico , Glioblastoma/patología , Humanos
12.
Hum Pathol ; 33(3): 375-8, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11979381

RESUMEN

Multiple meningiomas are rare, and only 13 cases have been subjected to molecular genetic analysis to detect mutations of the tumor-suppressor gene neurofibromatosis type 2 (NF2) located on chromosome 22. Most of these cases display NF2 gene mutations parallel to loss of the chromosome 22 homolog, indicating that inactivation of this gene may represent an early event in the development of multiple meningiomas. We report a case of a 61-year-old woman who developed multiple (dorsal and intracranial) meningiomas. Cytogenetic and molecular genetic studies demonstrated the loss of a copy of chromosome 22 in the 5 meningiomas studied and the absence of NF2 gene mutations in 4 of those available for this molecular analysis. These findings, together with similar data from 2 previously reported cases, suggest the participation of a tumor-suppressor gene other than NF2 on chromosome 22 in the pathogenesis of a subgroup of multiple meningiomas.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 22 , Genes de la Neurofibromatosis 2 , Neoplasias Meníngeas/genética , Meningioma/genética , Femenino , Humanos , Neoplasias Meníngeas/patología , Meningioma/patología , Persona de Mediana Edad , Mutación
13.
Cancer Genet Cytogenet ; 134(1): 1-5, 2002 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11996787

RESUMEN

Allelic losses of chromosome 22 are commonly found in ependymomas and oligodendrogliomas, suggesting that at least one tumor suppressor gene on chromosome 22 must be inactivated during the multistep process of tumorigenesis in these glial tumors. The neurofibromatosis 2 gene (NF2) located at 22q12, is a candidate tumor suppressor gene potentially involved in the pathogenesis of gliomas. Because there have been only a few studies of the NF2 gene in glial tumors other than astrocytoma, we screened the entire 17 NF2 exons for mutations in a series of 47 nonastrocytic tumors, including 40 oligodendrogliomas and 7 ependymomas. Only one mutation was detected, a 59-base pair insertion in exon 3 from a spinal anaplastic ependymoma. These results concur with previous findings proposing preferential inactivation of the NF2 gene in a subgroup of ependymomas, and suggest that the NF2 gene is not the target of chromosome 22 aberrations in oligodendrogliomas.


Asunto(s)
Neoplasias Encefálicas/genética , Ependimoma/genética , Neurofibromina 2/genética , Oligodendroglioma/genética , Adulto , Neoplasias Encefálicas/patología , Análisis Mutacional de ADN , ADN de Neoplasias/química , ADN de Neoplasias/genética , Ependimoma/patología , Exones/genética , Humanos , Persona de Mediana Edad , Mutagénesis Insercional , Mutación , Oligodendroglioma/patología , Polimorfismo Conformacional Retorcido-Simple
14.
Cancer Genet Cytogenet ; 144(2): 134-42, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12850376

RESUMEN

Promoter hypermethylation represents a primary mechanism in the inactivation of tumor suppressor genes during tumorigenesis. To determine the frequency and timing of hypermethylation during carcinogenesis of nonastrocytic tumors, we analyzed promoter methylation status of 10 tumor-associated genes in a series of 41 oligodendrogliomas (22 World Health Organization [WHO] grade II; 13 WHO grade III; 6 WHO grade II-III oligoastrocytomas) and 7 WHO grade II-III ependymomas, as well as 2 nonneoplastic brain samples, by a methylation-specific polymerase chain reaction. Aberrant CpG island methylation was detected in 9 of 10 genes analyzed, and all but one sample displayed anomalies in at least one gene. The frequencies of hypermethylation for the 10 genes were as follows, in oligodendrogliomas and ependymomas, respectively: 80% and 28% for MGMT; 70% and 28% for GSTP1; 66% and 57% for DAPK; 44% and 28% for TP14(ARF); 39% and 0% for THBS1; 24% and 28% for TIMP3; 24% and 14% for TP73; 22% and 0% for TP16(INK4A); 3% and 14% for RB1; and 0% in both neoplasms for TP53. No methylation of these genes was detected in normal brain tissue samples. We conclude that a high frequency of aberrant methylation of the 5' CpG island of the MGMT, GSTP1, TP14(ARF), THBS1, TIMP3, and TP73 genes is observed in nonastrocytic neoplasms. This aberration seems to occur early in the carcinogenesis process (it is already present in the low-grade forms), although in some instances (DAPK, THBS1, and TP73) it appears also associated with the genesis of anaplastic forms.


Asunto(s)
Metilación de ADN , Ependimoma/genética , Oligodendroglioma/genética , Regiones Promotoras Genéticas , Adulto , Islas de CpG , Femenino , Genes p16 , Genes p53 , Humanos , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , O(6)-Metilguanina-ADN Metiltransferasa/genética , Proteína p14ARF Supresora de Tumor/genética
15.
Cancer Genet Cytogenet ; 142(1): 21-4, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12660028

RESUMEN

We have determined the promoter CpG island methylation status of O(6)-methylguanine-DNA methyltransferase (MGMT), glutathione-S-transferase P1 (GSTP1), death-associated protein kinase (DAPK), p14(ARF), thrombospondin-1 (THBS1), tissue inhibitor of metalloproteinase-3 gene (TIMP-3), p73, p16(INK4A), RB1, and TP53 genes in three primary central nervous system lymphomas (PCNSL). Five genes (GSTP1, DAPK, TIMP-3, p16(INK4A), and RB1) were hypermethylated in two samples, whereas MGMT, THBS1, and p73 were aberrantly methylated in only one sample. No case presented CpG island methylation for the p14(ARF) and TP53 genes. These findings concur with previous data suggesting a frequent inactivation of p16(INK4A) and very limited involvement of TP53 in PCNSL and also provide insights into the epigenetic molecular involvement of other tumor-related genes in this neoplasm.


Asunto(s)
Neoplasias del Sistema Nervioso Central/genética , Islas de CpG , Metilación de ADN , Linfoma/genética , Anciano , Proteínas Reguladoras de la Apoptosis , Encéfalo/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteínas de Unión al ADN/genética , Proteínas Quinasas Asociadas a Muerte Celular , Femenino , Genes Supresores de Tumor , Genes p53 , Gutatión-S-Transferasa pi , Glutatión Transferasa/genética , Humanos , Inmunocompetencia , Isoenzimas/genética , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , O(6)-Metilguanina-ADN Metiltransferasa/genética , Regiones Promotoras Genéticas , Valores de Referencia , Proteína de Retinoblastoma/genética , Trombospondina 1/genética , Inhibidor Tisular de Metaloproteinasa-3/genética , Proteína Tumoral p73 , Proteína p14ARF Supresora de Tumor/genética , Proteínas Supresoras de Tumor
16.
Oncol Rep ; 9(5): 951-4, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12168053

RESUMEN

The retinoblastoma gene family RB1, p107 and RB2/p130 cooperate to regulate cell cycle progression through the G1 phase of the cell cycle. Previous data demonstrated that RB2/p130 inhibits proliferation of the glioblastoma cell line T98G, which is resistant to the growth suppressive effects of both RB1 and p107, and that RB2/p130 gene overexpresion induces astrocyte differentiation. We screened by single-strand conformation polymorphism and sequence analysis the structure of exons 19 through 22 of the RB2/p130 gene, which encodes the B domain and C terminus, in a series of 42 glioblastomas (32 primary and 10 secondary). Sequence variations were identified in one tumor, suggesting that mutation inactivation of RB2/p130 is a rare event in glioblastoma.


Asunto(s)
Glioblastoma/genética , Mutación , Fosfoproteínas/genética , Proteínas , Astrocitos/citología , Secuencia de Bases , Ciclo Celular , Diferenciación Celular , División Celular , Análisis Mutacional de ADN , Exones , Fase G1 , Humanos , Intrones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Polimorfismo Conformacional Retorcido-Simple , Estructura Terciaria de Proteína , Proteína p130 Similar a la del Retinoblastoma , Células Tumorales Cultivadas
17.
Oncol Rep ; 10(5): 1519-23, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12883734

RESUMEN

Aberrant methylation of the promoter CpG island of human genes is an alternative gene inactivation mechanism that contributes to the carcinogenesis of human tumours. We have determined the methylation status of the CpG island of 11 tumour-related genes (RB1, p14ARF, p16INK4a, p73, TIMP-3, MGMT, DAPK, THBS1, caspase 8, TP53 and GSTP1) in 18 neurofibromas (including one plexiform neurofibroma) and three neurofibrosarcomas, as well as two non-neoplastic peripheral nerve sheath samples, using methylation-specific polymerase chain reaction. The series included sporadic and neurofibromatosis type 1-associated tumours. The incidence of aberrant methylation in the tumour samples was 52% for THBS1, 43% for MGMT, 33% for TIMP-3, 19% each for p16INK4a and p73, 14% for RB1, 5% for p14ARF, and 0% for DAPK, caspase 8, TP53 and GSTP1. No methylation of these genes was detected in the two samples of non-neoplastic peripheral nerve sheath. All but three samples in the study displayed aberrant methylation in at least one of the studied genes, and there was no correlation between methylation status and the patients' clinical parameters. These findings suggest that methylation of some tumour-related genes may play a significant role in the tumourigenesis of neurofibromas/neurofibrosarcomas.


Asunto(s)
Neoplasias del Sistema Nervioso Central/genética , Islas de CpG , Metilación de ADN , Neurofibroma/genética , Neurofibrosarcoma/genética , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa
18.
Oncol Rep ; 12(3): 663-6, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15289853

RESUMEN

Aberrant methylation of promoter CpG islands in human genes is an alternative genetic inactivation mechanism that contributes to the development of human tumors. Nevertheless, few studies have analyzed methylation in medulloblastomas. We determined the frequency of aberrant CpG island methylation for Caspase 8 (CASP8) in a group of 24 medulloblastomas arising in 8 adult and 16 pediatric patients. Complete methylation of CASP8 was found in 15 tumors (62%) and one case displayed hemimethylation. Three samples amplified neither of the two primer sets for methylated or unmethylated alleles, suggesting that genomic deletion occurred in the 5' flanking region of CASP8. Our findings suggest that methylation commonly contributes to CASP8 silencing in medulloblastomas and that homozygous deletion or severe sequence changes involving the promoter region may be another mechanism leading to CASP8 inactivation in this neoplasm.


Asunto(s)
Neoplasias Encefálicas/genética , Caspasas/biosíntesis , Caspasas/genética , Islas de CpG , Metilación de ADN , Meduloblastoma/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Secuencia de Bases , Neoplasias Encefálicas/metabolismo , Caspasa 8 , Línea Celular Tumoral , Niño , Preescolar , ADN/metabolismo , Cartilla de ADN/química , Cartilla de ADN/farmacología , Femenino , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Homocigoto , Humanos , Masculino , Meduloblastoma/metabolismo , Persona de Mediana Edad , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Homología de Secuencia de Ácido Nucleico , Transcripción Genética
19.
Oncol Rep ; 10(4): 1031-3, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12792765

RESUMEN

Death-associated protein (DAP) kinase is a gene that participates in apoptosis induced by interferon gamma. It appears to play a role in lung cancer metastasis in animal models, suggesting that DAP-kinase may function as a metastasis suppressor by inducing apoptosis. Expression silencing through CpG island methylation of DAP-kinase has been frequently found in connection with adverse survival, as cells lacking DAP-kinase appear to be more invasive and more metastatic in lung cancer. The purpose of this study was to analyze the promoter methylation status of DAP-kinase gene in brain metastases of solid tumors. Methylation-specific PCR was performed on ten brain metastasis samples derived from malignant melanoma (three cases), lung cancer (two), breast carcinoma (two), ovarian carcinoma (two) and colon carcinoma (one case), and in corresponding peripheral blood DNA samples. Two normal brain tissue samples were also analyzed, no promoter hypermethylation was observed in either case. DAP-kinase hypermethylated alleles were identified in nine metastases (90%), and in peripheral blood lymphocytes DNA from four cases. Our data suggest that silencing of DAP-kinase through promoter hypermethylation is a common event in the multistep process of tumor metastasis, including brain involvement.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundario , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Metilación de ADN , Regiones Promotoras Genéticas , Adulto , Proteínas Reguladoras de la Apoptosis , Neoplasias Encefálicas/enzimología , Neoplasias de la Mama/sangre , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Proteínas Quinasas Dependientes de Calcio-Calmodulina/sangre , Neoplasias del Colon/sangre , Neoplasias del Colon/enzimología , Neoplasias del Colon/genética , ADN de Neoplasias/genética , Proteínas Quinasas Asociadas a Muerte Celular , Femenino , Humanos , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Masculino , Melanoma/sangre , Melanoma/enzimología , Melanoma/genética , Persona de Mediana Edad , Neoplasias Ováricas/sangre , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/genética , Reacción en Cadena de la Polimerasa , Sulfitos
20.
Int J Mol Med ; 13(1): 93-8, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14654977

RESUMEN

The aberrant methylation of the CpG island promoter regions acquired by tumor cells is one mechanism for loss of gene function. The high methylation rate for RB1 and death-associated protein-kinase gene (DAP-kinase) (60 and 90%, respectively) previously found in brain metastases suggests this mechanism could be non-randomly associated to tumor progression and metastasis. Thus, in addition to these two genes, we determined the methylation status of the genes p16INK4a, glutathione S-transferase P1 (GSTP1), O6-methylguanine DNA methyltransferase (MGMT), thrombospondin-1 (THBS1), p14ARF, TP53, p73, and tissue inhibitor of metalloproteinase 3 (TIMP-3), in 18 brain metastases of solid tumors, with methylation specific PCR. The metastases were derived from malignant melanoma (three cases), lung carcinoma (six cases), breast carcinoma (three cases), ovarian carcinoma (two cases) and one each from colon, kidney, bladder and undifferentiated carcinoma. We detected methylation levels in the tumor samples of 83% in p16INK4a, 72% in DAP-kinase, 56% in THBS1, 50% in RB1, 39% in MGMT, 33% in GSTP1 and p14ARF each, 22% in p73 and TIMP-3 each, and 11% in TP53. The methylation index (number of genes methylated/number of genes tested) varied between 0.1 and 0.6, with an average of 0.42, indicating that a high grade of gene methylation accumulates parallel to the tumor metastasis process. Our data suggest an important role for gene methylation in the development of brain metastases, primarily involving epigenetic silencing of DAP-kinase, THBS1 and the cell-cycle regulators RB1/p16INK4a.


Asunto(s)
Neoplasias Encefálicas/genética , Metilación de ADN , Regiones Promotoras Genéticas , Adulto , Anciano , Neoplasias Encefálicas/secundario , Femenino , Gutatión-S-Transferasa pi , Glutatión Transferasa/genética , Humanos , Isoenzimas/genética , Masculino , Persona de Mediana Edad , Inhibidor Tisular de Metaloproteinasa-3/genética , Proteína p14ARF Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA