Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Genes Chromosomes Cancer ; 54(4): 197-209, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25533176

RESUMEN

Schwannomas are tumors that develop from Schwann cells in the peripheral nerves and commonly arise from the vestibular nerve. Vestibular schwannomas can present unilaterally and sporadically or bilaterally when the tumor is associated with neurofibromatosis Type 2 (NF2) syndrome. The molecular hallmark of the disease is biallelic inactivation of the NF2 gene. The epigenetic signature of schwannomas remains poorly understood and is mostly limited to DNA methylation of the NF2 gene, whose altered expression due to epigenetic factors in this tumor is controversial. In this study, we tested the genomewide DNA methylation pattern of schwannomas to shed light on this epigenetic alteration in these particular tumors. The methodology used includes Infinium Human Methylation 450K BeadChip microarrays in a series of 36 vestibular schwannomas, 4 nonvestibular schwannomas, and 5 healthy nerves. Our results show a trend toward hypomethylation in schwannomas. Furthermore, homeobox (HOX) genes, located at four clusters in the genome, displayed hypomethylation in several CpG sites in the vestibular schwannomas but not in the nonvestibular schwannomas. Several microRNA (miRNA) and protein-coding genes were also found to be hypomethylated at promoter regions and were confirmed as upregulated by expression analysis; including miRNA-21, Met Proto-Oncogene (MET), and PMEPA1. We also detected methylation patterns that might be involved in alternative transcripts of several genes such as NRXN1 or MBP, which would increase the complexity of the methylation and expression patterns. Overall, our results show specific epigenetic signatures in several coding genes and miRNAs that could potentially be used as therapeutic targets.


Asunto(s)
Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Genes Homeobox , Neuroma Acústico/genética , Empalme Alternativo , Femenino , Genoma Humano , Humanos , Masculino , MicroARNs/metabolismo , Familia de Multigenes , Proto-Oncogenes Mas
2.
Int J Oncol ; 34(2): 493-504, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19148485

RESUMEN

Microarray gene expression profiling is a high-throughput system used to identify differentially expressed genes and regulation patterns, and to discover new tumor markers. As the molecular pathogenesis of meningiomas and schwannomas, characterized by NF2 gene alterations, remains unclear and suitable molecular targets need to be identified, we used low density cDNA microarrays to establish expression patterns of 96 cancer-related genes on 23 schwannomas, 42 meningiomas and 3 normal cerebral meninges. We also performed a mutational analysis of the NF2 gene (PCR, dHPLC, Sequencing and MLPA), a search for 22q LOH and an analysis of gene silencing by promoter hypermethylation (MS-MLPA). Results showed a high frequency of NF2 gene mutations (40%), increased 22q LOH as aggressiveness increased, frequent losses and gains by MLPA in benign meningiomas, and gene expression silencing by hypermethylation. Array analysis showed decreased expression of 7 genes in meningiomas. Unsupervised analyses identified 2 molecular subgroups for both meningiomas and schwannomas showing 38 and 20 differentially expressed genes, respectively, and 19 genes differentially expressed between the two tumor types. These findings provide a molecular subgroup classification for meningiomas and schwannomas with possible implications for clinical practice.


Asunto(s)
Neoplasias Meníngeas/genética , Meningioma/genética , Neurilemoma/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Adulto , Anciano , ADN Complementario/genética , ADN de Neoplasias/genética , Femenino , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias Meníngeas/clasificación , Meningioma/clasificación , Repeticiones de Microsatélite/genética , Persona de Mediana Edad , Neurilemoma/clasificación , Neurofibromatosis 2/genética
3.
Acta Otorrinolaringol Esp ; 60(1): 9-18, 2009.
Artículo en Español | MEDLINE | ID: mdl-19268124

RESUMEN

BACKGROUND AND OBJECTIVES: The lesions that involve the paranasal sinuses and the anterior cranial base at the same time are not unusual. These diseases have different features. The aim of this study is to set out the particularities of the non-malignant lesions involving both zones. MATERIAL AND METHODS: Retrospective study of 32 patients between 1986 and 2007 diagnosed with: non-malignant tumours (31.2 %), tumorlike lesions (3.1 %), fibrous-osseous lesions (12.5 %), congenital or acquired malformations (18.7 %) and infection disease (34.3 %). We analyse the diagnostic imaging, the treatment and pathogen mechanism. RESULTS: Only 6 of 43 osteomas involved the paranasal sinuses and anterior cranial fossa (13.04 %): 3 cases have developed meningitis and 1 developed a pneumocephalus. 2 cases are meningiomas: 1 was asymptomatic and the other one caused destruction at subtotal frontal bone. 1 giant hemangioma associated with Klippel-Trenaunay syndrome is treated by combined craniofacial approach. The fibrous-osseous lesions were specifically fibrous dysplasia and affected the ethmoides. The encephalocele were predominating in the malformations group, 2 were diagnosed after repeated meningitis. 11 cases are included by infection: 10 cases caused osteomielitis and the eleventh is a patient with a mucormycosis. Surgery has been used in 84.3 % of the cases: frontal craniotomy 37 %, combined craniofacial approach 18.5 %, subfrontal approach 18.5 %, osteoplastic technique 18.5 %, lateronasal approach 3.7 %, endonasal microscopic resection 3.7 %. CONCLUSIONS: In this study the diagnosis, extension and surgical management were supported in the imaging. A closed separation between the anterior cranial fossa ant the sinus is necessary after the resection. The reconstruction was performed using a pedicled pericranial flap and titanium mesh in most of the cases.


Asunto(s)
Enfermedades Óseas/diagnóstico , Enfermedades de los Senos Paranasales/diagnóstico , Base del Cráneo , Humanos
4.
Clin Endocrinol (Oxf) ; 69(6): 906-10, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18681855

RESUMEN

Head and neck paragangliomas are usually asymptomatic and benign tumours arising mainly from the carotid body and the vagal, tympanic or jugular glomus. The majority of patients develop sporadic masses, and around 30% of cases harbour germline mutations in one of the succinate dehydrogenase genes: SDHB, SDHC or SDHD. In these hereditary cases, the presence of familial antecedents of the disease, multiplicity/bilaterality, young age at onset, and more recently, presence of gastrointestinal stromal tumours, are main factors to be considered. Here we describe a new mutation (c.256-257insTTT) affecting the SDHC gene in a 60-year-old-patient with a single head and neck paraganglioma, and without familial antecedents of the disease. In silico splice site analysis showed that this variant created a cryptic splice acceptor site and loss of heterozygosity (LOH) supported the pathogenic role of the mutation. Control population analyses did not detect this variant but revealed a novel SDHC polymorphism that exhibited a frequency of 0.3% (3/1020). This latter finding highlights the importance of assessing the clinical relevance of variants of unknown significance by means of analysing sufficient controls. Despite having found a germline mutation in an older, apparently sporadic patient, we consider that the high costs of analysing all susceptibility genes related to the disease support the recommendation of screening for mutations only in patients fulfilling the above criteria.


Asunto(s)
Proteínas de la Membrana/genética , Adenoma/genética , Adulto , Femenino , Mutación de Línea Germinal , Humanos , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Paraganglioma/genética , Neoplasias Hipofisarias/genética
5.
Int J Oncol ; 30(1): 209-15, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17143531

RESUMEN

We have studied amplification/gene-dosage and sequence variations of the EGFR gene in 41 oligodendroglial tumours graded according to the WHO classification (21 oligodendrogliomas grade II, 13 oligodendrogliomas grade III and 6 oligoastrocytomas grade II-III), using multiplex ligation-dependent probe amplification (MLPA), real-time quantitative PCR, and PCR/SSCP techniques. To determine gene-dose we studied exons 11 (extracellular domain) and 25 (intracellular domain) in the EGFR gene. Overdose (1- to 5-fold increase) was present in exon 11 in 21 of 41 samples (52.5% of cases) and in exon 25, in 7 of 41 samples (17.5% of cases). Gene amplification > 5-fold increase) was present in exon 11, in 17 of 41 samples (42.5% of cases), and in exon 25 in 6 of 41 samples (15% of cases). Three tumours (two grade II oligodendrogliglioma, one mixed oligoastrocytoma) displayed high level amplifications: > 100 gene copies were identified by both real-time quantitative PCR and MLPA analyses. Gene sequence alterations were identified by PCR/SSCP and sequencing in four cases: two missense mutations: G1051A (Ala351Thr) and G2216A (Arg739Hys); one nonsense mutation: C2934T (Asp978Asp); and an 18 bp deletion in position 2423-2441 of E19. These changes were present only in tumoral DNA, not in the corresponding constitutional patients' DNA. We also found four previously unidentified polymorphic variants: G2025A (Ala675Ala), C2233T (Leu745Leu), C2895T (Treo965Treo) and C3168T (Asp1056Asp), and three previously described polymorphic changes: E12+22 Tright curved arrow A, G1748A (Arg583Lys) and A2547T (Glu849Glu). Our findings demonstrate that mutations and amplification/overdose in the EGFR gene are present in low-grade oligodendroglial tumours, and may contribute to the development of these brain neoplasms.


Asunto(s)
Neoplasias Encefálicas/genética , Análisis Mutacional de ADN , Receptores ErbB/genética , Dosificación de Gen , Oligodendroglioma/genética , Sustitución de Aminoácidos , Secuencia de Bases , Biopsia , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Cartilla de ADN , Exones , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Humanos , Oligodendroglioma/patología , Oligodendroglioma/cirugía , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Polimorfismo Conformacional Retorcido-Simple , Eliminación de Secuencia
6.
Cancer Genet Cytogenet ; 173(1): 63-7, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17284372

RESUMEN

Clinical response to Gefitinib (Iressa, ZD1839) has been found to be associated with somatic mutations, primarily of exons 18-21, of the epidermal growth factor receptor gene (EGFR) in non-small cell lung cancer (NSCLC). Evidence of a positive response was also reported recently on a patient with brain metastasis from NSCLC. On the other hand, amplification of EGFR appears to be associated with a poor prognosis. To determine whether EGFR mutations and amplification are involved in the tumorigenesis of brain metastases, we performed polymerase chain reaction/single-strand conformation polymorphism to examine exons 1, 2, and 7-26 of EGFR in a series of 18 brain metastases. The metastases 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. In addition to several sequence polymorphisms, we identified two mutations on E19 consisting of 18-base pair (bp) deletions: 2423-24440del and 2426-2443del. These mutations presented in lesions derived from kidney carcinoma and lung adenocarcinoma. By real-time quantitaive polymerase chain reaction technique, we determined the amplification/overdose status of EGFR by analyzing exons 11 and 25. Amplification (5- to 100-fold) was identified in three tumors, and overdose (low-level gene amplification corresponding to increases of 1- to 5-fold) presented in four additional metastases. These findings suggest that EGFR mutations and polymorphisms are not exclusively present in metastases derived from lung carcinoma. Accordingly, targeting of EGFR to determine molecular alterations of this gene may be useful in the management of patients with brain metastases.


Asunto(s)
Neoplasias Encefálicas/secundario , Receptores ErbB/genética , Mutación , Neoplasias/patología , Reacción en Cadena de la Polimerasa/métodos , Adulto , Anciano , Secuencia de Aminoácidos , Secuencia de Bases , Neoplasias Encefálicas/genética , Análisis Mutacional de ADN , ADN de Neoplasias/química , ADN de Neoplasias/genética , Femenino , Dosificación de Gen , Humanos , Masculino , Persona de Mediana Edad , Mutagénesis Insercional , Neoplasias/genética , Polimorfismo Genético , Polimorfismo Conformacional Retorcido-Simple , Eliminación de Secuencia
7.
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
8.
Head Neck ; 38 Suppl 1: E673-9, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-25867206

RESUMEN

BACKGROUND: Endolymphatic sac tumors (ELSTs) are, with a prevalence of up to 16%, a component of von Hippel-Lindau (VHL) disease. Data from international registries regarding heritable fraction and characteristics, germline VHL mutation frequency, and prevalence are lacking. METHODS: Systematic registration of ELSTs from international centers of otorhinolaryngology and from multidisciplinary VHL centers' registries was performed. Molecular genetic analyses of the VHL gene were offered to all patients. RESULTS: Our population-based registry comprised 93 patients with ELST and 1789 patients with VHL. The prevalence of VHL germline mutations in apparently sporadic ELSTs was 39%. The prevalence of ELSTs in patients with VHL was 3.6%. ELST was the initial manifestation in 32% of patients with VHL-ELST. CONCLUSION: Prevalence of ELST in VHL disease is much lower compared to the literature. VHL-associated ELSTs can be the first presentation of the syndrome and mimic sporadic tumors, thus emphasizing the need of molecular testing in all presentations of ELST. © 2015 Wiley Periodicals, Inc. Head Neck 38: 673-679, 2016.


Asunto(s)
Neoplasias del Oído/patología , Saco Endolinfático/patología , Enfermedad de von Hippel-Lindau/complicaciones , Adolescente , Adulto , Anciano , Niño , Femenino , Mutación de Línea Germinal , Humanos , Masculino , Persona de Mediana Edad , Sistema de Registros , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Adulto Joven
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Hum Mutat ; 23(2): 160-169, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14722919

RESUMEN

Von Hippel-Lindau (VHL) disease is a hereditary cancer syndrome caused by germline mutations in the VHL gene. This gene, located in the 3p25-26 chromosome, is a tumor suppressor gene associated with the inhibition of angiogenesis and apoptosis, cell cycle exit, fibronectin matrix assembly, and proteolysis. To define the molecular basis of VHL in a Spanish population, we studied 33 patients suspected of suffering familial or de novo VHL disease and two familial pheochromocytoma cases. Sequence analysis of the coding regions of the VHL gene revealed germline sequence variants in 68.7% (24 out of 35) of the patients, and four of them presented with undescribed germline alterations: g.5429-5430insG, p.Leu128Arg, p.Tyr175Cys, and p.Tyr175Asn. For the remaining 11 patients who showed negative for point mutations, we performed Southern blot analysis and detected gross rearrangements in eight cases (22.8% of the index cases). Our results support the relevance of VHL gene analysis in familial pheochromocytoma cases and also in those with no familial history. In order to investigate the relevance of different amino acid changes in the VHL phenotype, we also analyzed the genotype-phenotype correlations using structural analysis to assess protein stability and complexes. The association of clear cell renal carcinoma (CCRC) development with a relatively high loss of structural stability in pVHL missense-mutants was consistent. Structural stability data in the genotype-phenotype correlations therefore provides us with a better understanding of VHL clinical implications. It is also a suitable approach to the evaluation of unknown significance changes.


Asunto(s)
Enfermedad de von Hippel-Lindau/genética , Adenocarcinoma de Células Claras/genética , Adolescente , Adulto , Edad de Inicio , Carcinoma de Células Renales/genética , Niño , Femenino , Genes Supresores de Tumor , Genotipo , Humanos , Neoplasias Renales/genética , Sustancias Macromoleculares , Masculino , Persona de Mediana Edad , Modelos Moleculares , Mutación Missense/genética , Fenotipo , España , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau
15.
J Clin Endocrinol Metab ; 89(9): 4545-52, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15356060

RESUMEN

Pituitary adenomas are very common in humans. They are of monoclonal origin, very heterogeneous, and produce frequently paradoxical secretion. The normal anterior pituitary (AP) contains some unorthodox multifunctional cells able to store more than one AP hormone (polyhormonal) and/or to express multiple hypothalamic-releasing hormone receptors (multiresponsive). Multifunctional AP cells seem to be involved in plasticity processes such as transdifferentiation or paradoxical secretion. Here, we have characterized the single-cell phenotypes of 15 human pituitary tumors, including prolactinomas, nonfunctioning adenomas, and adenomas from multiple endocrine neoplasia type I (MEN-I) and pituitary Cushing's disease patients. Individual tumor cells were typed according to expression of AP hormones and hypothalamic-releasing hormone receptors by combination of calcium imaging and multiple sequential immunocytochemistry in the same cells. We found a large heterogeneity among the different tumors. In eight of the 15 tumors studied, more than 80% of the cells presented a multifunctional phenotype. This may explain the occurrence of paradoxical secretion. In addition, our results suggest that human pituitary adenomas might derive from multifunctional cells. This is consistent with the existence of a link between pituitary plasticity and tumorigenesis.


Asunto(s)
Adenoma/patología , Hormonas Adenohipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Receptores de Hormona Reguladora de Hormona Hipofisaria/análisis , Adenoma/etiología , Adenoma/metabolismo , Adulto , Anciano , Línea Celular Tumoral , Síndrome de Cushing/metabolismo , Síndrome de Cushing/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/patología , Fenotipo , Neoplasias Hipofisarias/etiología , Neoplasias Hipofisarias/metabolismo , Prolactinoma/metabolismo , Prolactinoma/patología
16.
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
17.
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
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA