Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Psychiatry ; 19(6): 676-81, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23752245

RESUMO

Over 200 rare and fully penetrant pathogenic mutations in amyloid precursor protein (APP), presenilin 1 and 2 (PSEN1 and PSEN2) cause a subset of early-onset familial Alzheimer's disease (EO-FAD). Of these, 21 cases of EO-FAD families carrying unique APP locus duplications remain the only pathogenic copy number variations (CNVs) identified to date in Alzheimer's disease (AD). Using high-density DNA microarrays, we performed a comprehensive genome-wide analysis for the presence of rare CNVs in 261 EO-FAD and early/mixed-onset pedigrees. Our analysis revealed 10 novel private CNVs in 10 EO-FAD families overlapping a set of genes that includes: A2BP1, ABAT, CDH2, CRMP1, DMRT1, EPHA5, EPHA6, ERMP1, EVC, EVC2, FLJ35024 and VLDLR. In addition, CNVs encompassing two known frontotemporal dementia genes, CHMP2B and MAPT were found. To our knowledge, this is the first study reporting rare gene-rich CNVs in EO-FAD and early/mixed-onset AD that are likely to underlie pathogenicity in familial AD and perhaps related dementias.


Assuntos
Doença de Alzheimer/genética , Variações do Número de Cópias de DNA , Adulto , Idade de Início , Idoso , Estudos de Coortes , Família , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Linhagem
2.
Neuropathol Appl Neurobiol ; 38(2): 213-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21988727

RESUMO

AIMS: Atypical (World Health Organization grade II) meningiomas have moderately high recurrence rates; even for completely resected tumours, approximately one-third will recur. Post-operative radiotherapy may aid local control and improve survival, but carries the risk of side effects. More accurate prediction of recurrence risk is therefore needed for patients with atypical meningioma. Previously, we used high-resolution array comparative genomic hybridization to identify genetic variations in 47 primary atypical meningiomas and found that approximately 60% of tumours show gain of 1q at 1q25.1 and 1q25.3 to 1q32.1 and that 1q gain appeared to correlate with shorter progression-free survival. This study aimed to validate and extend these findings in an independent sample. METHODS: Eighty-six completely resected atypical meningiomas (with 25 recurrences) from two neurosurgical centres in Ireland were identified and clinical follow-up was obtained. Utilizing a dual-colour interphase fluorescence in situ hybridization assay, 1q gain was assessed using Bacterial Artificial Chromosome probes directed against 1q25.1 and 1q32.1. RESULTS: The results confirm the high prevalence of 1q gain at these loci in atypical meningiomas. We further show that gain at 1q32.1 and age each correlate with progression-free survival in patients who have undergone complete surgical resection of atypical meningiomas. CONCLUSIONS: These independent findings suggest that assessment of 1q copy number status can add clinically useful information for the management of patients with atypical meningiomas.


Assuntos
Aberrações Cromossômicas , Cromossomos Humanos Par 1 , Neoplasias Meníngeas/genética , Meningioma/genética , Recidiva Local de Neoplasia/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Hibridização in Situ Fluorescente , Masculino , Neoplasias Meníngeas/mortalidade , Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/radioterapia , Meningioma/mortalidade , Meningioma/patologia , Meningioma/radioterapia , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/patologia , Prognóstico , Adulto Jovem
3.
Br J Cancer ; 99(8): 1302-6, 2008 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-18797466

RESUMO

Germline mutations in both BRCA2 and CHEK2 are associated with an increased risk for male breast cancer. To search for potential interactions between the products of these breast cancer susceptibility genes, we undertook systematic mapping of BRCA2 for potential phosphorylation sites by CHEK2. In vitro kinase assays and mass spectrometric analysis identified a 50 amino-acid fragment within the N-terminus of BRCA2 potentially targeted by CHEK2, containing two major phosphopeptides. Inducible overexpression of this peptide, but not a derivative with mutated phosphorylation sites, leads to increased chromosome fragmentation and suppression of cellular proliferation. These results suggest a link between CHEK2 and BRCA2 pathways, which may contribute to the spectrum of cancers associated with germline CHEK2 mutations.


Assuntos
Proteína BRCA2/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Proteína BRCA2/genética , Neoplasias da Mama Masculina/genética , Neoplasias da Mama Masculina/metabolismo , Linhagem Celular , Quinase do Ponto de Checagem 2 , Fragmentação do DNA , Feminino , Predisposição Genética para Doença , Humanos , Immunoblotting , Técnicas In Vitro , Masculino , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Proteínas Serina-Treonina Quinases/genética
4.
Cancer Res ; 61(20): 7683-8, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11606412

RESUMO

Astrocytomas are brain tumors with variable responses to radiation and chemotherapy. Tumor grade and patient age are important prognostic factors but do not account for the variability in clinical outcome. We hypothesized that genetic subgroups play a role in the outcome of grade III astrocytomas and studied 80 grade III astrocytomas by comparative genomic hybridization. Some chromosomal aberrations (+7p/q, -9p, -10q, -13q, +19q) were related to aberrations that are frequent in grade IV astrocytoma, whereas others (+10p, -11q, +11p, -Xq) were more frequent in grade III astrocytoma. +7p, +19 and -4q were more frequent in tumors from older patients while -11p was more frequent in tumors from younger patients. Finally, gains of 7p and 7q were associated with shorter patient survival, independent of age. Our results indicate that genetic events underlie the well-known effects of age on survival in grade III astrocytoma and demonstrate the importance of molecular classification in astrocytic tumors.


Assuntos
Astrocitoma/genética , Neoplasias Encefálicas/genética , Adolescente , Adulto , Fatores Etários , Criança , Pré-Escolar , Aberrações Cromossômicas , Transtornos Cromossômicos , Progressão da Doença , Feminino , Dosagem de Genes , Glioblastoma/genética , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/genética , Hibridização de Ácido Nucleico , Prognóstico , Taxa de Sobrevida
5.
Oncogene ; 19(7): 870-7, 2000 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-10702795

RESUMO

The hyaluronidase first isolated from human plasma, Hyal-1, is expressed in many somatic tissues. The Hyal-1 gene, HYAL1, also known as LUCA-1, maps to chromosome 3p21.3 within a candidate tumor suppressor gene locus defined by homozygous deletions and by functional tumor suppressor activity. Hemizygosity in this region occurs in many malignancies, including squamous cell carcinomas of the head and neck. We have investigated whether cell lines derived from such malignancies expressed Hyal-1 activity, using normal human keratinocytes as controls. Hyal-1 enzyme activity and protein were absent or markedly reduced in six of seven carcinoma cell lines examined. Comparative genomic and fluorescence in situ hybridization identified chromosomal deletions of one allele of HYAL1 in six of seven cell lines. Initial RT - PCR analyses demonstrated marked discrepancies between levels of HYAL1 mRNA and protein. Despite repeated sequence analyses, no mutations were found. However, two species of transcripts were identified when primers were used that included the 5' untranslated region. The predominant mRNA species did not correlate with protein translation and contained a retained intron. A second spliced form lacking this intron was found only in cell lines that produced Hyal-1 protein. Inactivation of HYAL1 in these tumor lines is a result of incomplete splicing of its pre-mRNA that appears to be epigenetic in nature. Oncogene (2000) 19, 870 - 877.


Assuntos
Processamento Alternativo/genética , Carcinoma de Células Escamosas/enzimologia , Cromossomos Humanos Par 3/genética , Regulação Enzimológica da Expressão Gênica , Genes Supressores de Tumor/genética , Neoplasias de Cabeça e Pescoço/enzimologia , Hialuronoglucosaminidase/genética , Precursores de RNA/genética , Carcinoma de Células Escamosas/genética , Linhagem Celular , DNA Complementar/genética , DNA Complementar/isolamento & purificação , Indução Enzimática/genética , Éxons/genética , Regulação da Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Humanos , Íntrons/genética , Fases de Leitura Aberta/genética , Regiões Promotoras Genéticas/genética , Células Tumorais Cultivadas
6.
Oncogene ; 18(28): 4144-52, 1999 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10435596

RESUMO

Allelic alterations of chromosomes 1 and 19 are frequent events in human diffuse gliomas and have recently proven to be strong predictors of chemotherapeutic response and prolonged survival in oligodendrogliomas (Cairncross et al., 1998; Smith et al., submitted). Using 115 human diffuse gliomas, we localized regions of common allelic loss on chromosomes 1 and 19 and assessed the association of these deletion intervals with glioma histological subtypes. Further, we evaluated the capacity of multiple modalities to detect these alterations, including loss of heterozygosity (LOH), fluorescence in situ hybridization (FISH), and comparative genomic hybridization (CGH). The correlation coefficients for detection of 1p and 19q alterations, respectively, between modalities were: 0.98 and 0.87 for LOH and FISH, 0.79 and 0.60 for LOH and CGH, and 0.79 and 0.53 for FISH and CGH. Minimal deletion regions were defined on 19q13.3 (D19S412-D19S596) and 1p (D1S468-D1S1612). Loss of the 1p36 region was found in 18% of astrocytomas (10/55) and in 73% (24/33) of oligodendrogliomas (P < 0.0001), and loss of the 19q13.3 region was found in 38% (21/55) of astrocytomas and 73% (24/33) of oligodendrogliomas (P = 0.0017). Loss of both regions was found in 11% (6/55) of astrocytomas and in 64% (21/33) of oligodendrogliomas (P < 0.0001). All gliomas with LOH on either 1p or 19q demonstrated loss of the corresponding FISH probe, 1p36 or 19q13.3, suggesting not only locations of putative tumor suppressor genes, but also a simple assay for assessment of 1p and 19q alterations as diagnostic and prognostic markers.


Assuntos
Neoplasias Encefálicas/genética , Cromossomos Humanos Par 19/genética , Cromossomos Humanos Par 1/genética , Glioma/genética , Deleção de Sequência , Astrocitoma/genética , Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Cromossomos Humanos Par 1/ultraestrutura , Cromossomos Humanos Par 19/ultraestrutura , Glioma/classificação , Glioma/patologia , Humanos , Hibridização in Situ Fluorescente , Perda de Heterozigosidade , Oligodendroglioma/genética , Oligodendroglioma/patologia
7.
Clin Cancer Res ; 5(6): 1435-43, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10389929

RESUMO

Glioblastoma multiforme (GM) is the most common and most malignant astrocytoma in adults. After surgery, radiation therapy extends patient survival; however, in vivo response to radiation therapy is variable. The purpose of this investigation was to determine whether the cytogenetic abnormalities of GM differ according to patient response to radiation therapy. Radiation response was defined by either progression [radiation-resistant (RR)] or resolution [radiation-sensitive (RS)] of tumor at the first postradiation radiographic imaging evaluation. Twenty RR and 10 RS frozen tissue specimens were subjected to cytogenetic analysis by comparative genomic hybridization. RS and RR specimens had different cytogenetic aberrations that mapped predominantly to chromosomes 7, 9, 10, 13, and 19. Relative gain of 7 occurred in 70% of the RR and 30% of the RS cases and was the most significant difference involving a single change between the two groups (P = 0.06). RR and RS specimens also differed in their patterns of simultaneous cytogenetic aberrations. A simultaneous gain of chromosomes 7 and 19 was found in 30% of the RR cases but was absent in the RS group. Concurrent loss of 9p23-24 and 13q14 regions was absent in the RS cohort but occurred in 30% of the RR series. This latter cytogenetic pattern was also associated with older age. Amplifications were more common in the RR series, but the difference did not reach statistical significance. The data suggest that GM with different in vivo responses to radiation therapy also differ cytogenetically.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/radioterapia , Aberrações Cromossômicas/genética , Glioblastoma/genética , Glioblastoma/radioterapia , Adulto , Idoso , Neoplasias Encefálicas/mortalidade , Transtornos Cromossômicos , Mapeamento Cromossômico , Estudos de Coortes , Progressão da Doença , Feminino , Amplificação de Genes , Glioblastoma/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Hibridização de Ácido Nucleico , Deleção de Sequência , Taxa de Sobrevida , Resultado do Tratamento
8.
J Neuropathol Exp Neurol ; 56(9): 999-1008, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9291941

RESUMO

The p16 protein plays a key role in cell cycle control by preventing CDK4 from inactivating the retinoblastoma protein (pRb). The corresponding tumor suppressor gene (p16/MTS1/CDKN2) has recently been implicated in malignant progression of astrocytomas and could potentially serve as an important marker for patient prognosis and for guiding specific therapeutic strategies. We have undertaken a study to evaluate 2 methods of detecting p16 deletion. Thirty diffuse gliomas were analyzed for p16 gene dosage. Dual color fluorescence in situ hybridization (FISH) was performed on cytologic preparations using paired centromeric (CEN) and locus-specific probes for CEN9/p16, CEN8/RB, and CEN12/CDK4. Quantitative PCR was performed using primers for p16, MTAP, and reference genes. Eleven cases were also studied using comparative genomic hybridization (CGH). Abnormalities of the p16-CDK4-RB pathway were identified in 21 (70%) cases by FISH and/or PCR. These included 15 (50%) with p16 deletion, 9 of which were detected by both techniques, 3 by FISH alone, and 3 by PCR alone (concordance rate = 81%). FISH analysis further revealed tetraploidy/aneuploidy in 14 (47%), RB deletion in 11 (37%) and CDK4 amplification in 1 (3.3%). There were 94% and 100% concordance rates between CGH and FISH or PCR, respectively. Quantitative PCR was noninformative in 4 cases. Although FISH and quantitative PCR are both reliable techniques, each has limitations. PCR is likely to miss p16 deletions when there is significant normal cell contamination or clonal heterogeneity, whereas the p16 YAC probe used for FISH analysis may miss small deletions. Replacement of the latter with a cosmid probe may improve the sensitivity of FISH in future experiments.


Assuntos
Neoplasias Encefálicas/genética , Proteínas de Transporte/genética , Deleção de Genes , Glioma/genética , Inibidor p16 de Quinase Dependente de Ciclina , Humanos , Hibridização in Situ Fluorescente , Hibridização de Ácido Nucleico , Reação em Cadeia da Polimerase
9.
Cancer Genet Cytogenet ; 83(2): 127-35, 1995 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7553582

RESUMO

The short arm of chromosome 9 is frequently deleted in malignant gliomas. We used locus-specific probes for interferon-A (IFNA) and D9S3 in combination with a chromosome 9 centromeric probe to detect genetic aberrations on a cell-by-cell basis in touch preparations of 30 glioblastomas by fluorescence in situ hybridization. Seven (23%) of 30 tumors had deletions in > 70% of cells; the IFNA locus was deleted in all seven, but the D9S3 locus was deleted in only five of the seven. The latter data confirm that a tumor suppressor gene on 9p relevant to glioblastoma multiforme lies between D9S3 and IFNA. Eleven tumors had deletions in 20-40% of cells, more than three standard deviations above the level in control tissues. The remaining tumors had deletions in < 20% of cells. The seven tumors with the lowest percentage of deleted cells each had more than one genetically abnormal population of cells. In total, 10 cases were of this type (i.e., aneusomic for chromosome 9). Three of these 10 tumors had hybridization patterns consistent with polyploidy.


Assuntos
Aneuploidia , Neoplasias Encefálicas/genética , Cromossomos Humanos Par 9 , Deleção de Genes , Heterogeneidade Genética , Glioblastoma/genética , Poliploidia , Adulto , Idoso , Humanos , Hibridização in Situ Fluorescente , Interferons/genética , Pessoa de Meia-Idade
10.
Cancer Genet Cytogenet ; 100(1): 77-83, 1998 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-9406586

RESUMO

Gliomas that aggregate in otherwise unremarkable families may have a heritable genetic basis. To determine the spectrum of genetic alterations in glioma-susceptible families, we examined tumor DNA from familial cases for regions of chromosomal gain or loss using comparative genomic hybridization (CGH). We compared chromosomal alterations within and among glioma families to those found in sporadic gliomas. A specific chromosomal abnormality common to the tumors of multiple unrelated probands with glioma or a specific chromosomal abnormality common to multiple affected persons in a single glioma-prone family would support the hypothesis of an inherited predisposition to glioma and at the same time identify specific regions of the genome harboring putative glioma susceptibility genes. Tumor DNA from 11 patients from seven families with two or more individuals with glioma was analyzed, including three members of a remarkable family having 10 affected individuals. We found no chromosomal abnormality common to all tumors of all probands nor did we find family-specific abnormalities in two of three glioma-prone kindreds. There were frequent copy number aberrations (CNAs) on chromosomes 7, 10, 19, and the sex chromosomes; other CNAs included +3q(13.3-29), -4q, +5q, -9q34, +12, -13q(21-->33), -15, -16p, +17qter, -18, -21, and -22. Amplifications occurred at +2 7p(11.1-->12), +2 7q(21.2-->33), +2 12q(13.2-->14), and +2 12p(11-->12). Although there were several novel CNAs [-16p, and +2 12p(11-p12)], none could readily explain the inheritance of these tumors.


Assuntos
Aberrações Cromossômicas , Glioma/genética , Hibridização de Ácido Nucleico , Adolescente , Adulto , Idoso , Criança , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
11.
J Neurosurg ; 90(2): 306-14, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9950502

RESUMO

OBJECT: This study was conducted to determine whether comparative genomic hybridization (CGH) is a more sensitive method for detecting genetic aberrations than other tests currently in use. METHODS: The authors used CGH to examine 40 primary and 13 recurrent adenomas obtained from 52 patients for loss and gain of genetic material. Copy number aberrations (CNAs) were detected in 25 (48%) of the 52 patients studied. The chromosomes affected were, in order of decreasing frequency, 11, 7, X, 1, 8, 13, 5, 14, 2, 6, 9, 10, 12, 3, 18, 21, 4, 16, 15, 19, 22, and Y. Endocrinologically active adenomas were more likely to contain (p = 0.009) and had a greater number (p = 0.003) of CNAs. Of 26 adenomas with CNAs, 18 showed multiple aberrations involving entire chromosomes or chromosome arms. The most frequent CNA involving a chromosome subregion, which was present in four (8%) of 53 adenomas, was the loss of all chromosome 11 material except for a preserved common segment containing 11q13. Immunoperoxidase staining did not detect cyclin D1 expression in those four cases, making cyclin D1 an unlikely target of this rearrangement. CONCLUSIONS: These findings indicate that genetic abnormalities are present in pituitary adenomas at a higher rate than previously reported, are associated with endocrinological activity, and often involve several chromosomes. Rearrangement at 11q13 may inactivate a tumor suppressor gene or activate an oncogene that is important in the initiation or progression of sporadic pituitary adenomas.


Assuntos
Adenoma/genética , Aberrações Cromossômicas/genética , Cromossomos Humanos Par 11 , Rearranjo Gênico/genética , Neoplasias Hipofisárias/genética , Adenoma/metabolismo , Adolescente , Adulto , Idoso , Ciclina D1/metabolismo , Feminino , Humanos , Técnicas Imunoenzimáticas , Masculino , Pessoa de Meia-Idade , Hibridização de Ácido Nucleico , Neoplasias Hipofisárias/metabolismo , Sensibilidade e Especificidade
12.
Oncogene ; 33(33): 4226-35, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-24056967

RESUMO

Ovarian cancers are thought to result from the accumulation of multiple genetic aberrations that transform ovarian and/or fallopian tube surface epithelial cells, allowing for their abnormal growth, proliferation and metastasis. In the report presented here, we carried out genome-wide copy-number analysis using comparative genomic hybridization on a panel of mouse ovarian cancer (OVCA) cell lines previously established in our laboratory. We identified a recurrent focal amplification on mouse chromosomal region 2qB, which contains the LIM-homeodomain-containing transcription factor 1B (Lmx1b) gene. LMX1B is not expressed in normal human ovary, but is expressed in many human OVCA cell lines and primary tumors. High expression of LMX1B correlates with poor outcome. To clarify the role of LMX1B in ovarian carcinogenesis, we transduced LMX1B into a panel of mouse and human OVCA cell lines and demonstrated that LMX1B strongly promotes migration of cancer cells in culture and promotes xenograft growth in nude mice. Conversely, knockdown of LMX1B in a human cell line with endogenous high expression of LMX1B inhibits cell migration in vitro and tumor growth in vivo. Microarray analysis of cells overexpressing LMX1B identified the nuclear factor (NF)-κB pathway as a potential mediator of tumor progression and subsequent treatment of NFκB inhibitor decreased the migratory capacity of these cells. Thus, our data demonstrate that LMX1B is a novel oncogene in OVCA pathogenesis.


Assuntos
Proteínas com Homeodomínio LIM/genética , Oncogenes , Neoplasias Ovarianas/genética , Fatores de Transcrição/genética , Animais , Carcinogênese , Linhagem Celular Tumoral , Feminino , Amplificação de Genes , Humanos , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , NF-kappa B/metabolismo , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Transdução de Sinais , Fatores de Transcrição/metabolismo , Transcriptoma , Carga Tumoral
13.
Neurology ; 78(16): 1250-7, 2012 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-22491860

RESUMO

OBJECTIVES: More than 30 different rare mutations, including copy number variants (CNVs), in the amyloid precursor protein gene (APP) cause early-onset familial Alzheimer disease (EOFAD), whereas the contribution of common APP variants to disease risk remains controversial. In this study we systematically assessed the role of both rare and common APP DNA variants in Alzheimer disease (AD) families. METHODS: Families with EOFAD genetically linked to the APP region were screened for missense mutations and locus duplications of APP. Further, using genome-wide DNA microarray data, we examined the APP locus for CNVs in a total of 797 additional early- and late-onset AD pedigrees. Finally, 423 single nucleotide polymorphisms (SNPs) in the APP locus, including 2 promoter polymorphisms previously associated with AD risk, were tested in up to 4,200 individuals from multiplex AD families. RESULTS: Analyses of 8 21q21-linked families revealed one family carrying a nonsynonymous mutation in exon 17 (Val717Leu) and another family with a partially penetrant 3.5-Mb locus duplication encompassing APP. CNV analysis in the APP locus revealed an additional family carrying a fully penetrant 380-kb duplication, merely spanning APP. Last, contrary to previous reports, association analyses of more than 400 different SNPs in or near APP failed to show significant effects on AD risk. CONCLUSION: Our study shows that APP mutations and locus duplications are a very rare cause of EOFAD and that the contribution of common APP variants to AD susceptibility is insignificant. Furthermore, duplications of APP may not be fully penetrant, possibly indicating the existence of hitherto unknown protective genetic factors.


Assuntos
Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Predisposição Genética para Doença/genética , Idoso , Variações do Número de Cópias de DNA , Feminino , Loci Gênicos/genética , Estudo de Associação Genômica Ampla/métodos , Humanos , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto/genética , Linhagem , Polimorfismo de Nucleotídeo Único
14.
J Neurooncol ; 24(1): 47-55, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-8523075

RESUMO

The unregulated growth that is characteristic of human malignant gliomas is accompanied by, and may result from, losses and/or gains of genetic material. Understanding the mechanisms that underlie how particular genetic aberrations cause dysfunctional growth will help elucidate the pathogenesis of this disease. Two techniques are proving useful in evaluating the clinical relevance of specific genetic aberrations in malignant gliomas. Fluorescence in situ hybridization (FISH) permits direct visualization of gains and losses of genetic material in single cells and quantitation of cellular subpopulations that have particular genetic aberrations. Comparative genomic hybridization can identify regions of genetic gain and loss in tumor DNA.


Assuntos
Neoplasias Encefálicas/genética , Aberrações Cromossômicas/genética , DNA de Neoplasias/genética , Glioma/genética , Transtornos Cromossômicos , Amplificação de Genes , Genoma Humano , Humanos , Hibridização in Situ Fluorescente , Cariotipagem
15.
Genes Chromosomes Cancer ; 13(2): 86-93, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7542911

RESUMO

A protocol for comparative genomic hybridization by use of nucleotides directly labeled with fluorochromes was used to map regions of deletion and amplification in ten glioma cell lines. The protocol greatly reduced experimental artifacts. We detected several genetic aberrations, including whole chromosome loss and gain, partial loss and gain, possible isochromosome, and higher level DNA amplification. The most frequent losses (in order of frequency) occurred on chromosomes 10, 18, 13, 11, 9, 14, 4, 6, 1, and X. The most common gain occurred on chromosome 7. Several sites of previously known and unknown amplifications were observed.


Assuntos
Neoplasias Encefálicas/genética , Aberrações Cromossômicas/genética , Glioma/genética , Mapeamento Cromossômico , DNA de Neoplasias/análise , Feminino , Humanos , Masculino , Metáfase , Hibridização de Ácido Nucleico , Células Tumorais Cultivadas
16.
Int J Cancer ; 60(6): 812-9, 1995 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-7896451

RESUMO

Comparative genomic hybridization (CGH) is a recent molecular cytogenetic method that detects and localizes gains or losses in DNA copy number across the entire tumor genome. We used CGH to examine 9 glioma cell lines and 20 primary and 10 recurrent glioblastoma tumors. More than 25% of the primary tumors had gains on chromosome 7; they also had frequent losses on 9p, 10, 13 and Y. The losses on chromosome 13 included several interstitial deletions, with a common area of loss of 13q21. The recurrent tumors not only had gains on chromosome 7 and losses on 9p, 10, 13 and Y but also frequent losses on 6 and 14. One recurrent tumor had a deletion of 10q22-26. Cell lines showed gains of 5p, 7 and Xp; frequent amplifications at 8q22-24.2, 7q21-32 and 3q26.2-29 and frequent losses on 4, 10, 13, 14 and Y. Because primary and recurrent tumors and cell lines showed abnormalities of DNA copy number on chromosomes 7, 10, 13 and Y, these regions may play a fundamental role in tumor initiation and/or progression. The propensity for losses on chromosomes 6 and 14 to occur in recurrent tumors suggests that these aberrations play a role in tumor recurrence, the development of resistance to therapy or both. Analysis of common areas of loss and gain in these tumors and cell lines provides a basis for future attempts to more finely map these genetic changes.


Assuntos
Aberrações Cromossômicas , Glioblastoma/genética , Glioma/genética , Mapeamento Cromossômico , DNA de Neoplasias/análise , Genoma , Humanos , Hibridização de Ácido Nucleico , Células Tumorais Cultivadas
17.
EMBO J ; 16(11): 2985-95, 1997 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-9214616

RESUMO

The proto-oncogene MYCN is often amplified in human neuroblastomas. The assumption that the amplification contributes to tumorigenesis has never been tested directly. We have created transgenic mice that overexpress MYCN in neuroectodermal cells and develop neuroblastoma. Analysis of tumors by comparative genomic hybridization revealed gains and losses of at least seven chromosomal regions, all of which are syntenic with comparable abnormalities detected in human neuroblastomas. In addition, we have shown that increases in MYCN dosage or deficiencies in either of the tumor suppressor genes NF1 or RB1 can augment tumorigenesis by the transgene. Our results provide direct evidence that MYCN can contribute to the genesis of neuroblastoma, suggest that the genetic events involved in the genesis of neuroblastoma can be tumorigenic in more than one chronological sequence, and offer a model for further study of the pathogenesis and therapy of neuroblastoma.


Assuntos
Genes myc , Neuroblastoma/genética , Tumores Neuroectodérmicos/genética , Proteínas Proto-Oncogênicas c-myc/biossíntese , Abdome/patologia , Animais , Aberrações Cromossômicas , Transtornos Cromossômicos , DNA de Neoplasias/genética , Dosagem de Genes , Expressão Gênica , Marcação de Genes , Camundongos , Camundongos Transgênicos , Metástase Neoplásica , Neuroblastoma/etiologia , Neuroblastoma/patologia , Tumores Neuroectodérmicos/etiologia , Tumores Neuroectodérmicos/patologia , Hibridização de Ácido Nucleico , Proto-Oncogene Mas , Especificidade da Espécie , Tórax/patologia
18.
Am J Hum Genet ; 64(1): 70-6, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9915945

RESUMO

A Prader-Willi syndrome patient is described who has a de novo balanced translocation, (4;15)(q27;q11.2)pat, with breakpoints lying between SNRPN exons 2 and 3. Parental-origin studies indicate that there is no uniparental disomy and no apparent deletion. This patient expresses ZNF127, SNRPN exons 1 and 2, IPW, and D15S227E (PAR1) but does not express either SNRPN exons 3 and 4 or D15S226E (PAR5), as assayed by reverse transcription-PCR, of peripheral blood cells. Methylation studies showed normal biparental patterns of inheritance of loci DN34/ZNF127, D15S63, and SNRPN exon 1. Results for this patient and that reported by Sun et al. support the contention that an intact genomic region and/or transcription of SNRPN exons 2 and 3 play a pivotal role in the manifestations of the major clinical phenotype in Prader-Willi syndrome.


Assuntos
Autoantígenos/genética , Cromossomos Humanos Par 15 , Cromossomos Humanos Par 4 , Síndrome de Prader-Willi/genética , Ribonucleoproteínas Nucleares Pequenas/genética , População Negra/genética , Southern Blotting , Bandeamento Cromossômico , Éxons , Humanos , Hibridização in Situ Fluorescente , Masculino , Reação em Cadeia da Polimerase , Translocação Genética , Proteínas Centrais de snRNP
19.
Genomics ; 45(1): 42-7, 1997 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9339359

RESUMO

P1 clones near the telomeres and centromeres of each mouse chromosome except Y have been selected from a mouse genomic library and mapped using fluorescence in situ hybridization (FISH). Each clone was selected to contain a genetically mapped polymorphic DNA sequence as close as possible to the centromere or telomere of a chromosome. The genetic distance from the various P1 clones to the most distal genetically mapped polymorphic sequence ranged from 0 for about half of the clones to 6.7 cM for the probe at the telomere of chromosome 14. The average distance to the most distal or proximal chromosome marker was 1.5 cM. The use of FISH with these probes for mouse chromosome identification during comparative genomic hybridization is illustrated.


Assuntos
Mapeamento Cromossômico , Sondas de DNA , Hibridização in Situ Fluorescente , Animais , Clonagem Molecular , Camundongos , Polimorfismo Genético
20.
Am J Pathol ; 150(1): 81-9, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9006325

RESUMO

Neuroblastoma behavior is variable and outcome partially depends on genetic factors. However, tumors that lack high-risk factors such as MYCN amplification or 1p deletion may progress, possibly due to other genetic aberrations. Comparative genomic hybridization summarizes DNA copy number abnormalities in a tumor by mapping them to their positions on normal metaphase chromosomes. We analyzed 29 tumors from nearly equal proportions of children with stage I, II, III, IV, and IV-S disease by comparative genomic hybridization. We found two classes of copy number abnormalities: whole chromosome and partial chromosome. Whole chromosome losses were frequent at 11, 14, and X. The most frequent partial chromosome losses were on 1p and 11q. Gains were most frequent on chromosome 17 (72% of cases). The two patterns of gain for this chromosome were whole 17 gain and 17q gain, with 17q21-qter as a minimal common region of gain. Other common gains were on chromosomes 7, 6, and 18. High level amplifications were detected at 2p23-25 (MYCN region), at 4q33-35, and at 6p11-22. Chromosome 17q gains were associated with 1p and/or 11q deletions and advanced stage. The high frequency of chromosome 17 gain and its association with bad prognostic factors suggest an important role for this chromosome in the development of neuroblastoma.


Assuntos
Aberrações Cromossômicas/genética , Aberrações Cromossômicas/patologia , Cromossomos Humanos Par 17 , Hibridização in Situ Fluorescente , Neuroblastoma/genética , Neuroblastoma/patologia , Pré-Escolar , Transtornos Cromossômicos , Cromossomos Humanos Par 2 , Deleção de Genes , Genes myc , Humanos , Hibridização in Situ Fluorescente/métodos , Lactente , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA