Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Acta Neuropathol Commun ; 7(1): 59, 2019 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-31023364

RESUMO

BACKGROUND: Diffuse lower WHO grade II and III gliomas (LGG) are slowly progressing brain tumors, many of which eventually transform into a more aggressive type. LGG is characterized by widespread genetic and transcriptional heterogeneity, yet little is known about the heterogeneity of the DNA methylome, its function in tumor biology, coupling with the transcriptome and tumor microenvironment and its possible impact for tumor development. METHODS: We here present novel DNA methylation data of an LGG-cohort collected in the German Glioma Network containing about 85% isocitrate dehydrogenase (IDH) mutated tumors and performed a combined bioinformatics analysis using patient-matched genome and transcriptome data. RESULTS: Stratification of LGG based on gene expression and DNA-methylation provided four consensus subtypes. We characterized them in terms of genetic alterations, functional context, cellular composition, tumor microenvironment and their possible impact for treatment resistance and prognosis. Glioma with astrocytoma-resembling phenotypes constitute the largest fraction of nearly 60%. They revealed largest diversity and were divided into four expression and three methylation groups which only partly match each other thus reflecting largely decoupled expression and methylation patterns. We identified a novel G-protein coupled receptor and a cancer-related 'keratinization' methylation signature in in addition to the glioma-CpG island methylator phenotype (G-CIMP) signature. These different signatures overlap and combine in various ways giving rise to diverse methylation and expression patterns that shape the glioma phenotypes. The decrease of global methylation in astrocytoma-like LGG associates with higher WHO grade, age at diagnosis and inferior prognosis. We found analogies between astrocytoma-like LGG with grade IV IDH-wild type tumors regarding possible worsening of treatment resistance along a proneural-to-mesenchymal axis. Using gene signature-based inference we elucidated the impact of cellular composition of the tumors including immune cell bystanders such as macrophages. CONCLUSIONS: Genomic, epigenomic and transcriptomic factors act in concert but partly also in a decoupled fashion what underpins the need for integrative, multidimensional stratification of LGG by combining these data on gene and cellular levels to delineate mechanisms of gene (de-)regulation and to enable better patient stratification and individualization of treatment.


Assuntos
Neoplasias Encefálicas/genética , Metilação de DNA/genética , Dosagem de Genes , Glioma/genética , Transcriptoma , Neoplasias Encefálicas/complicações , Biologia Computacional , Epigênese Genética , Humanos , Gradação de Tumores , Microambiente Tumoral/genética , Organização Mundial da Saúde
2.
Clin Genet ; 94(1): 185-186, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29498415

RESUMO

Ataxia-oculomotor apraxia type 4 (AOA4) is a rare autosomal recessive neurologic disorder. The phenotype is characterized by ataxia, oculomotor apraxia, peripheral neuropathy and dystonia. AOA4 is caused by biallelic pathogenic variants in the PNKP gene encoding a polynucleotide kinase 3'-phosphatase with an important function in DNA-damage repair. By whole exome sequencing, we identified 2 variants within the PNKP gene in a 27-year-old German woman with a clinical AOA phenotype combined with a cerebellar pilocytic astrocytoma diagnosed at 23 years of age. One variant, a duplication in exon 14 resulting in the frameshift c.1253_1269dup p.(Thr424fs*49), has previously been described as pathogenic, for example, in cases of AOA4. The second variant, representing a nonsense mutation in exon 17, c.1545C>G p.(Tyr515*), has not yet been described and is predicted to cause a loss of the 7 C-terminal amino acids. This is the first description of AOA4 in a patient with central European descent. Furthermore, the occurrence of a pilocytic astrocytoma has not been described before in an AOA4 patient. Our data demonstrate compound heterozygous PNKP germline variants in a German patient with AOA4 and provide evidence for a possible link with tumor predisposition. Localization of the 2 variants in human PNKP NP_009185.2. NM_007254.3:c.1253_1269dup p.(Thr424fs*49) is predicted to cause a frameshift within the kinase domain, NM_007254.3:c.1545C>G p.(Tyr515*) is predicted to cause loss of 2 C-terminal amino acids of the kinase domain and 5 additional C-terminal amino acids.


Assuntos
Apraxias/congênito , Astrocitoma/genética , Síndrome de Cogan/genética , Enzimas Reparadoras do DNA/genética , Sequenciamento do Exoma , Heterozigoto , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Alelos , Sequência de Aminoácidos , Apraxias/diagnóstico , Apraxias/genética , Astrocitoma/diagnóstico , Síndrome de Cogan/diagnóstico , Dano ao DNA , Enzimas Reparadoras do DNA/química , Éxons , Feminino , Humanos , Mutação , Linhagem , Fosfotransferases (Aceptor do Grupo Álcool)/química
4.
Cytogenet Genome Res ; 136(4): 308-13, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22433391

RESUMO

Carriers of completely balanced chromosomal translocations have all necessary genetic information. Nevertheless, because of the possibility of maldistribution during gametogenesis, they are at increased risk for infertility, miscarriage, stillbirth or having a child with congenital anomalies including mental retardation. As postnatal clinical reports are infrequent, prediction of clinical course for specific unbalanced karyotypes diagnosed during pregnancy remains difficult. Here, we report the 6th case of partial trisomy 6p and partial monosomy 20p due to an unbalanced adjacent-1 segregation of the rare familial translocation t(6;20)(p21;p13). We give a thorough clinical description of the present case, demonstrating broad phenotypic overlap with the 5 previously published cases reviewed here, providing important data on postnatal outcome.


Assuntos
Translocação Genética , Trissomia/genética , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Adulto , Bandeamento Cromossômico , Deleção Cromossômica , Coloração Cromossômica , Cromossomos Humanos Par 20/genética , Cromossomos Humanos Par 6/genética , Feminino , Humanos , Lactente , Cariotipagem , Masculino , Linhagem , Fenótipo , Gravidez
5.
J Anim Ecol ; 76(3): 503-14, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17439467

RESUMO

1. Functional responses -- the relationship between resource intake rate and resource abundance -- are widely used in explaining predator-prey interactions yet many studies indicate that resource availability is crucial in dictating intake rates. 2. For time-stressed migrant birds refuelling at passage sites, correct decisions concerning patch use are crucial as they determine fattening rates and an individual's future survival and reproduction. Measuring availability alongside abundance is essential if spatial and temporal patterns of foraging are to be explained. 3. A suite of shorebird species stage in Delaware Bay where they consume horseshoe crab Limulus polyphemus eggs. Several factors including spawning activity and weather give rise to marked spatial and temporal variation in the abundance and availability of eggs. We undertook field experiments to determine and contrast the intake rates of shorebird species pecking for surface and probing for buried eggs. 4. Whether eggs were presented on the sand surface or buried, we demonstrate strong aggregative responses and rapid depletion (up to 80%). Depletion was greater at deeper depths when more eggs were present. No consistent give-up densities were found. Type II functional responses were found for surface eggs and buried eggs, with peck success twice as high in the former. Maximum intake rates of surface eggs were up to 83% higher than those of buried eggs. 5. Caution is needed when applying functional responses predicted on the basis of morphology. Our expectation of a positive relationship between body size and intake rate was not fully supported. The smallest species, semipalmated sandpiper, had the lowest intake rate but the largest species, red knot, achieved only the same intake rate as the mid-sized dunlin. 6. These functional responses indicate that probing is rarely more profitable than pecking. Currently, few beaches provide egg densities sufficient for efficient probing. Areas where eggs are deposited on the sand surface are critical for successful foraging and ongoing migration. This may be especially true for red knot, which have higher energetic demands owing to their larger body size yet appear to have depressed intake rates because they consume smaller prey than their body size should permit.


Assuntos
Migração Animal/fisiologia , Charadriiformes/fisiologia , Comportamento Alimentar/fisiologia , Caranguejos Ferradura/crescimento & desenvolvimento , Comportamento Predatório/fisiologia , Animais , Charadriiformes/anatomia & histologia , Delaware , Ingestão de Alimentos , Ovos , Caranguejos Ferradura/anatomia & histologia , Caranguejos Ferradura/embriologia , Densidade Demográfica , Dinâmica Populacional
6.
Neurology ; 68(10): 743-50, 2007 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-17339581

RESUMO

OBJECTIVE: Because in most patients with mental retardation (MR), who constitute 2 to 3% of the population, the etiology remains unknown, we wanted to identify novel chromosomal candidate regions and genes associated with the MR phenotype. METHODS: We screened for microimbalances in 60 clinically well-characterized patients with unexplained MR mostly combined with congenital anomalies. Genome-wide array-based comparative genomic hybridization was performed on DNA microarrays with an average resolution of <0.5 Mb. We verified every nonpolymorphic array clone outside the diagnostic thresholds by fluorescence in situ hybridization and performed breakpoint analyses on confirmed imbalances. RESULTS: Six presumably causal microimbalances were detected, five of which have not been reported. Microdeletions were found in five patients with MR and distinctive facial features, who also had neurologic findings (three cases), brain anomalies (two cases), and growth retardation (two cases), in chromosomal bands 6q11.1-q13 (10.8 Mb), Xq21.31-q21.33 (4.0 Mb), 1q24.1-q24.2 (3.8 Mb), 19p13.12 (2.1 Mb), and 4p12-p13 (1.1 Mb). One microduplication was detected in 22q11.2 (2.8 Mb) including the DiGeorge syndrome critical region in a patient with mild MR, microcephaly at birth, and dysmorphisms. Three imbalances were shown to be de novo and two inherited. The Xq21 microdeletion in a boy with borderline intellectual functioning was inherited from a normal mother; the 22q11.2 microduplication was inherited from a normal father and was present in two affected siblings. CONCLUSION: We could identify novel microimbalances as the probable cause of mental retardation in 10% of patients with unclear etiology. The gene content of the microimbalances was found to correlate with phenotype severity. Precise breakpoint analyses allowed the identification of deleted genes presumably causing mental retardation.


Assuntos
Perfilação da Expressão Gênica , Deficiência Intelectual/etiologia , Deficiência Intelectual/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Anormalidades Múltiplas , Criança , Aberrações Cromossômicas , Feminino , Humanos , Hibridização in Situ Fluorescente , Lactente , Deficiência Intelectual/patologia , Imageamento por Ressonância Magnética , Masculino
7.
Oncogene ; 26(7): 1088-97, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16909113

RESUMO

The molecular pathogenesis of pleomorphic xanthoastrocytoma (PXA), a rare astrocytic brain tumor with a relatively favorable prognosis, is still poorly understood. We characterized 50 PXAs by comparative genomic hybridization (CGH) and found the most common imbalance to be loss on chromosome 9 in 50% of tumors. Other recurrent losses affected chromosomes 17 (10%), 8, 18, 22 (4% each). Recurrent gains were identified on chromosomes X (16%), 7, 9q, 20 (8% each), 4, 5, 19 (4% each). Two tumors demonstrated amplifications mapping to 2p23-p25, 4p15, 12q13, 12q21, 21q21 and 21q22. Analysis of 10 PXAs with available high molecular weight DNA by high-resolution array-based CGH indicated homozygous 9p21.3 deletions involving the CDKN2A/p14(ARF)/CDKN2B loci in six tumors (60%). Interphase fluorescence in situ hybridization to tissue sections confirmed the presence of tumor cells with homozygous 9p21.3 deletions. Mutational analysis of candidate genes on 9q, PTCH and TSC1, revealed no mutations in PXAs with 9q loss and no evidence of TSC1 promoter methylation. However, PXAs consistently showed low TSC1 transcript levels. Taken together, our study identifies loss of chromosome 9 as the most common chromosomal imbalance in PXAs and suggests important roles for homozygous CDKN2A/p14(ARF)/CDKN2B deletion as well as low TSC1 mRNA expression in these tumors.


Assuntos
Astrocitoma/genética , Deleção Cromossômica , Cromossomos Humanos Par 9/genética , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Deleção de Genes , Proteína Supressora de Tumor p14ARF/genética , Proteínas Supressoras de Tumor/deficiência , Adolescente , Adulto , Criança , Pré-Escolar , Inibidor de Quinase Dependente de Ciclina p15/deficiência , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Feminino , Homozigoto , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/biossíntese , Proteína 1 do Complexo Esclerose Tuberosa , Proteína Supressora de Tumor p14ARF/deficiência , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética
8.
Acta Neurol Scand ; 114(2): 133-8, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16867037

RESUMO

BACKGROUND: Patients with the karyotypic finding of a terminal deletion in the long arm of chromosome 18 (18q- syndrome) commonly display cerebral dysmyelination and developmental delay. To our knowledge, all reported cases characterized by molecular analysis who had no mental retardation as confirmed by neuropsychological testing had a chromosomal breakpoint within the two most distal bands, 18q22 or 18q23, leading to a deletion of 16 Mb or less. AIMS OF THE STUDY: It was the aim of this study to improve the karyotype-phenotype correlation in 18q- syndrome by thoroughly analyzing the deletion size and the mental and radiologic status in a 23-year-old woman with a terminal 18q deletion. We performed cytogenetic and molecular cytogenetic analysis, brain MRI, and extended neuropsychological testing. RESULTS: Molecular karyotyping revealed a 17 Mb deletion of terminal 18q with a breakpoint in 18q21.33 and no evidence for mosaicism. While brain MRI demonstrated severe global dysmyelination, the patient showed a neuropsychological pattern that allowed for normal psychosocial and job achievement. After delayed development in childhood, the patient caught up during puberty and showed normal verbal intelligence and skills at 23 years. However, visual, visual-spatial, visual-constructional, and executive functions were found to be severely impaired. CONCLUSION: Here, we present a patient with one of the largest terminal 18q deletions reported in an individual without obvious mental retardation. Our analysis extends the phenotypic spectrum for individuals with breakpoints in 18q21.33. In addition, this study highlights the fact that severe global dysmyelination may not be associated with general cognitive deficits.


Assuntos
Aberrações Cromossômicas , Cromossomos Humanos Par 18/genética , Deficiências do Desenvolvimento/genética , Doenças Genéticas Inatas/genética , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/genética , Deleção de Sequência/genética , Adulto , Sequência de Bases , Encéfalo/patologia , Encéfalo/fisiopatologia , Criança , Transtornos Cognitivos/diagnóstico , Transtornos Cognitivos/genética , Transtornos Cognitivos/fisiopatologia , Análise Mutacional de DNA , Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/fisiopatologia , Feminino , Doenças Genéticas Inatas/diagnóstico , Doenças Genéticas Inatas/fisiopatologia , Testes Genéticos , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/diagnóstico , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/fisiopatologia , Humanos , Inteligência/genética , Imageamento por Ressonância Magnética , Mutação/genética , Transtornos Psicomotores/diagnóstico , Transtornos Psicomotores/genética , Transtornos Psicomotores/fisiopatologia , Síndrome
9.
J Pathol ; 208(4): 554-63, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16400626

RESUMO

Desmoplastic medulloblastoma (DMB) is a malignant cerebellar tumour composed of two distinct tissue components, pale islands and desmoplastic areas. Previous studies revealed mutations in genes encoding members of the sonic hedgehog pathway, including PTCH, SMOH and SUFUH in DMBs. However, little is known about other genomic aberrations. We performed comparative genomic hybridization (CGH) analysis of 22 sporadic DMBs and identified chromosomal imbalances in 20 tumours (91%; mean, 4.9 imbalances/tumour). Recurrent chromosomal gains were found on chromosomes 3, 9 (six tumours each), 20, 22 (five tumours each), 2, 6, 7, 17 (four tumours each) and 1 (three tumours). Recurrent losses involved chromosomes X (eight tumours), Y (six of eleven tumours from male patients), 9, 12 (four tumours each), as well as 10, 13 and 17 (three tumours each). Four tumours demonstrated high-level amplifications involving sequences from 1p22, 5p15, 9p, 12p13, 13q33-q34 and 17q22-q24, respectively. Further analysis of the 9p and 17q22-q24 amplicons by array-based CGH (matrix-CGH) and candidate gene analyses revealed amplification of JMJD2C at 9p24 in one DMB and amplification of RPS6KB1, APPBP2, PPM1D and BCAS3 from 17q23 in three DMBs. Among the 17q23 genes, RPS6KB1 showed markedly elevated transcript levels as compared to normal cerebellum in five of six DMBs and four of five classic medulloblastomas investigated. Finally, CGH analysis of microdissected pale islands and desmoplastic areas showed common chromosomal imbalances in five of six informative tumours. In summary, we have identified several novel genetic alterations in DMBs and provide genetic evidence for a monoclonal origin of their different tissue components.


Assuntos
Neoplasias Cerebelares/genética , Perfilação da Expressão Gênica , Meduloblastoma/genética , Análise de Sequência com Séries de Oligonucleotídeos , Sequência de Bases , Neoplasias Cerebelares/patologia , Aberrações Cromossômicas , Análise Citogenética , Feminino , Marcadores Genéticos , Humanos , Perda de Heterozigosidade , Masculino , Meduloblastoma/patologia , Dados de Sequência Molecular , Proteínas Quinases S6 Ribossômicas 70-kDa/genética
10.
Am J Pathol ; 159(2): 661-9, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11485924

RESUMO

We investigated 67 meningothelial tumors (20 benign meningiomas, 34 atypical meningiomas, and 13 anaplastic meningiomas) for losses of genetic information from chromosome arms 1p and 9p, as well as for deletion, mutation, and expression of the tumor suppressor genes CDKN2A (p16(INKa)/MTS1), p14(ARF), CDKN2B (p15(INK4b)/MTS2) (all located at 9p21) and CDKN2C (1p32). Comparative genomic hybridization and microsatellite analysis showed losses on 1p in 11 anaplastic meningiomas (85%), 23 atypical meningiomas (68%), and 5 benign meningiomas (25%). One atypical meningioma with loss of heterozygosity on 1p carried a somatic CDKN2C mutation (c.202C>T: R68X). Losses on 9p were found in five anaplastic meningiomas (38%), six atypical meningiomas (18%), and one benign meningioma (5%). Six anaplastic meningiomas (46%) and one atypical meningioma (3%) showed homozygous deletions of the CDKN2A, p14(ARF), and CDKN2B genes. Two anaplastic meningiomas carried somatic point mutations in CDKN2A (c.262G>T: E88X and c.262G>A: E88K) and p14(ARF) (c.305G>T: G102V and c.305G>A: G102E). One anaplastic meningioma, three atypical meningiomas, and one benign meningioma without a demonstrated homozygous deletion or mutation of CDKN2A, p14(ARF), or CDKN2B lacked detectable transcripts from at least one of these genes. Hypermethylation of CDKN2A, p14(ARF), and CDKN2B could be demonstrated in one of these cases. Taken together, our results indicate that CDKN2C is rarely altered in meningiomas. However, the majority of anaplastic meningiomas either show homozygous deletions of CDKN2A, p14(ARF), and CDKN2B, mutations in CDKN2A and p14(ARF), or lack of expression of one or more of these genes. Thus, inactivation of the G(1)/S-phase cell-cycle checkpoint is an important aberration in anaplastic meningiomas.


Assuntos
Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Inibidor p16 de Quinase Dependente de Ciclina , Ciclinas/genética , Inibidores Enzimáticos , Genes Supressores de Tumor , Neoplasias Meníngeas/genética , Meningioma/genética , Proteínas/genética , Proteínas Supressoras de Tumor , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Criança , Mapeamento Cromossômico , Cromossomos Humanos Par 1 , Cromossomos Humanos Par 9 , Inibidor de Quinase Dependente de Ciclina p15 , Inibidor de Quinase Dependente de Ciclina p18 , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Feminino , Dosagem de Genes , Humanos , Perda de Heterozigosidade , Masculino , Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/cirurgia , Meningioma/patologia , Meningioma/cirurgia , Repetições de Microssatélites , Pessoa de Meia-Idade , Dados de Sequência Molecular , Proteína Supressora de Tumor p14ARF
11.
J Neurosurg ; 94(4): 605-11, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11302659

RESUMO

The authors report on a patient who had undergone resection of a left-sided temporal giant cell glioblastoma at the age of 69 years and who survived for more than 17 years. This man had not undergone postoperative radiotherapy or adjuvant chemotherapy. He died at the age of 86 years without clinical evidence of tumor recurrence. Histologically, the lesion was characterized by highly pleomorphic tumor cells (including bizarre multinucleated giant cells) with high mitotic activity, large necroses, and prominent mononuclear infiltration. A point mutation in the TP53 tumor suppressor gene (c.524G>A; R175H) and no epidermal growth factor receptor gene amplification were revealed on molecular genetic analysis. No diagnostic chromosomal imbalances were identified on comparative genomic hybridization, although the average ratio profile for chromosome 10 indicated loss of 10p15 in a subpopulation of tumor cells. This patient is exceptional because tumor resection, probably in conjunction with a marked antitumor immune response, apparently resulted in eradication of the lesion.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Neoplasias Encefálicas/cirurgia , Glioblastoma/fisiopatologia , Glioblastoma/cirurgia , Lobo Temporal , Idoso , Sequência de Bases/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Genes p53/genética , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Imuno-Histoquímica , Masculino , Biologia Molecular , Hibridização de Ácido Nucleico , Mutação Puntual/genética , Fatores de Tempo
12.
Int J Cancer ; 92(4): 551-4, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11304690

RESUMO

We have previously reported that losses of genomic material from the long arm of chromosome 18 are frequent in atypical and anaplastic meningiomas but rare in benign meningiomas. In the present study, we have investigated a series of 37 meningiomas for mutation and expression of 4 tumor suppressor genes (MADH2, MADH4, APM-1 and DCC) located at 18q21. Comparative genomic hybridization or loss of heterozygosity analysis showed losses on chromosome 18 that included sequences from 18q21 in 15 of 37 tumors. Mutation analysis of APM-1 revealed a missense mutation (c. 1819G>A: G607S) in 1 atypical meningioma. None of the tumors showed mutations of MADH2 and MADH4 or loss of detectable transcripts from MADH2, MADH4, APM-1 and DCC. In contrast to human brain tissue, normal leptomeninges and meningiomas showed preferential expression of a DCC splice variant lacking 60 base pairs from exon 17. Taken together, our data do not support a significant role for MADH2, MADH4, APM-1 and DCC alterations in the pathogenesis of meningiomas. The targeted gene that is inactivated in most meningiomas with 18q losses remains to be identified.


Assuntos
Neoplasias Encefálicas/genética , Moléculas de Adesão Celular/genética , Aberrações Cromossômicas , Cromossomos Humanos Par 18/genética , Proteínas de Ligação a DNA/genética , Peptídeos e Proteínas de Sinalização Intercelular , Meningioma/genética , Proteínas/genética , Transativadores/genética , Proteínas Supressoras de Tumor , Adiponectina , Adolescente , Adulto , Idoso , Alelos , Processamento Alternativo , Encéfalo/metabolismo , Criança , Receptor DCC , Análise Mutacional de DNA , Éxons , Feminino , Humanos , Perda de Heterozigosidade , Masculino , Meninges/metabolismo , Pessoa de Meia-Idade , Mutação , Mutação de Sentido Incorreto , Hibridização de Ácido Nucleico , Polimorfismo Conformacional de Fita Simples , RNA Mensageiro/metabolismo , Receptores de Superfície Celular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad2 , Proteína Smad4
13.
Int J Cancer ; 91(2): 213-8, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11146447

RESUMO

The median survival for human malignant glioma patients treated with neurosurgery and postoperative radiotherapy does not exceed one year. Only a minority of patients benefit from adjuvant chemotherapy. It was the aim of our study to determine which genomic alterations in malignant gliomas modulate the sensitivity to chemotherapy or cytotoxic cytokines such as CD95 ligand (CD95L) or Apo2L/tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL). Therefore, we analyzed 12 human malignant glioma cell lines for chromosomal gains and losses by comparative genomic hybridization (CGH). The gains most commonly identified were on chromosomes 7q, 19, 1, and 20q, whereas the most frequent losses were on 13q, 11q, 18q, and 4q. By comparison with previously published data on this panel of glioma cell lines1112, we defined candidate regions which may carry genes responsible for sensitivity to chemotherapy or cytotoxic cytokines. All but one of the chromosomal regions associated with response to chemotherapy, i.e. 1p12, 3p21, 11p11.2-p13, 12q23, 17p11. 2-p13, were different from those associated with response to cytotoxic cytokines, i.e. lp12, 1q22, 12q12-q21. Genomic regions known to harbor major candidate genes, including genes encoding death ligands, death receptors, caspases and BCL-2 family proteins, were not found to be imbalanced. In addition, we identified 5q13-q14, 5q34, 10p11.2, 9q21-q34 as genomic regions associated with the proliferative activity of malignant glioma cell lines. Cell lines with gain on proximal 5q, where CCNB1 and CCNH reside, showed an increased growth rate, suggesting that cyclins activating cdc2, the dominant G2/M phase kinase, may play a role in glioma tumorigenes.


Assuntos
Aberrações Cromossômicas , Citocinas/farmacologia , Glioma/genética , Apoptose/efeitos dos fármacos , Ciclo Celular , Cicloeximida/farmacologia , Resistencia a Medicamentos Antineoplásicos , Glioma/tratamento farmacológico , Glioma/patologia , Humanos , Hibridização de Ácido Nucleico , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/análise
14.
Am J Pathol ; 157(2): 571-8, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10934159

RESUMO

As data on the genomic alterations in hepatoblastoma (HB) are limited, 34 HB tumors and three HB cell lines were screened for DNA copy number changes by comparative genomic hybridization. The average number of chromosomal imbalances per tumor was 2.3 +/- 0.5 (mean +/- SEM) with gains sevenfold more frequent than losses. The most frequent gains of chromosomal material in HB tumors were on 2q (44%), 1q (41%), 2p (29%), 20 (24%), 22q (18%), 8q (15%), 8p and 12q (9% each), as well as 7q, 12p, and 17 (6% each) and the only recurrent loss was on 4q in 12% of cases. Highly amplified sequences were identified in four tumors and mapped to 2q24 in two cases, to 8q in two cases (once to 8q11.2-q13 and once to 8q11.2-q21.3) as well as to 10q24-q26 in one case. In one cell line, highly amplified DNA sequences were mapped to 7p and 8q. Comparison to previously published data on this series of HB revealed that the number of chromosomal imbalances was significantly higher in HB tumors with loss of heterozygosity on 11p (P = 0.03), whereas in five of 10 HB biopsies without chromosomal imbalances, beta-catenin gene mutations were found. HB patients were divided into a good (no evidence of disease) and a poor (died of disease) outcome group according to their clinical course after standard therapy. Two alterations were found to be significantly associated with poor outcome: gain on 8q (P = 0.007) and gain on 20 (P = 0.009). In summary, our analysis allowed the identification of gains on chromosomes 1q and 2 as hallmark DNA copy number changes in HB with 2q24 as a critical chromosomal band. Furthermore, this study provided evidence that gains on 8q and 20 play a role as markers of prognostic significance in HB.


Assuntos
Aberrações Cromossômicas , Hepatoblastoma/genética , Neoplasias Hepáticas/genética , Transativadores , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia , Criança , Pré-Escolar , Cromossomos Humanos Par 20/genética , Cromossomos Humanos Par 8/genética , Proteínas do Citoesqueleto/genética , Feminino , Hepatoblastoma/mortalidade , Hepatoblastoma/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/patologia , Perda de Heterozigosidade , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas , beta Catenina
15.
Brain Pathol ; 10(1): 73-84, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10668897

RESUMO

We performed a genome wide screening for genomic alterations on a series of 19 sporadic primary central nervous system lymphomas (PCNSL) of the diffuse large B-cell type by comparative genomic hybridization (CGH). The tumors were additionally analyzed for amplification and rearrangement of the BCL2 gene at 18q21 as well as for mutation of the recently cloned BCL10 gene at 1p22. Eighteen tumors showed genomic imbalances on CGH analysis. On average, 2.1 losses and 4.7 gains were detected per tumor. The chromosome arm most frequently affected by losses of genomic material was 6q (47%) with a commonly deleted region mapping to 6q21-q22. The most frequent gains involved chromosome arms 12q (63%), 18q and 22q (37% each), as well as 1q, 9q, 11q, 12p, 16p and 17q (26% each). High-level amplifications were mapped to 9p23-p24 (1 tumor) and to 18q21-q23 (2 tumors). However, PCR-based analysis, Southern blot analysis and high-resolution matrix-CGH of the BCL2 gene revealed neither evidence for amplification nor for genetic rearrangement. Mutational analysis of BCL10 in 16 PCNSL identified four distinct sequence polymorphisms but no mutation. Taken together, our data do not support a role of BCL2 rearrangement/amplification and BCL10 mutation in PCNSL but indicate a number of novel chromosomal regions that likely carry yet unknown tumor suppressor genes or proto-oncogenes involved in the pathogenesis of these tumors.


Assuntos
Neoplasias do Sistema Nervoso Central/genética , Aberrações Cromossômicas/genética , Linfoma de Células B/genética , Linfoma Difuso de Grandes Células B/genética , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Hibridização de Ácido Nucleico
16.
Brain Pathol ; 9(4): 617-26, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10517500

RESUMO

Chordoid glioma of the third ventricle was recently reported as a novel tumor entity of the central nervous system with characteristic clinical and histopathological features (Brat et al., J Neuropathol Exp Neurol 57: 283-290, 1998). Here, we report on a histopathological, immunohistochemical and molecular genetic analysis of five cases of this rare neoplasm. All tumors were immunohistochemically investigated for the expression of various differentiation antigens, the proliferation marker Ki-67, and a panel of selected proto-oncogene and tumor suppressor gene products. These studies revealed a strong expression of GFAP, vimentin, and CD34. In addition, most tumors contained small fractions of neoplastic cells immunoreactive for epithelial membrane antigen, S-100 protein, or cytokeratins. The percentage of Ki-67 positive cells was generally low (<5%). All tumors showed immunoreactivity for the epidermal growth factor receptor and schwannomin/merlin. There was no nuclear accumulation of the p53, p21 (Waf-1) and Mdm2 proteins. To examine genomic alterations associated with the development of chordoid gliomas, we screened 4 tumors by comparative genomic hybridization (CGH) analysis. No chromosomal imbalances were detected. More focussed molecular genetic analyses revealed neither aberrations of the TP53 and CDKN2A tumor suppressor genes nor amplification of the EGFR, CDK4, and MDM2 proto-oncogenes. Our data strongly support the hypothesis that chordoid glioma of the third ventricle constitutes a novel tumor entity characterized by distinct morphological and immunohistochemical features, as well as a lack of chromosomal and genetic alterations commonly found in other types of gliomas or in meningiomas.


Assuntos
Neoplasias do Plexo Corióideo/genética , Glioma/genética , Adulto , Idoso , Neoplasias do Plexo Corióideo/patologia , Feminino , Deleção de Genes , Genes p16/genética , Glioma/patologia , Humanos , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Hibridização de Ácido Nucleico , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Proto-Oncogene Mas , Terceiro Ventrículo/patologia , Tomografia Computadorizada por Raios X
17.
Brain Pathol ; 9(3): 435-42; discussion 432-3, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10416984

RESUMO

Malignant gliomas frequently show genetic aberrations of genes coding for cell cycle regulatory proteins involved in the control of G1/S phase transition. These include mutation and/or deletion of the retinoblastoma (RB1) gene, homozygous deletion of the CDKN2A and CDKN2B genes, as well as amplification and overexpression of the CDK4 and CDK6 genes. The D-type cyclins (cyclin D1, D2, and D3) promote cell cycle progression from G1 to S phase by binding to and activating the cyclin dependent kinases Cdk4 and Cdk6. Here, we have investigated a series of 110 primary malignant gliomas and 8 glioma cell lines for amplification and expression of the D-type cyclin genes CCND1 (11q13), CCND2 (12p13), and CCND3 (6p21). We found the CCND1 gene amplified and overexpressed in one anaplastic astrocytoma of our tumor series. Two glioblastomas and one anaplastic astrocytoma showed CCND2 gene amplification, but lacked significant overexpression of CCND2 transcripts. Amplification and overexpression of the CCND3 gene was detected in the glioblastoma cell line CCF-STTG1, as well as in one primary glioblastoma and in the sarcomatous component of one gliosarcoma. Our data thus suggest that amplification and increased expression of CCND1 and CCND3 contribute to the loss of cell cycle control in a small fraction of human malignant gliomas.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Ciclinas/biossíntese , Ciclinas/genética , Glioma/genética , Glioma/metabolismo , Southern Blotting , Ciclo Celular/genética , Ciclina D , Ciclina D1/biossíntese , Ciclina D1/genética , Ciclina D2 , Ciclina D3 , Amplificação de Genes , Glioblastoma/genética , Glioblastoma/metabolismo , Gliossarcoma/genética , Gliossarcoma/metabolismo , Humanos , Imuno-Histoquímica , Hibridização de Ácido Nucleico , Reação em Cadeia da Polimerase
18.
Genes Chromosomes Cancer ; 24(1): 87-93, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9892114

RESUMO

Somatic genetic alterations of 9p have been seen in a wide range of human cancers, including breast cancer. Loss of heterozygosity analysis of primary breast cancer tumors has revealed a high frequency of deletion of DNA from 9p21-22 encompassing the MTSI (P16/CDKN2A) gene. We report the approximately tenfold increase in copy number of DNA from 9p23-24, which is far distal to P16/CDKN2A in female breast cancer cell line COLO 824, as revealed by fluorescence in situ hybridization, comparative genomic hybridization, and microsatellite analysis. Amplification of DNA has been reported previously to encompass multiple sites of the genome of the breast cancer cell, but increase in DNA copy number has not been seen in distal 9p.


Assuntos
Aneuploidia , Neoplasias da Mama/genética , Cromossomos Humanos Par 9/genética , Evolução Molecular , Feminino , Dosagem de Genes , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Repetições de Microssatélites , Hibridização de Ácido Nucleico , Células Tumorais Cultivadas
19.
Am J Pathol ; 153(1): 295-303, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9665491

RESUMO

Biopsies routinely performed for the histopathological diagnosis of oral epithelial lesions before treatment were screened for chromosomal imbalances by comparative genomic hybridization. Comparative genomic hybridization was performed on 12 oral premalignant lesions (OPLs; dysplasias and carcinomas in situ) and 14 oral squamous cell carcinomas (OSCCs). Eight biopsies displayed areas of different histopathological appearance, so that OPLs and OSCCs from the same patient were analyzed. To avoid contamination with nonneoplastic cells, defined cell populations were isolated by micromanipulation with a glass needle. Before comparative genomic hybridization analysis, universal DNA amplification was performed using the DOP-polymerase chain reaction protocol. In the 14 OSCCs examined, the average number of chromosomal imbalances was significantly higher than in the 12 OPLs (mean +/- SEM: 11.9 +/- 1.9 versus 3.2 +/- 1.2; P = 0.003). The DNA copy number changes identified in more than one OPL were gains on 8q (3 of 12) and 16p (2 of 12), as well as losses on 3p (5 of 12); 5q (4 of 12); 13q (3 of 12); and 4q, 8p, and 9p (2 of 12 each). In more than 30% of OSCCs, gains of chromosomal material were identified on 20q (8 of 14); 8q, 11q, 22q (7 of 14 each); 3q, 15q, and 17p (6 of 14 each); and 14q, 17q, and 20p (5 of 14 each), and losses were identified on 3p and 4q (9 of 14 each), 5q (7 of 14), 13q (6 of 14), and 2q and 9p (5 of 14 each). These results were validated by positive and negative control comparative genomic hybridization experiments and microsatellite analysis for the detection of allelic loss. The vast majority of genomic alterations found in OPLs were again identified in OSCCs from the same biopsy, supporting the hypothesis that multiple lesions in the same patient are clonally related. In summary, we show that comprehensive information on the genomic alterations in oral epithelial lesions can be obtained from small biopsies. Such data may identify prognostic indicators that could eventually assist in designing therapeutic strategies.


Assuntos
Neoplasias Bucais/genética , Lesões Pré-Cancerosas/genética , Adulto , Idoso , Biópsia , Carcinoma in Situ/genética , Carcinoma in Situ/patologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Aberrações Cromossômicas , Transtornos Cromossômicos , DNA de Neoplasias/análise , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mucosa Bucal/química , Mucosa Bucal/patologia , Neoplasias Bucais/patologia , Hibridização de Ácido Nucleico , Reação em Cadeia da Polimerase , Lesões Pré-Cancerosas/patologia
20.
Genomics ; 50(3): 341-5, 1998 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9676428

RESUMO

The glycine receptor of the human CNS comprises ligand-binding alpha 1 and structural beta subunits encoded by the GLRA1 and GLRB genes, respectively. Screening of a human hippocampal cDNA library resulted in the identification of the novel subunit transcript beta B, differing in the 5'-UTR. Analysis of the genomic organization of GLRB showed that the coding region is distributed over nine exons, highly homologous to the GLRA1 gene. By in situ hybridization, the chromosomal localization of GLRB was refined to band 4q31.3. Based on the identical phenotypes of mouse lines carrying mutant alleles of the alpha 1 and beta subunit genes, GLRB was assumed to be a candidate gene for those cases of hyperekplexia that cannot be associated with mutations of GLRA1. Therefore, flanking intronic sequences were determined, and DNA samples from more than 30 index patients were subjected to SSCP screening of the entire GLRB coding region. A polymorphism in exon 8 was found both in the normal population and in families affected by hyperekplexia, although no coding mutation was detectable.


Assuntos
Cromossomos Humanos Par 4 , Éxons , Polimorfismo Genético , Receptores de Glicina/genética , Alelos , Sequência de Aminoácidos , Sequência de Bases , Mapeamento Cromossômico , DNA Complementar , Biblioteca Genômica , Hipocampo , Humanos , Hibridização in Situ Fluorescente , Íntrons , Dados de Sequência Molecular , Doenças do Sistema Nervoso/genética , Mutação Puntual , Polimorfismo Conformacional de Fita Simples , Reflexo de Sobressalto/genética , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...