Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Oncogene ; 20(36): 4917-25, 2001 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-11526476

RESUMO

Multiple endocrine neoplasia type 1 is an autosomal dominant tumor syndrome. Manifestations include neoplasms of the parathyroid glands, enteropancreatic neuroendocrine cells, and the anterior pituitary gland. The MEN1 tumor suppressor gene encodes menin, a 610 amino acid nuclear protein without sequence homology to other proteins. To elucidate menin function, we used immunoprecipitation to identify interacting proteins. The NF-kappaB proteins p50, p52 and p65 were found to interact specifically and directly with menin in vitro and in vivo. The region of NF-kappaB proteins sufficient for binding to menin is the N-terminus. Furthermore, amino acids 305-381 of menin are essential for this binding. Menin represses p65-mediated transcriptional activation on NF-kappaB sites in a dose-dependent and specific manner. Also, PMA (phorbol 12-myristate 13-acetate)-stimulated NF-kappaB activation is suppressed by menin. These observations suggest that menin's ability to interact with NF-kappaB proteins and its modulation of NF-kappaB transactivation contribute to menin's tumor suppressor function.


Assuntos
Genes Supressores de Tumor , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas de Neoplasias/fisiologia , Proteínas Proto-Oncogênicas , Animais , Células COS , Linhagem Celular , Glutationa Transferase/química , Células HeLa , Humanos , NF-kappa B/química , Proteínas de Neoplasias/química , Proteínas de Neoplasias/imunologia , Testes de Precipitina , Estrutura Terciária de Proteína , Acetato de Tetradecanoilforbol/farmacologia , Ativação Transcricional
2.
Gene ; 263(1-2): 31-8, 2001 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-11223240

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) is a familial cancer syndrome characterized by tumors of the parathyroid, entero-pancreatic neuroendocrine and pituitary tissues and caused by inactivating mutations in the MEN1 gene. Menin, the 610-amino acid nuclear protein encoded by MEN1, binds to the transcription factor JunD and can repress JunD-induced transcription. We report here the identification of a MEN1 ortholog in Drosophila melanogaster, Menin1, that encodes a 763 amino acid protein sharing 46% identity with human menin. Additionally, 69% of the missense mutations and in-frame deletions reported in MEN1 patients appear in amino acid residues that are identical in the Drosophila and human protein, suggesting the importance of the conserved regions. Drosophila Menin1 gene transcripts use alternative polyadenylation sites resulting in 4.3 and 5-kb messages. The 4.3-kb transcript appears to be largely maternal, while the 5-kb transcript appears mainly zygotic. The binding of Drosophila menin to human JunD or Drosophila Jun could not be demonstrated by the yeast two-hybrid analysis. The identification of the MEN1 ortholog from Drosophila melanogaster will provide an opportunity to utilize Drosophila genetics to enhance our understanding of the function of human menin.


Assuntos
Drosophila melanogaster/genética , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogênicas , Sequência de Aminoácidos , Animais , Northern Blotting , DNA Complementar/química , DNA Complementar/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/crescimento & desenvolvimento , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário , Éxons , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos/genética , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Humanos , Hibridização In Situ , Íntrons , Masculino , Camundongos , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/genética , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência , Transcrição Gênica , Técnicas do Sistema de Duplo-Híbrido , Peixe-Zebra
3.
Mol Cell Biol ; 20(14): 5087-95, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10866665

RESUMO

Snail/Slug family proteins have been identified in diverse species of both vertebrates and invertebrates. The proteins contain four to six zinc fingers and function as DNA-binding transcriptional regulators. Various members of the family have been demonstrated to regulate cell movement, neural cell fate, left-right asymmetry, cell cycle, and apoptosis. However, the molecular mechanisms of how these regulators function and the target genes involved are largely unknown. In this report, we demonstrate that human Slug (hSlug) is a repressor and modulates both activator-dependent and basal transcription. The repression depends on the C-terminal DNA-binding zinc fingers and on a separable repression domain located in the N terminus. This domain may recruit histone deacetylases to modify the chromatin and effect repression. Protein localization study demonstrates that hSlug is present in discrete foci in the nucleus. This subnuclear pattern does not colocalize with the PML foci or the coiled bodies. Instead, the hSlug foci overlap extensively with areas of the SC-35 staining, some of which have been suggested to be sites of active splicing or transcription. These results lead us to postulate that hSlug localizes to target promoters, where activation occurs, to repress basal and activator-mediated transcription.


Assuntos
Núcleo Celular/metabolismo , Proteínas Nucleares , Proteínas Repressoras/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Sequência de Bases , Sítios de Ligação , Núcleo Celular/genética , Cromatina/metabolismo , Sequência Conservada , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Imunofluorescência , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Dados de Sequência Molecular , Proteínas de Neoplasias/imunologia , Proteínas de Neoplasias/metabolismo , Proteína da Leucemia Promielocítica , Splicing de RNA , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Deleção de Sequência , Fatores de Transcrição da Família Snail , Fatores de Transcrição/imunologia , Proteínas Supressoras de Tumor , Dedos de Zinco
4.
Recent Prog Horm Res ; 54: 397-438; discussion 438-9, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10548885

RESUMO

MEN1 is a syndrome of parathyroid adenomas, gastrinomas, prolactinomas, and other endocrine tumors. Collagenomas and facial angiofibromas are newly recognized but common skin expressions. Many tumors in MEN1 are benign; however, many entero-pancreatic neuroendocrine tumors and foregut carcinoid tumors are malignant. MEN1 is thus the expression of a cancer gene but without available prevention or cure for malignancy. Hereditary (as compared to sporadic) endocrine tumors show early onset age and multiplicity, because each cell of the body has "one hit" by inheritance. Multiple neoplasia syndromes with endocrine tumor(s) all include nonendocrine components; their known defective genes seem mainly to disturb cell accumulation. Hereditary neoplasia/hyperplasia of one endocrine tissue reflects a defect that is tissue selective and directed at cell secretion. Though the hereditary endocrine neoplasias are rare, most of their identified genes also contribute to common sporadic endocrine neoplasms. Hereditary tumors may be caused by activation of an oncogene (e.g., RET) or, more often, by inactivation of a tumor suppressor gene (e.g., P53, MEN1). Recently, MEN1 was identified by positional cloning. This strategy included narrowing the gene candidate interval, identifying many or all genes in that interval, and testing the newly identified candidate genes for mutation in MEN1 cases. MEN1 was identified because it showed mutation in 14 of 15 MEN1 cases. NIH testing showed germline MEN1 mutations in 47 of 50 MEN1 index cases and in seven of eight cases with sporadic MEN1. Despite proven capacity to find germline MEN1 mutation, NIH testing found no MEN1 mutation among five families with isolated hyperparathyroidism, suggesting that this often arises from mutation of other gene(s). Analogous studies in Japan found that familial isolated pituitary tumors also did not show MEN1 germline mutation. MEN1 mutation testing can now be considered for cases of MEN1 and its phenocopies and for asymptomatic members of families with known MEN1 mutation. Germline MEN1 testing does not have the urgency of RET testing in MEN2a and 2b, as MEN1 testing does not commonly lead to an important intervention. Somatic MEN1 mutation was found in sporadic tumors: parathyroid adenoma (21%), gastrinoma (33%), insulinoma (17%), and bronchial carcinoid (36%). For each of these, MEN1 was the known gene most frequently mutated. MEN1 has a widely expressed mRNA that encodes a protein (menin) of 610 amino acids. The protein sequence is not informative about domains or functions. The protein was mainly nuclear. Menin binds to JunD, an AP-1 transcription factor, inhibiting JunD's activation of transcription. Most of the germline and somatic MEN1 mutations predict truncation of menin, a likely destructive change. Inactivating MEN1 mutations in germline and in sporadic neoplasms support prior predictions that MEN1 is a tumor suppressor gene. Germline MEN1 mutation underlies all or most cases of MEN1 (familial or sporadic). Somatic MEN1 mutation is the most common gene mutation in many sporadic endocrine tumor types.


Assuntos
Neoplasia Endócrina Múltipla Tipo 1/fisiopatologia , Sequência de Aminoácidos , Hormônios/metabolismo , Humanos , Dados de Sequência Molecular , Neoplasia Endócrina Múltipla Tipo 1/epidemiologia , Neoplasia Endócrina Múltipla Tipo 1/terapia , Linhagem , Prevalência , Taxa Secretória
5.
Autoimmunity ; 29(4): 311-22, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10433087

RESUMO

Circulating auto-antibodies (Abs) against lens antigens (Ags) are highly prevalent in patients with cataract, but their origin and pathogenic significance are unknown. We hypothesized that Abs raised after exposure to infectious microbes could cross-react with lens Ags. To test this hypothesis, we generated a monoclonal Ab to human betaA3-crystallin. Epitope analysis indicated that the ETQAE sequence in the N-terminus region of betaA3-crystallin was critical for mounting a humoral response. Similar sequences were found in three microbial Ags. Mice injected with a microbial oligopeptide containing ETQAE emulsified with complete Freund's adjuvant (CFA) raised Abs which cross-reacted with betaA3-crystallin and developed lens epithelial cell (LEC) damage in vitro. We also genetically engineered an betaA3-crystallin-expressing E. coli. Mice immunized with the recombinant E. coli developed LEC damage. These results support the hypothesis that exposure to microbes having Ags homologous to self Ags can trigger a humoral immune response that leads to LEC damage in mice.


Assuntos
Cristalinas/imunologia , Cristalino/imunologia , Sequência de Aminoácidos , Animais , Autoimunidade , Reações Cruzadas , Cristalinas/genética , Células Epiteliais/imunologia , Células Epiteliais/patologia , Epitopos/genética , Epitopos/imunologia , Humanos , Imunização , Cristalino/patologia , Camundongos , Dados de Sequência Molecular , Oligopeptídeos/genética , Oligopeptídeos/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Cadeia A de beta-Cristalina
6.
Bone ; 25(1): 119-22, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10423035

RESUMO

Multiple endocrine neoplasia type 1 (MENI) is a promising model to understand endocrine and other tumors. Its most common endocrine expressions are tumors of parathyroids, entero-pancreatic neuro-endocrine tissue, and anterior pituitary. Recently, collagenomas and multiple angiofibromas of the dermis also have been recognized as very common. MEN1 can be characterized from different perspectives: (a) as a hormone (parathyroid hormone, gastrin, prolactin, etc.) excess syndrome with excellent therapeutic options; (b) as a syndrome with sometimes lethal outcomes from malignancy of entero-pancreatic neuro-endocrine or foregut carcinoid tissues; or (c) as a disorder than can give insight about cell regulation in the endocrine, the dermal, and perhaps other tissue systems. The MEN1 gene was identified recently by positional cloning, a comprehensive strategy of narrowing the candidate interval and evaluating all or most genes in that interval. This discovery has opened new approaches to basic and clinical issues. Germline MEN1 mutations have been identified in most MEN1 families. Germline MENI mutations were generally not found in families with isolated hyperparathyroidism or with isolated pituitary tumor. Thus, studies with the MENI gene helped establish that mutation of other gene(s) is likely causative of these two MEN1 phenocopies. MEN1 proved to be the gene most frequent L4 mutated in common-variety, nonhereditary parathyroid tumor, gastrinoma, insulinoma, or bronchial carcinoid. For example, in common-variety parathyroid tumors, mutation of several other genes (such as cyclin D1 and P53) has been found, but much less frequently than MEN1 mutation. The majority of germline and somatic MEN1 mutations predicted truncation of the encoded protein (menin). Such inactivating mutations strongly supported prior predictions that MEN1 is a tumor suppressor gene insofar as stepwise mutational inactivation of both copies can release a cell from normal growth suppression. Menin is principally a nuclear protein; menin interacts with junD. Future studies, such as discovery of menin's metabolic pathway, could lead to new opportunities in cell biology and in tumor therapy.


Assuntos
Neoplasias das Glândulas Endócrinas/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasia Endócrina Múltipla Tipo 1/genética , Proteínas Proto-Oncogênicas , Genótipo , Mutação em Linhagem Germinativa , Humanos , Proteínas de Neoplasias/fisiologia , Linhagem , Fenótipo
7.
Mamm Genome ; 10(6): 592-6, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10341092

RESUMO

The mouse homolog of the human MEN1 gene, which is defective in a dominant familial cancer syndrome, multiple endocrine neoplasia type 1 (MEN1), has been identified and characterized. The mouse Men1 transcript contains an open reading frame encoding a protein of 611 amino acids which has 97% identity and 98% similarity to human menin. Sequence of the entire Men1 gene (9.3 kb) was assembled, revealing 10 exons, with exon 1 being non-coding; a polymorphic tetranucleotide repeat was located in the 5'- flanking region. The exon-intron organization and the size of the coding exons 2-9 were well conserved between the human and mouse genes. Fluorescence in situ hybridization localized the Men1 gene to mouse Chromosome (Chr) 19, a region known to be syntenic to human Chr 11q13, the locus for the MEN1 gene. Northern analysis indicated two messages-2.7 kb and 3.1 kb-expressed in all stages of the embryo analyzed and in all eight adult tissues tested. The larger transcript differs from the smaller by the inclusion of an unspliced intron 1. Whole-mount in situ hybridization of 10.5-day and 11.5-day embryos showed ubiquitous expression of Men1 RNA. Western analysis with antibodies raised against a conserved C-terminal peptide identified an approximately 67-kDa protein in the lysates of adult mouse brain, kidney, liver, pancreas, and spleen tissues, consistent with the size of human menin. The levels of mouse menin do not appear to fluctuate during the cell cycle.


Assuntos
Mapeamento Cromossômico , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogênicas , Sequência de Aminoácidos , Animais , Northern Blotting , Ciclo Celular/genética , Clonagem Molecular , DNA Complementar , Desoxirribonucleases de Sítio Específico do Tipo II/genética , Embrião de Mamíferos/fisiologia , Éxons , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização In Situ/métodos , Hibridização in Situ Fluorescente , Íntrons , Camundongos , Dados de Sequência Molecular , Proteínas de Neoplasias/metabolismo , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
8.
Cell ; 96(1): 143-52, 1999 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-9989505

RESUMO

MEN1 is a tumor suppressor gene that encodes a 610 amino acid nuclear protein (menin) of previously unknown function. Using a yeast two-hybrid screen with menin as the bait, we have identified the transcription factor JunD as a direct menin-interacting partner. Menin did not interact directly with other Jun and Fos family members. The menin-JunD interaction was confirmed in vitro and in vivo. Menin repressed transcriptional activation mediated by JunD fused to the Gal4 DNA-binding domain from a Gal4 responsive reporter, or by JunD from an AP1-responsive reporter. Several naturally occurring and clustered MEN1 missense mutations disrupted menin interaction with JunD. These observations suggest that menin's tumor suppressor function involves direct binding to JunD and inhibition of JunD activated transcription.


Assuntos
Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional , Animais , Sítios de Ligação , Células HeLa , Humanos , Mutação de Sentido Incorreto , Proteínas de Neoplasias/genética , Hibridização de Ácido Nucleico , Testes de Precipitina , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Leveduras
9.
Hum Mutat ; 12(2): 75-82, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9671267

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant disorder that manifests as varying combinations of tumors of endocrine and other tissues (parathyroids, pancreatic islets, duodenal endocrine cells, the anterior pituitary and others). The MEN1 gene is on chromosome 11q13; it was recently identified by positional cloning. We previously reported 32 different germline mutations in 47 of the 50 familial MEN1 probands studied at the NIH. Eight different germline MEN1 mutations were encountered repeatedly in two or more apparently unrelated families. We analyzed the haplotypes of families with recurrent MEN1 mutations with seven polymorphic markers in the 11q13 region surrounding the MEN1 gene (from D11S1883 to D11S4908). Disease haplotypes were inferred from germline DNA and also from tumors with 11ql3 loss of heterozygosity. Two different disease haplotype cores were shared by apparently unrelated families for two mutations in exon 2 (five families with 416delC and six families with 512delC). These two repeat mutations were associated with the two founder effects that we reported in a prior haplotype analysis. The disease haplotypes for each of the other six repeat mutations (seen twice each) were discordant, suggesting independent origins of these recurrent mutations. Most of the MEN1 germline mutations including all of those recurring independently occur in regions of CpG/CpNpG, short DNA repeats or single nucleotide repeat motifs. In conclusion, recurring germline mutations account for about half of the mutations in North American MEN1 families. They result from either founder effects or independent occurrence of one mutation more than one time.


Assuntos
Mutação em Linhagem Germinativa/genética , Neoplasia Endócrina Múltipla Tipo 1/genética , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogênicas , Ilhas de CpG , DNA , Saúde da Família , Feminino , Efeito Fundador , Genes Supressores de Tumor , Marcadores Genéticos , Haplótipos/genética , Humanos , Masculino , Mutação Puntual , Polimorfismo Genético , Sequências Repetitivas de Ácido Nucleico , Análise de Sequência de DNA
10.
J Intern Med ; 243(6): 433-9, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9681840

RESUMO

For nearly a decade since the mapping of the multiple endocrine neoplasia type 1 (MEN1) locus to 11q13 and the suggestion that it is a tumour suppressor gene, efforts have been made to identify the gene responsible for this familial cancer syndrome. Recently, we have identified the MEN1 gene by the positional cloning approach. This effort involved construction of a 2.8-Mb physical map (D11S480-D11S913) based primarily on a bacterial clone contig. Using these resources, 20 new polymorphic markers were isolated which helped to reduce the interval for candidate genes by haplotype analysis in families and by loss of heterozygosity (LOH) studies in approximately 200 tumours, utilizing laser-assisted microdissection to obtain tumour cells with minimal or no admixture by normal cells. The interval was narrowed by LOH to only 300 kb, and nearly 20 new transcripts that map to this region of 11q13 were isolated and characterized. One of the transcripts was found by dideoxyfingerprinting and cycle sequencing to harbour deleterious germline mutations in affected individuals from MEN-1 kindreds and therefore identified as the MEN1 gene. The type of germline mutations and the identification of mutations in sporadic tumours support the Knudson's two-hit model of tumorigenesis for MEN-1. Efforts are being made to identify the function of the MEN1 gene-encoded protein, menin, and to study its role in tumorigenesis.


Assuntos
Clonagem Molecular/métodos , Neoplasia Endócrina Múltipla Tipo 1/genética , Sequência de Aminoácidos , Sequência de Bases , Cromossomos Humanos Par 11/genética , Ligação Genética , Marcadores Genéticos , Haplótipos , Humanos , Perda de Heterozigosidade/genética , Dados de Sequência Molecular , Mutação
11.
J Intern Med ; 243(6): 447-53, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9681842

RESUMO

Dideoxyfingerprinting was used to screen for germline and somatic MEN1 mutations. This method, applied to a panel of germline DNA from 15 probands with multiple endocrine neoplasia type 1 (MEN-1), allowed confident discovery of the MEN1 gene. Germline MEN1 mutation has been found in 47 out of 50 probands with familial MEN-1, in 7 out of 8 cases with sporadic MEN-1, and in 1 out of 3 cases with atypical sporadic MEN-1. Germline MEN1 mutation was not found in any of five probands with familial hyperparathyroidism. Somatic MEN1 mutations were found in 7 out of 33 parathyroid tumours not associated with MEN-1. Allowing for repeating mutations, a total of 47 different germline or somatic MEN1 mutations have been identified. Most predict inactivation of the encoded 'menin' protein. supporting expectations that MEN1 is a tumour suppressor gene. The 16 observed missense mutations were distributed across the gene, suggesting that many domains are important to its as yet unknown functions.


Assuntos
Mutação em Linhagem Germinativa , Neoplasia Endócrina Múltipla Tipo 1/genética , Códon/genética , DNA de Neoplasias/genética , Genes Supressores de Tumor/genética , Humanos , Hiperparatireoidismo/genética , Neoplasias das Paratireoides/genética
12.
Mol Genet Metab ; 63(2): 151-5, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9562970

RESUMO

We analyzed constitutional and tumor DNA from 27 MEN1 kindreds not known to be related to each other. Disease allele haplotypes were constructed for each pedigree based on shared alleles from two or more affected members and from determination of allelic loss patterns in their tumors. Analysis of disease allele haplotypes showed unexpected linkage disequilibrium at marker PYGM. Further haplotype analysis indicated this could be explained by the presence of two founder chromosomes, one in four families, the other in three. A shared disease haplotype was not observed among two MEN1 kindreds with the prolactinoma phenotype of MEN1.


Assuntos
Alelos , Cromossomos Humanos Par 11/genética , Efeito Fundador , Neoplasia Endócrina Múltipla Tipo 1/genética , Marcadores Genéticos , Haplótipos , Humanos , América do Norte , Linhagem , Reação em Cadeia da Polimerase , Polimorfismo Genético
13.
Hum Mutat ; 11(4): 264-9, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9554741

RESUMO

Familial multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant disorder with affected individuals developing parathyroid, gastrointestinal (GI) endocrine, and anterior pituitary tumors. Four large kindreds from the Burin peninsula/Fortune Bay area of Newfoundland with prominent features of prolactinomas, carcinoids, and parathyroid tumors (referred to as MEN1Burin) have been described, and they show linkage to 11q13, the same locus as that of MEN1. Haplotype analysis with 16 polymorphic markers now reveals that representative affected individuals from all four families share a common haplotype over a 2.5 Mb region. A nonsense mutation in the MEN1 gene has been found to be responsible for the disease in the affected members in all four of the MEN1Burin families, providing convincing evidence of a common founder.


Assuntos
Neoplasia Endócrina Múltipla Tipo 1/genética , Neoplasias Hipofisárias/genética , Mutação Puntual , Prolactinoma/genética , Alelos , Sequência de Aminoácidos , Sequência de Bases , Cromossomos Humanos Par 11/genética , DNA/genética , Feminino , Efeito Fundador , Ligação Genética , Marcadores Genéticos , Haplótipos , Humanos , Masculino , Dados de Sequência Molecular , Terra Nova e Labrador , Linhagem , Reação em Cadeia da Polimerase
14.
Proc Natl Acad Sci U S A ; 95(4): 1630-4, 1998 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-9465067

RESUMO

The MEN1 gene, mutations in which are responsible for multiple endocrine neoplasia type 1 (MEN1), encodes a 610-amino acid protein, denoted menin. The amino acid sequence of this putative tumor suppressor offers no clue to the function or subcellular location of the protein. We report herein, based on immunofluorescence, Western blotting of subcellular fractions, and epitope tagging with enhanced green fluorescent protein, that menin is located primarily in the nucleus. Enhanced green fluorescent protein-tagged menin deletion constructs identify at least two independent nuclear localization signals (NLS), both located in the C-terminal fourth of the protein. Among the 68 known independent disease-associated mutations, none of the 22 missense and 3 in-frame deletions affect either of the putative NLS sequences. However, if expressed, none of the truncated menin proteins resulting from the 43 known frameshift/nonsense mutations would retain both the NLSs. The precise role(s) of menin in the nucleus remain to be understood.


Assuntos
Núcleo Celular/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas , Sequência de Aminoácidos , Compartimento Celular , Técnica Indireta de Fluorescência para Anticorpo , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes , Microscopia de Fluorescência , Dados de Sequência Molecular , Proteínas de Neoplasias/imunologia , Peptídeos/química , Peptídeos/imunologia
16.
Cancer Res ; 57(21): 4682-6, 1997 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9354421

RESUMO

Gastrinomas and insulinomas are frequent in multiple endocrine neoplasia type 1 (MEN1). The MEN1 tumor suppressor gene was recently identified. To elucidate the etiological role of the MEN1 gene in sporadic enteropancreatic endocrine tumorigenesis, we analyzed tumors (28 gastrinomas and 12 insulinomas) from 40 patients for MEN1 gene mutations and allelic deletions. One copy of the MEN1 gene was found to be deleted in 25 of 27 (93%) sporadic gastrinomas and in 6 of 12 (50%) sporadic insulinomas. MEN1 gene mutations were identified in 9 of 27 (33%) sporadic gastrinomas and 2 of 12 (17%) insulinomas and were not seen in corresponding germ-line DNA sequence. A specific MEN1 mutation was detected in one gastrinoma and in the corresponding germ-line DNA of a patient who had no family history of MEN1. Somatic MEN1 gene mutations and deletions play a critical role in the tumorigenesis of sporadic gastrinomas and may also contribute to the development of a subgroup of insulinomas.


Assuntos
Gastrinoma/genética , Genes Supressores de Tumor/genética , Insulinoma/genética , Neoplasias do Jejuno/genética , Neoplasia Endócrina Múltipla Tipo 1/genética , Mutação/genética , Neoplasias Pancreáticas/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise Mutacional de DNA , DNA de Neoplasias/genética , Feminino , Deleção de Genes , Mutação em Linhagem Germinativa , Humanos , Hibridização in Situ Fluorescente , Masculino , Pessoa de Meia-Idade
17.
Nat Genet ; 16(4): 375-8, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9241276

RESUMO

Primary hyperparathyroidism is a common disorder with an annual incidence of approximately 0.5 in 1,000 (ref. 1). In more than 95% of cases, the disease is caused by sporadic parathyroid adenoma or sporadic hyperplasia. Some cases are caused by inherited syndromes, such as multiple endocrine neoplasia type 1 (MEN1; ref. 2). In most cases, the molecular basis of parathyroid neoplasia is unknown. Parathyroid adenomas are usually monoclonal, suggesting that one important step in tumour development is a mutation in a progenitor cell. Approximately 30% of sporadic parathyroid tumours show loss of heterozygosity (LOH) for polymorphic markers on 11q13, the site of the MEN1 tumour suppressor gene. This raises the question of whether such sporadic parathyroid tumours are caused by sequential inactivation of both alleles of the MEN1 gene. We recently cloned the MEN1 gene and identified MEN1 germline mutations in fourteen of fifteen kindreds with familial MEN1 (ref. 10). We have studied parathyroid tumours not associated with MEN1 to determine whether somatic mutations in the MEN1 gene are present. Among 33 tumours we found somatic MEN1 gene mutation in 7, while the corresponding MEN1 germline sequence was normal in each patient. All tumours with MEN1 gene mutation showed LOH on 11q13, making the tumour cells hemi- or homozygous for the mutant allele. Thus, somatic MEN1 gene mutation for the mutant allele. Thus, somatic MEN1 gene mutation contributes to tumorigenesis in a substantial number of parathyroid tumours not associated with the MEN1 syndrome.


Assuntos
Neoplasia Endócrina Múltipla Tipo 1/genética , Mutação , Proteínas de Neoplasias/genética , Neoplasias das Paratireoides/genética , Proteínas Proto-Oncogênicas , Cromossomos Humanos Par 11 , Impressões Digitais de DNA , Análise Mutacional de DNA , DNA de Neoplasias/análise , Deleção de Genes , Heterozigoto , Humanos
18.
Hum Mol Genet ; 6(7): 1169-75, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9215689

RESUMO

Familial multiple endocrine neoplasia type 1 (FMEN1) is an autosomal dominant trait characterized by tumors of the parathyroids, gastro-intestinal endocrine tissue, anterior pituitary and other tissues. We recently cloned the MEN1 gene and confirmed its identity by finding mutations in FMEN1. We have now extended our mutation analysis to 34 more unrelated FMEN1 probands and to two related states, sporadic MEN1 and familial hyperparathyroidism. There was a high prevalence of heterozygous germline MEN1 mutations in sporadic MEN1 (8/11 cases) and in FMEN1 (47/50 probands). One case of sporadic MEN1 was proven to be a new MEN1 mutation. Eight different mutations were observed more than once in FMEN1. Forty different mutations (32 FMEN1 and eight sporadic MEN1) were distributed across the MEN1 gene. Most predicted loss of function of the encoded menin protein, supporting the prediction that MEN1 is a tumor suppressor gene. No MEN1 germline mutation was found in five probands with familial hyperparathyroidism, suggesting that familial hyperparathyroidism often is caused by mutation in another gene or gene(s).


Assuntos
Mutação em Linhagem Germinativa , Hiperparatireoidismo/genética , Neoplasia Endócrina Múltipla Tipo 1/genética , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogênicas , Adulto , Idoso , Humanos , Pessoa de Meia-Idade , Linhagem , Polimorfismo Genético
19.
Genome Res ; 7(7): 725-35, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9253601

RESUMO

Multiple endocrine neoplasia type 1 (MEN 1) is an inherited cancer syndrome in which affected individuals develop multiple parathyroid, enteropancreatic, and pituitary tumors. The locus for MEN1 is tightly linked to the marker PYGM on chromosome 11q13, and linkage analysis places the MEN1 gene within a 2-Mb interval flanked by the markers D11S1883 and D11S449. Loss of heterozygosity studies in MEN 1 and sporadic tumors suggest that the MEN1 gene encodes a tumor suppressor and have helped to narrow the location of the gene to a 600-kb interval between PYGM and D11S449. Focusing on this smaller MEN1 interval, we have identified and mapped 12 transcripts to this 600-kb region. A precise ordered map of 33 transcripts, including 12 genes known to map to this region, was generated for the 2.8-Mb D11S480-D11S913 interval. Fifteen candidate genes (of which 10 were examined exhaustively) were evaluated by Southern blot and/or dideoxy fingerprinting analysis to identify the gene harboring disease-causing mutations.


Assuntos
Mapeamento Cromossômico , Cromossomos Humanos Par 11 , Genoma Humano , Neoplasia Endócrina Múltipla Tipo 1/genética , Ligação Genética , Humanos , Dados de Sequência Molecular , Transcrição Gênica
20.
Biochim Biophys Acta ; 1353(1): 39-49, 1997 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-9256063

RESUMO

The human gene for the seventh largest subunit of RNA polymerase II complex, hsRPB7 was cloned, sequenced and mapped. This complex is an integral part of the transcription-coupled DNA repair mechanism and has been shown to be involved in several human genetic diseases and implicated in many others. The hsRPB7 gene consists of 8 exons and spans approximately 5.1 kb. Southern blots of genomic and cloned DNA suggest that hsRPB7 is coded for by a single gene. Using human radiation hybrids and YACs, the gene was localized to 11q13.1, within 70 kb of marker D11S1765. The sequence of the 5' flanking region does not contain a TATA element, but does contain several Sp1 binding sites, an AP-1 site and a novel inverted polymorphic GATA tandem repeat. This novel GATA repeat can be used for linkage analysis. The hsRPB7 gene seems to be highly conserved among eukaryotic species, showing general sequence conservation to yeast and Drosophila. Northern blot analysis reveals a high degree of tissue-specific expression. For example, adult retina, brain and kidney exhibit a relatively high level of expression. A moderate level of expression is observed in heart, lung, testis, cornea, retinal pigmented epithelium/choroid and placenta with a lower level of expression in the uterus, small intestine and skeletal muscle. A very low level of expression was observed in stomach and liver. Comparison between four fetal and adult tissues also demonstrate a surprising level of developmental specificity. Expression in fetal retina is considerably lower than fetal brain but similar to adult retina.


Assuntos
Cromossomos Humanos Par 11 , RNA Polimerase II/genética , Sequência de Aminoácidos , Sequência de Bases , Northern Blotting , Southern Blotting , Clonagem Molecular , Cosmídeos , DNA Complementar/biossíntese , DNA Complementar/isolamento & purificação , Expressão Gênica , Humanos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , RNA Polimerase II/química , RNA Mensageiro/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...