Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Biol Lett ; 20(7): 20240177, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38982849

RESUMO

While various marine predators form associations, the most commonly studied are those between subsurface predators and seabirds, with gulls, shearwaters or terns frequently co-occurring with dolphins, billfish or tuna. However, the mechanisms underlying these associations remain poorly understood. Three hypotheses have been proposed to explain the prevalence of these associations: (1) subsurface predators herd prey to the surface and make prey accessible to birds, (2) subsurface predators damage prey close to the surface and thereby provide food scraps to birds, and (3) attacks of underwater predators lower the cohesion of prey groups and thereby their collective defences making the prey easier to be captured by birds. Using drone footage, we investigated the interaction between Indo-Pacific sailfish (Istiophorus platypterus) and terns (Onychoprion sp.) preying on schooling fish off the eastern coast of the Malaysian peninsula. Through spatio-temporal analysis of the hunting behaviour of the two predatory species and direct measures of prey cohesion we showed that terns attacked when school cohesion was low, and that this decrease in cohesion was frequently caused by sailfish attacks. Therefore, we propose that sailfish created a by-product benefit for the bird species, lending support to the hypothesis that lowering cohesion can facilitate associations between subsurface predators and seabirds.


Assuntos
Comportamento Predatório , Animais , Charadriiformes/fisiologia , Peixes/fisiologia , Malásia , Cadeia Alimentar , Aves/fisiologia , Comportamento Alimentar
2.
R Soc Open Sci ; 5(12): 181132, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30662732

RESUMO

Despite the frequency with which mixed-species groups are observed in nature, studies of collective behaviour typically focus on single-species groups. Here, we quantify and compare the patterns of interactions between three fish species, threespine sticklebacks (Gasterosteus aculeatus), ninespine sticklebacks (Pungitius pungitius) and roach (Rutilus rutilus) in both single- and mixed-species shoals in the laboratory. Pilot data confirmed that the three species form both single- and mixed-species shoals in the wild. In our laboratory study, we found that single-species groups were more polarized than mixed-species groups, while single-species groups of threespine sticklebacks and roach were more cohesive than mixed shoals of these species. Furthermore, while there was no difference between the inter-individual distances between threespine and ninespine sticklebacks within mixed-species groups, there was some evidence of segregation by species in mixed groups of threespine sticklebacks and roach. There were differences between treatments in mean pairwise transfer entropy, and in particular we identify species-differences in information use within the mixed-species groups, and, similarly, differences in responses to conspecifics and heterospecifics in mixed-species groups. We speculate that differences in the patterns of interactions between species in mixed-species groups may determine patterns of fission and fusion in such groups.

3.
Horm Metab Res ; 37(6): 369-74, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16001329

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant disease predisposed by heterozygous germline mutations in the MEN1 tumor suppressor gene. Biallelic loss of MEN1 resulting from small mutation and/or loss of heterozygosity occurs in a large tissue spectrum of MEN1 tumors or non-hereditary tumors. Mouse models of MEN1 underexpression or overexpression have also supported the tumor-suppressor effect of the MEN1 gene. Menin, the 610-amino-acid protein encoded by MEN1, is expressed ubiquitously and found predominantly in the nucleus. Sequence analyses do not reveal motifs of known function other than two nuclear localization sequences. Menin has been found to partner in vitro with a variety of proteins that comprise transcription factors, DNA processing factors, DNA repair proteins, and cytoskeletal proteins. The diverse functions of menin interactors suggest roles for menin in multiple biological pathways. Inactivation of menin switches its JunD partner from a downstream action of growth suppression to growth promotion. This is a plausible mechanism for menin tumorigenesis.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasia Endócrina Múltipla Tipo 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Núcleo Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , DNA/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasia Endócrina Múltipla Tipo 1/genética , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
4.
J Invest Dermatol ; 117(5): 1027-35, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11710909

RESUMO

Previous studies have documented that the ability to heal wounds declines with age. Although many factors contribute to this age-associated deficit, one variable that has not been carefully examined is leukocyte recruitment and function in wounds. This investigation compares the inflammatory response in excisional wounds of young (age 8 wk) and aged (age 22 mo) mice. In the early inflammatory response, neutrophil content of wounds was similar for both aged and young mice. In contrast, macrophage levels were 56% higher in aged versus young mice (81 +/- 20 vs 52 +/- 13 cells per mm2). In the later inflammatory response, wounds of aged mice exhibited a delay in T cell infiltration, with maximum T cell levels at day 10 in aged mice versus day 7 in young mice. Despite this delay, the eventual peak concentration of T cells was 23% higher in the wounds of aged mice (152 +/- 11 cells per mm2 vs 124 +/- 21cells per mm2). The observed alterations in inflammatory cell content suggested that chemokine production might be altered with age. An elevation of monocyte chemoattractant protein (MCP-1) levels was observed in wounds of aged mice. RNase protection studies, however, revealed that the production of most chemokines, including MIP-2, MIP-1alpha, MIP-1beta, and eotaxin, tended to decline with age. Because optimal wound healing requires both appropriate macrophage infiltration and phagocytic activity, phagocytosis was examined. Compared to young mice, wound macrophages from aged mice exhibited a 37%-43% reduction in phagocytic capacity. Taken together, the data demonstrate age-related shifts in both macrophage and T cell infiltration into wounds, alterations in chemokine content, and a concurrent decline in wound macrophage phagocytic function. These alterations may contribute to the delayed repair response of aging.


Assuntos
Envelhecimento/fisiologia , Dermatite/etiologia , Pele/lesões , Ferimentos Penetrantes/complicações , Animais , Quimiocinas/metabolismo , Feminino , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Infiltração de Neutrófilos , Receptores de IgG/metabolismo , Pele/metabolismo , Pele/patologia , Pele/fisiopatologia , Ferimentos Penetrantes/metabolismo , Ferimentos Penetrantes/patologia , Ferimentos Penetrantes/fisiopatologia
5.
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
7.
Trends Endocrinol Metab ; 12(4): 173-8, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11295574

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) provides a prime example of how a rare disease can advance our understanding of basic cell biology, neoplasia and common endocrine tumors. MEN1 is expressed mainly as parathyroid, enteropancreatic neuroendocrine, anterior pituitary and foregut carcinoid tumors. It is an autosomal dominant disease caused by mutation of the MEN1 gene. Since its identification, the MEN1 gene has been implicated in many common endocrine and non-endocrine tumors. This is a brief overview of recent scientific advances relating to MEN1, including newly recognized clinical features that are now better characterized by genetic analysis, insights into the function of the MEN1-encoded protein menin, and refined recommendations for mutation testing and tumor screening, which highlight our increasing understanding of this complex syndrome.


Assuntos
Neoplasia Endócrina Múltipla Tipo 1 , Proteínas Proto-Oncogênicas , Neoplasias do Córtex Suprarrenal/genética , Angiofibroma/genética , Humanos , Leiomioma/genética , Neoplasia Endócrina Múltipla Tipo 1/genética , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Feocromocitoma/genética , Neoplasias Cutâneas/genética , Neoplasias da Glândula Tireoide/genética
8.
Cytotechnology ; 35(2): 127-35, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19003289

RESUMO

A process was developed for producing human menin from transformed Drosophila Schneider 2 cells. Protein expression was achieved after inducing the metallothionein promoter by adding copper sulfate to cells growing in suspension in a stirred-tank reactor. Experiments in shake flasks showed that the production of menin was improved when the induction was conducted late in the exponential phase of cell growth at a concentration of 1-2 x 10(7) cells ml(-1), with a copper concentration of 0.2 mM for no more than 24 h. This observation was confirmed by experiments in bench-scale fermentors. Subsequently, a pilot-scale fermentation yielded 1 mg l(-1) culture of purified menin.

9.
Oncogene ; 19(41): 4706-12, 2000 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-11032020

RESUMO

Menin, the product of the MEN1 tumor suppressor gene, binds to the AP1 transcription factor JunD and represses JunD transcriptional activity. The effects of human or mouse JunD missense mutations upon menin interaction were studied by random and alanine scanning mutagenesis of the menin binding region of JunD (amino acids 1-70). JunD mutant proteins were tested for menin binding in a reverse yeast two-hybrid assay, and for transcriptional regulation by menin in AP1-reporter assays. Random mutagenesis identified two different mutations that disrupted menin interaction at mouse JunD amino acid 42 (G42E and G42R). Mutation G42A generated by alanine scanning did not affect menin binding, likely reflecting the conserved nature of this amino acid substitution. Furthermore, by size exclusion chromatography menin co-migrated with wild type JunD but not with the JunD mutant tested (G42E). Alanine scanning mutagenesis of residues 30-55 revealed two different amino acids, P41 and P44, of mouse JunD that were critical for interaction with menin. Mouse JunD missense mutants P41A, G42R, G42E and P44A failed to bind menin and also escaped menin's control over their transcriptional activity. At lower amounts of transfected menin, the transcriptional effect of menin on the mutants P41A, G42R and G42E was changed from repression to activation, similar to that with c-jun. In conclusion, a small N-terminal region of JunD mediates a key difference between JunD and c-jun, and a component of this difference is dependent on JunD binding to menin.


Assuntos
Substituição de Aminoácidos , Mutação de Sentido Incorreto , Proteínas de Neoplasias/metabolismo , Isoformas de Proteínas/genética , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas , Sequência de Aminoácidos , Animais , Linhagem Celular , Cromatografia em Gel , Humanos , Rim , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ratos , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Ativação Transcricional , Transfecção , Técnicas do Sistema de Duplo-Híbrido
10.
J Clin Endocrinol Metab ; 85(1): 116-23, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10634374

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) gene mutations are reported in some gastrinomas occurring in patients without MEN1 as well as in some other pancreatic endocrine tumors (PETs). In some inherited syndromes phenotype-genotype correlations exist for disease severity, location, or other manifestations. The purpose of the present study was to correlate mutations of the MEN1 gene in a large cohort of patients with sporadic gastrinomas to disease activity, tumor location, extent, and growth pattern. DNA was extracted from frozen gastrinomas from 51 patients and screened by dideoxyfinger-printing (ddF) for abnormalities in the 9 coding exons and adjacent splice junctions of the MEN1 gene. Tumor DNA exhibiting abnormal ddF patterns was sequenced for mutations. The findings were correlated with clinical manifestations of the disease, primary tumor site, disease extent, and tumor growth postoperatively. Tumor growth was determined by serial imaging studies. Sixteen different MEN1 gene mutations in the 51 sporadic gastrinomas (31%) were identified (11 truncating, 4 missense, and 1 in-frame deletion). Nine of the 16 mutations were located in exon 2 compared to 7 of 16 in the remaining 8 coding exons (P = 0.005 on a per nucleotide basis). Primary pancreatic or lymph node gastrinomas with a mutation had only exon 2 mutations, whereas duodenal tumors uncommonly harbored exon 2 mutations (P = 0.011). Similarly, small primary tumors (<1 cm) more frequently contained a nonexon 2 mutation (P = 0.02). There was no difference between patients with or without a mutation with respect to clinical characteristics, primary tumor site, disease extent, or proportion of patients disease free after surgery. Postoperative tumor growth tended to be more aggressive in patients with a mutation (P = 0.09). No correlation in the rate of disease-free status or postoperative tumor growth in patients with active disease to the location of the mutation was seen. These results demonstrate that the MEN1 gene is mutated in 31% of sporadic gastrinomas, and mutations are clustered between amino acids 66-166, which differs from patients with familial MEN1, in whom mutations occur throughout the gene. The presence of an MEN1 gene mutation does not correlate with clinical characteristics of patients with gastrinomas, gastrinoma extent, or growth pattern; however, the location of the mutation differed with gastrinoma location. These data suggest that mutations in the MEN1 gene are important in a proportion of sporadic gastrinomas, but the presence or absence of these mutations will not identify the clinically important subgroups with different growth patterns.


Assuntos
Gastrinoma/genética , Neoplasia Endócrina Múltipla Tipo 1/genética , Mutação/genética , Neoplasias Pancreáticas/genética , Sequência de Bases , DNA de Neoplasias/análise , DNA de Neoplasias/genética , Éxons/genética , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Fenótipo , Reação em Cadeia da Polimerase , Síndrome de Zollinger-Ellison/genética
11.
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
12.
Oncogene ; 18(43): 5936-42, 1999 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-10557080

RESUMO

Although there is indirect genetic evidence that MEN1, the gene for multiple endocrine neoplasia type 1, is a tumor suppressor gene, little is known about the MEN1-encoded protein, menin. Menin was stably overexpressed in a well-characterized murine tumor cell line, (valine-12)-RAS-transformed NIH3T3 cells. Menin overexpression reverted the morphology of the RAS-transformed NIH3T3 cells towards the more flattened and more spread, fibroblastic shape of wild type NIH3T3 cells. The proliferation rate of the RAS-transformed cells in 0.5% calf serum was also slower with menin overexpression. Menin overexpression reduced the RAS-induced clonogenicity in soft agar. Menin also reduced tumor growth after injection of cells in nude mice. In conclusion, stable overexpression of MEN1 suppressed partially the RAS-mediated tumor phenotype in vitro and in vivo. Overexpressed menin protein had biological effects, directly supporting MEN1 gene function as a tumor suppressor.


Assuntos
Transformação Celular Neoplásica , Genes Supressores de Tumor , Neoplasia Endócrina Múltipla Tipo 1/genética , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas , Proteínas ras/genética , Células 3T3 , Ágar , Sequência de Aminoácidos , Animais , Divisão Celular , Núcleo Celular/metabolismo , Células Cultivadas , Meios de Cultura , Fibroblastos/citologia , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Transfecção
13.
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
14.
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
15.
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
16.
J Clin Endocrinol Metab ; 84(1): 216-9, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9920087

RESUMO

Adrenocortical tumors occur as sporadic tumors, as part of the multiple endocrine neoplasia type 1 (MEN1) syndrome or as part of other hereditary disorders. We recently cloned the MEN1 gene, a tumor-suppressor gene located on chromosome 11q13. Subsequently, we showed that sequential somatic inactivation of both alleles of the MEN1 gene contributes to the development of some sporadic endocrine neoplasms (parathyroid, enteropancreatic neuroendocrine, bronchial carcinoid, and pituitary tumors). We now studied whether somatic inactivation of the MEN1 gene contributes to the pathogenesis of sporadic adrenocortical neoplasms. Seven adrenocortical carcinomas, 2 adrenocortical carcinoma cell lines, and 11 aldosterone-secreting, 8 cortisol-secreting, and 5 nonsecreting benign adrenocortical tumors were studied. Seven tumors (5 of 5 carcinomas, 2 of 21 nonsecreting benign adenomas; P < 0.001) exhibited loss of heterozygosity on 11q13. All 33 tumors and cell lines were screened for mutation throughout the MEN1 open-reading frame and adjacent splice junctions. None exhibited a mutation within the MEN1-coding region. We conclude that somatic MEN1 mutation within the MEN1-coding region does not occur commonly in sporadic adrenocortical tumors, although the majority of adrenocortical carcinomas exhibit 11q13 loss of heterozygosity.


Assuntos
Neoplasias do Córtex Suprarrenal/genética , Neoplasia Endócrina Múltipla Tipo 1/genética , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogênicas , Cromossomos Humanos Par 11 , Humanos , Perda de Heterozigosidade , Mutação
17.
Cancer Genet Cytogenet ; 106(1): 30-6, 1998 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9772906

RESUMO

Primary hyperparathyroidism is characterized by hypercalcemia and elevated parathyroid hormone levels. It can be caused by overactivity of one (adenoma or carcinoma) or more (hyperplasia or multiple adenoma) parathyroid glands. Parathyroid adenoma and hyperplasia are usually mono- or oligoclonal neoplasms. To establish whether parathyroid cancer has a genetic composition distinct from parathyroid adenoma, we analyzed 10 adenoma and 10 carcinoma cases by comparative genomic hybridization (CGH). Results show clear differences between the constitution of adenoma and carcinoma genomic DNA. The most frequent genomic alterations in adenoma included deletions on chromosomes 11, 17 (5 of 10 cases), and 22 (7 of 10 cases). In parathyroid carcinoma, frequent chromosomal deletions were on chromosome arm 1p (4 of 10 cases) and chromosome 17 (3 of 10 cases), and gains were on chromosome 5 (3 of 10 cases). Our data indicate that different genetic changes could contribute to the development of parathyroid adenoma and carcinoma; genomic losses predominate in adenoma, and gains along with some losses are found in carcinoma. Furthermore, the CGH results implicate several chromosomal regions that may harbor genes that could be potentially involved in the development of parathyroid adenoma and carcinoma.


Assuntos
Adenoma/genética , Carcinoma/genética , Aberrações Cromossômicas , Neoplasias das Paratireoides/genética , Deleção Cromossômica , Humanos , Hibridização in Situ Fluorescente
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA