Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Mol Psychiatry ; 23(3): 683-690, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28322272

RESUMEN

The study of monozygotic twins discordant for attention deficit hyperactivity disorder can elucidate mechanisms that contribute to the disorder, which affects ~7% of children. First, using in vivo neuroanatomic imaging on 14 pairs of monozygotic twins (mean age 9.7, s.d. 1.9 years), we found that discordance for the disorder is mirrored by differing dimensions of deep brain structures (the striatum and cerebellum), but not the cerebral cortex. Next, using whole-blood DNA from the same twins, we found a significant enrichment of epigenetic differences in genes expressed in these 'discordant' brain structures. Specifically, there is differential methylation of probes lying in the shore and shelf and enhancer regions of striatal and cerebellar genes. Notably, gene sets pertaining to the cerebral cortex (which did not differ in volume between affected and unaffected twins) were not enriched by differentially methylated probes. Genotypic differences between the twin pairs-such as copy number and rare, single-nucleotide variants-did not contribute to phenotypic discordance. Pathway analyses of the genes implicated by the most differentially methylated probes implicated γ-aminobutyric acid (GABA), dopamine and serotonin neurotransmitter systems. The study illustrates how neuroimaging can help guide the search for epigenomic mechanisms in neurodevelopmental disorders.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/diagnóstico por imagen , Trastorno por Déficit de Atención con Hiperactividad/genética , Epigénesis Genética/genética , Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Encéfalo/anatomía & histología , Encéfalo/fisiopatología , Cerebelo/diagnóstico por imagen , Cerebelo/fisiología , Niño , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/fisiopatología , Metilación de ADN , Enfermedades en Gemelos/genética , Epigenómica , Femenino , Genotipo , Humanos , Masculino , Trastornos del Neurodesarrollo/genética , Fenotipo , Gemelos Monocigóticos/genética , Gemelos Monocigóticos/psicología
2.
Oncogene ; 36(22): 3168-3177, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28068329

RESUMEN

Neurofibromatosis type 1 (NF1) is a common tumor-predisposition disorder due to germline mutations in the tumor suppressor gene NF1. A virtually pathognomonic finding of NF1 is the plexiform neurofibroma (PN), a benign, likely congenital tumor that arises from bi-allelic inactivation of NF1. PN can undergo transformation to a malignant peripheral nerve sheath tumor, an aggressive soft-tissue sarcoma. To better understand the non-NF1 genetic contributions to PN pathogenesis, we performed whole-exome sequencing, RNASeq profiling and genome-wide copy-number determination for 23 low-passage Schwann cell cultures established from surgical PN material with matching germline DNA. All resected tumors were derived from routine debulking surgeries. None of the tumors were considered at risk for malignant transformation at the time; for example, there was no pain or rapid growth. Deep (~500X) NF1 exon sequencing was also conducted on tumor DNA. Non-NF1 somatic mutation verification was performed using the Ampliseq/IonTorrent platform. We identified 100% of the germline NF1 mutations and found somatic NF1 inactivation in 74% of the PN. One individual with three PNs had different NF1 somatic mutations in each tumor. The median number of somatic mutations per sample, including NF1, was one (range 0-8). NF1 was the only gene that was recurrently somatically inactivated in multiple tumors. Gene Set Enrichment Analysis of transcriptome-wide tumor RNA sequencing identified five significant (FDR<0.01) and seven trending (0.01⩽FDR<0.02) gene sets related to DNA replication, telomere maintenance and elongation, cell cycle progression, signal transduction and cell proliferation. We found no recurrent non-NF1 locus copy-number variation in PN. This is the first multi-sample whole-exome and whole-transcriptome sequencing study of NF1-associated PN. Taken together with concurrent copy-number data, our comprehensive genetic analysis reveals the primacy of NF1 loss as the driver of PN tumorigenesis.


Asunto(s)
Neurofibroma Plexiforme/patología , Neurofibromatosis 1/patología , Neurofibromina 1/deficiencia , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Replicación del ADN , Dosificación de Gen , Genes Supresores de Tumor , Mutación de Línea Germinal , Humanos , Neurofibroma Plexiforme/genética , Neurofibroma Plexiforme/metabolismo , Neurofibromatosis 1/genética , Neurofibromatosis 1/metabolismo , Neurofibromina 1/genética , Transcriptoma
4.
Mol Syndromol ; 4(1-2): 27-31, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23653574

RESUMEN

Exome sequencing offers an efficient and affordable method to interrogate genetic factors involved in human disease. Performing exome sequencing of monozygotic twins discordant for VACTERL (Vertebral anomalies, Anal atresia, Cardiac malformations, Tracheo-Esophageal fistula, Renal anomalies, and Limb abnormalities) association-type congenital malformations was hypothesized to potentially reveal discordant variants that could demonstrate disease cause(s). After demonstrating monozygosity, we applied high-density microarrays and exome sequencing to 2 twin pairs in which 1 twin had features of VACTERL association while the other was phenotypically normal (demonstrated through comprehensive clinical and radiological evaluation). No obvious discordant genotypic results were found that would explain phenotypic discordance. We conclude that VACTERL association is a complex disease, and while performing microarray analysis and exome sequencing on phenotypically discordant monozygotic twins may hypothetically reveal genetic causes of disorders, challenges remain in applying these methods in this circumstance.

5.
Mol Psychiatry ; 18(5): 568-75, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22710270

RESUMEN

The literature on GWAS (genome-wide association studies) data suggests that very large sample sizes (for example, 50,000 cases and 50,000 controls) may be required to detect significant associations of genomic regions for complex disorders such as Alzheimer's disease (AD). Because of the challenges of obtaining such large cohorts, we describe here a novel sequential strategy that combines pooling of DNA and bootstrapping (pbGWAS) in order to significantly increase the statistical power and exponentially reduce expenses. We applied this method to a very homogeneous sample of patients belonging to a unique and clinically well-characterized multigenerational pedigree with one of the most severe forms of early onset AD, carrying the PSEN1 p.Glu280Ala mutation (often referred to as E280A mutation), which originated as a consequence of a founder effect. In this cohort, we identified novel loci genome-wide significantly associated as modifiers of the age of onset of AD (CD44, rs187116, P=1.29 × 10⁻¹²; NPHP1, rs10173717, P=1.74 × 10⁻¹²; CADPS2, rs3757536, P=1.54 × 10⁻¹°; GREM2, rs12129547, P=1.69 × 10⁻¹³, among others) as well as other loci known to be associated with AD. Regions identified by pbGWAS were confirmed by subsequent individual genotyping. The pbGWAS methodology and the genes it targeted could provide important insights in determining the genetic causes of AD and other complex conditions.


Asunto(s)
Alanina/genética , Enfermedad de Alzheimer/genética , Predisposición Genética a la Enfermedad , Ácido Glutámico/genética , Presenilina-1/genética , Edad de Inicio , Enfermedad de Alzheimer/epidemiología , Estudios de Cohortes , Bases de Datos Factuales/estadística & datos numéricos , Femenino , Efecto Fundador , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Mutación/genética , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo
6.
Horm Metab Res ; 37(6): 369-74, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16001329

RESUMEN

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.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Núcleo Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasia Endocrina Múltiple Tipo 1/genética , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/metabolismo , Transcripción Genética
7.
J Intern Med ; 253(6): 606-15, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12755956

RESUMEN

Multiple endocrine neoplasia type 1 is an autosomal dominant cancer syndrome affecting primarily parathyroid, enteropancreatic endocrine and pituitary tissues. The inactivating germline and somatic mutations spread throughout the gene and the accompanying loss of the second allele in tumours show that the MEN1 gene is a tumour suppressor. The MEN1-encoded protein, menin, is a novel nuclear protein. Menin binds and alters JunD-, NF-kappaB-, Smad3-mediated transcriptional activation. The mouse Men1 knockout model mimicks the human MEN1 condition contributing to the understanding of tumorigenesis in MEN1.


Asunto(s)
Genes Supresores de Tumor , Neoplasia Endocrina Múltiple Tipo 1/genética , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas , Animales , Secuencia de Bases , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Drosophila melanogaster , Humanos , Linfocitos/metabolismo , Ratones , Ratones Noqueados , Modelos Animales , Datos de Secuencia Molecular , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , FN-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Mapeo de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Alineación de Secuencia , Proteína smad3 , Transactivadores/metabolismo
8.
Genesis ; 30(4): 259-63, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11536432

RESUMEN

In an effort to create a conventional knockout mouse model for multiple endocrine neoplasia type 1 (MEN1), we targeted disruption of the mouse Men1 gene through homologous recombination in ES cells. Men1 exons 2-4 were replaced by a PGK-neomycin cassette inserted in the opposite direction of Men1 transcription (Men1(MSK/+)). Unexpectedly, the Men1 conventional knockout was lethal in heterozygous, chimeric animals. Analysis of embryos revealed late gestational lethality with some embryos showing omphalocele. This was a very surprising phenotype, given that humans and mice that are heterozygotes for loss of function mutations in MEN1 are phenotypically normal except for a risk of endocrine tumors. Northern analysis of Men1(MSK/+) embryonic stem cell RNA revealed the presence of an abundant, novel transcript of 2.1 kb, in addition to the expected wild-type transcripts of 2.7 kb and 3.1 kb. RT-PCR analysis identified this aberrant transcript as arising from the antisense strand of the PGK promoter. We hypothesize that this transcript is producing either a toxic effect at the RNA level, or a dominant negative effect through the production of an amino-terminal truncated protein product. This example serves as a cautionary reminder that mouse knockouts using PGK-neo may sometimes display phenotypes that reflect more than just the loss of function of the targeted gene.


Asunto(s)
Pérdida del Embrión/genética , Genes Letales/genética , Heterocigoto , Mutagénesis Insercional/genética , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas , Transcripción Genética/genética , Animales , Western Blotting , Quimera/genética , Embrión de Mamíferos/metabolismo , Exones/genética , Eliminación de Gen , Marcación de Gen/métodos , Genes Dominantes/genética , Genes Reporteros/genética , Hernia Umbilical/genética , Ratones , Ratones Noqueados , Neomicina/biosíntesis , Fenotipo , Pruebas de Precipitina , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
Oncogene ; 20(36): 4917-25, 2001 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-11526476

RESUMEN

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.


Asunto(s)
Genes Supresores de Tumor , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas , Animales , Células COS , Línea Celular , Glutatión Transferasa/química , Células HeLa , Humanos , FN-kappa B/química , Proteínas de Neoplasias/química , Proteínas de Neoplasias/inmunología , Pruebas de Precipitina , Estructura Terciaria de Proteína , Acetato de Tetradecanoilforbol/farmacología , Activación Transcripcional
11.
Gene ; 263(1-2): 31-8, 2001 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-11223240

RESUMEN

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.


Asunto(s)
Drosophila melanogaster/genética , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas , Secuencia de Aminoácidos , Animales , Northern Blotting , ADN Complementario/química , ADN Complementario/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/crecimiento & desarrollo , Embrión no Mamífero/metabolismo , Desarrollo Embrionario , Exones , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes de Insecto/genética , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Humanos , Hibridación in Situ , Intrones , Masculino , Ratones , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Saccharomyces cerevisiae/genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia , Transcripción Genética , Técnicas del Sistema de Dos Híbridos , Pez Cebra
12.
Proc Natl Acad Sci U S A ; 98(3): 1118-23, 2001 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-11158604

RESUMEN

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant cancer syndrome, characterized primarily by multiple tumors in the parathyroid glands, endocrine pancreas, and anterior pituitary. Other tumors, including gastrinoma, carcinoid, adrenal cortical tumors, angiofibroma, collagenoma, and lipoma, also occur in some patients. Individuals with MEN1 almost always have loss-of-function mutations in the MEN1 gene on chromosome 11, and endocrine tumors arising in these patients usually show somatic loss of the remaining wild-type allele. To examine the role of MEN1 in tumor formation, a mouse model was generated through homologous recombination of the mouse homolog Men1. Homozygous mice die in utero at embryonic days 11.5-12.5, whereas heterozygous mice develop features remarkably similar to those of the human disorder. As early as 9 months, pancreatic islets show a range of lesions from hyperplasia to insulin-producing islet cell tumors, and parathyroid adenomas are also frequently observed. Larger, more numerous tumors involving pancreatic islets, parathyroids, thyroid, adrenal cortex, and pituitary are seen by 16 months. All of the tumors tested to date show loss of the wild-type Men1 allele, further supporting its role as a tumor suppressor gene.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/genética , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas , Adenoma/genética , Adenoma/patología , Animales , Mapeo Cromosómico , Cromosomas Artificiales Bacterianos , Cromosomas Humanos Par 11 , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Exones , Femenino , Muerte Fetal , Genes Letales , Genes Supresores de Tumor , Homocigoto , Humanos , Hiperparatiroidismo/genética , Hiperparatiroidismo/patología , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Neoplasia Endocrina Múltiple Tipo 1/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias de las Paratiroides/genética , Neoplasias de las Paratiroides/patología , Fosfoglicerato Quinasa/genética , Embarazo , Recombinación Genética
13.
Nature ; 407(6806): 913-6, 2000 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-11057671

RESUMEN

The vertebrate organizer can induce a complete body axis when transplanted to the ventral side of a host embryo by virtue of its distinct head and trunk inducing properties. Wingless/Wnt antagonists secreted by the organizer have been identified as head inducers. Their ectopic expression can promote head formation, whereas ectopic activation of Wnt signalling during early gastrulation blocks head formation. These observations suggest that the ability of head inducers to inhibit Wnt signalling during formation of anterior structures is what distinguishes them from trunk inducers that permit the operation of posteriorizing Wnt signals. Here we describe the zebrafish headless (hdl) mutant and show that its severe head defects are due to a mutation in T-cell factor-3 (Tcf3), a member of the Tcf/Lef family. Loss of Tcf3 function in the hdl mutant reveals that hdl represses Wnt target genes. We provide genetic evidence that a component of the Wnt signalling pathway is essential in vertebrate head formation and patterning.


Asunto(s)
Proteínas HMGB , Cabeza/embriología , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Proteínas de Pez Cebra , Animales , Mapeo Cromosómico , Clonación Molecular , Perfilación de la Expresión Génica , Cabeza/anomalías , Mutación , Organizadores Embrionarios , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Transducción de Señal , Factores de Transcripción TCF , Proteína 1 Similar al Factor de Transcripción 7 , Factores de Transcripción/genética , Proteínas Wnt , Pez Cebra/embriología
14.
Oncogene ; 19(41): 4706-12, 2000 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-11032020

RESUMEN

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.


Asunto(s)
Sustitución de Aminoácidos , Mutación Missense , Proteínas de Neoplasias/metabolismo , Isoformas de Proteínas/genética , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas , Secuencia de Aminoácidos , Animales , Línea Celular , Cromatografía en Gel , Humanos , Riñón , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Activación Transcripcional , Transfección , Técnicas del Sistema de Dos Híbridos
15.
Genes Chromosomes Cancer ; 29(2): 157-65, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10959095

RESUMEN

Previous cytogenetic and molecular genetic studies have shown that the HeLa (cervical carcinoma) cell line D98/AH-2 contains two apparently normal copies of chromosome 11 and additional 11q13-25 material translocated onto a chromosome 3 marker. To determine the 11q13 breakpoint, we performed fluorescence in situ hybridization (FISH) using 18 different 11q13 specific BAC (bacterial artificial chromosome) and cosmid probes spanning a 5.6 Mb interval. Markers localized to the multiple endocrine neoplasia type 1 (MEN1) gene (menin) were also included in the analysis. The FISH study identified an interstitial deletion between markers D11S449 and GSTP1, an interval of 2.3 Mb, in the marker chromosome. This deletion did not include the MEN1 gene. Because point mutations and methylations can inactivate the MEN1 gene, single stranded conformational polymorphism (SSCP) and Northern and Western blot analyses were performed with MEN1 specific probes and antibody. SSCP did not reveal mutations of the MEN1 gene in HeLa or in seven other cervical cancer cell lines. Northern and Western blot studies revealed normal levels of expression of this gene in the cervical cancer cell lines as well as in HeLa cell derived tumorigenic hybrids. Because deletions of tumor suppressor genes often occur in cancer progression, we hypothesize that the inactivation of a tumor suppressor gene other than MEN1, localized to the 2.3 Mb interval on 11q13, might play a role in the abnormal growth behavior of HeLa cells in vitro or in vivo.


Asunto(s)
Cromosomas Humanos Par 11/genética , Proteínas Proto-Oncogénicas , Eliminación de Secuencia , Transformación Celular Neoplásica/genética , Femenino , Células HeLa , Humanos , Células Híbridas , Neoplasia Endocrina Múltiple Tipo 1/etiología , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Mutación , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/genética
16.
Hum Mutat ; 16(1): 92, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10874319

RESUMEN

Alagille syndrome (AGS) is an autosomal dominant, developmental disorder affecting multiple organ systems including liver, heart, vertebrae, eye and face. Recurrent deletions of the 20p12 region led to the localization, and ultimately to the identification of mutations in the Jagged1 gene (JAG1) in AGS patients. A translocation t(3;20)(q13.3;p12.2) in an AGS patient was characterized using fluorescent in situ hybridization (FISH). The involvement of 3q and 20p in this translocation was demonstrated using probes for 3q and 20p. Three overlapping YAC clones, 940D11, 953A2, and 675G11 extending to nearly 4 Mb including the JAG1 were used as probes for FISH analysis to define the translocation breakpoint. The translocated chromosome was found to have a deletion of more than 3 Mb including the entire JAG1 gene. The observation of an accompanying large deletion, revealed by molecular characterization of the t(3;20) translocation, is similar to the only other translocation reported in an AGS patient; a t(2;20) translocation was also found to have a large deletion of the JAG1 region at 20p12.


Asunto(s)
Síndrome de Alagille/genética , Deleción Cromosómica , Cromosomas Humanos Par 20/genética , Cromosomas Humanos Par 3/genética , Proteínas/genética , Translocación Genética/genética , Síndrome de Alagille/diagnóstico , Proteínas de Unión al Calcio , Humanos , Lactante , Recién Nacido , Péptidos y Proteínas de Señalización Intercelular , Proteína Jagged-1 , Masculino , Proteínas de la Membrana , Proteínas Serrate-Jagged
17.
Mamm Genome ; 11(6): 448-54, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10818209

RESUMEN

Mutations in the MEN1 gene lead to an autosomal dominant disorder, multiple endocrine neoplasia type 1 (MEN1), which is characterized by tumors of the parathyroid, entero-pancreatic neuroendocrine, and pituitary tissues. The protein encoded by MEN1, 610-amino acid menin, resides primarily in the nucleus and binds to the transcription factor JunD, resulting in the repression of JunD-induced transcription. We report here a detailed characterization of the zebrafish men1 gene and its full-length (2551 nt) transcript, encoding a 617-amino acid protein with 67% identity and 80% similarity to human menin. Of the 81 missense mutations and in-frame deletions reported in MEN1 patients, 72 occur in residues that are identical in zebrafish, suggesting the importance of the conserved regions. The zebrafish men1 gene maps 61 cM from the top of linkage group 7 (LG7), a region that appears to show conserved synteny to the MEN1 loci at human 11q13. A 2.7-kb men1 message is detected at all stages of zebrafish development analyzed, from one-cell embryos to adult fish. Whole-mount in situ hybridization showed ubiquitous distribution of men1 message in zebrafish embryos at cleavage, blastula, gastrula, and early segmentation stages, with relatively abundant expression in blood cell progenitors (24 h post fertilization) and mesenchymal tissues (48 h post fertilization) at later stages. Zebrafish menin binds both human and mouse JunD, and represses JunD-induced transcription, indicating that the JunD-binding ability of menin is evolutionarily conserved.


Asunto(s)
ADN Complementario/genética , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting , Mapeo Cromosómico , ADN/química , ADN/genética , ADN Complementario/química , ADN Complementario/aislamiento & purificación , Embrión no Mamífero/metabolismo , Desarrollo Embrionario , Exones , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación in Situ , Intrones , Ratones , Datos de Secuencia Molecular , Proteínas de Neoplasias/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN/genética , ARN/metabolismo , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Activación Transcripcional , Pez Cebra/embriología
18.
Genes Chromosomes Cancer ; 28(1): 58-65, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10738303

RESUMEN

Neuroendocrine tumors of the lung consist of a spectrum of neoplasms, including typical carcinoids, atypical carcinoids, large-cell neuroendocrine carcinomas (LCNEC), and small-cell lung carcinomas (SCLC). We previously reported frequent inactivation of the gene responsible for multiple endocrine neoplasia type 1 (MEN1) in both typical and atypical carcinoid tumors. In the present study, we extend the analysis of human NE lung tumors to include 9 primary SCLCs, 36 SCLC cell lines, and 13 primary LCNECs for MEN1 gene inactivation. In SCLC, loss of heterozygosity (LOH) at the MEN1 gene on chromosome band 11q13 was detected in one primary tumor and two cell lines. The coding sequence and splice junctions of the MEN1 gene were screened for mutations in all 44 tumors and cell lines, and no mutations were detected. Northern blot analysis of 13 SCLC cell lines showed the MEN1 transcript to be present and of normal size. In LCNECs, a somatic frameshift in the MEN1 gene (1226delC) was found in one of 13 tumors, representing the first mutation observed outside the spectrum of neoplasms associated with MEN1. Interestingly, neither a deletion nor a mutation was detected in the other allele, and wild-type mRNA sequence was expressed in the tumor, suggesting that the MEN1 gene was not inactivated by a conventional two-hit mechanism. The data support the hypothesis that SCLC and lung carcinoids develop via distinct molecular pathways; however, further investigation is necessary to determine the significance of the MEN1 gene mutation observed in a single case of LCNEC. Published 2000 Wiley-Liss, Inc.


Asunto(s)
Carcinoma Neuroendocrino/genética , ADN de Neoplasias/análisis , Neoplasias Pulmonares/genética , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas , Carcinoma de Células Grandes/genética , Carcinoma de Células Pequeñas/genética , Cromosomas Humanos Par 11/genética , Análisis Mutacional de ADN/métodos , Humanos , Pérdida de Heterocigocidad/genética , Neoplasia Endocrina Múltiple Tipo 1/genética , Células Tumorales Cultivadas
19.
Genomics ; 62(1): 21-33, 1999 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-10585764

RESUMEN

The region of chromosome 2 encompassed by the polymorphic markers D2S378 (centromeric) and D2S391 (telomeric) spans an approximately 10-cM distance in cytogenetic bands 2p15-p21. This area is frequently involved in cytogenetic alterations in human cancers. It also harbors the genes for several genetic disorders, including Type I hereditary nonpolyposis colorectal cancer (HNPCC), familial male precocious puberty (FMPP), Carney complex (CNC), Doyne's honeycomb retinal dystrophy (DHRD), and one form of familial dyslexia (DYX-3). Only a handful of known genes have been mapped to 2p16. These include MSH2, which is responsible for HNPCC, FSHR, the gene responsible for FMPP, EFEMP-1, the gene mutated in DHRD, GTBP, a DNA repair gene, and SPTBN1, nonerythryocytic beta-spectrin. The genes for CNC and DYX-3 remain unknown, due to lack of a contig of this region and its underrepresentation in the existing maps. This report presents a yeast- and bacterial-artificial chromosome (YAC and BAC, respectively) resource for the construction of a sequence-ready map of 2p15-p21 between the markers D2S378 and D2S391 at the centromeric and telomeric ends, respectively. The recently published Genemap'98 lists 146 expressed sequence tags (ESTs) in this region; we have used our YAC-BAC map to place each of these ESTs within a framework of 40 known and 3 newly cloned polymorphic markers and 37 new sequence-tagged sites. This map provides an integration of genetic, radiation hybrid, and physical mapping information for the region corresponding to cytogenetic bands 2p15-p21 and is expected to facilitate the identification of disease genes from the area.


Asunto(s)
Mapeo Cromosómico , Cromosomas Humanos Par 2/genética , Enfermedades Genéticas Congénitas/genética , Mapeo Cromosómico/métodos , Paseo de Cromosoma , Cromosomas Artificiales de Levadura , Cromosomas Bacterianos , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , ADN Recombinante/genética , Dislexia/genética , Etiquetas de Secuencia Expresada , Marcadores Genéticos , Enfermedades de los Genitales Masculinos/genética , Humanos , Hibridación Fluorescente in Situ , Masculino , Síndromes Neoplásicos Hereditarios/genética , Enfermedades de la Retina/genética
20.
Recent Prog Horm Res ; 54: 397-438; discussion 438-9, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10548885

RESUMEN

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.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/fisiopatología , Secuencia de Aminoácidos , Hormonas/metabolismo , Humanos , Datos de Secuencia Molecular , Neoplasia Endocrina Múltiple Tipo 1/epidemiología , Neoplasia Endocrina Múltiple Tipo 1/terapia , Linaje , Prevalencia , Tasa de Secreción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...