Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Am J Intellect Dev Disabil ; 115(2): 83-112, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20441388

RESUMEN

Down syndrome, the most common genetic cause of intellectual disability, is associated with brain disorders due to chromosome 21 gene overdosage. Molecular and cellular mechanisms involved in the neuromorphological alterations and cognitive impairments are reported herein in a global model. Recent advances in Down syndrome research have lead to the identification of altered molecular pathways involved in intellectual disability, such as Calcineurin/NFATs pathways, that are of crucial importance in understanding the molecular basis of intellectual disability pathogenesis in this syndrome. Potential treatments in mouse models of Down syndrome, including antagonists of NMDA or GABA(A) receptors, and microRNAs provide new avenues to develop treatments of intellectual disability. Nevertheless, understanding the links between molecular pathways and treatment strategies in human beings requires further research.


Asunto(s)
Síndrome de Down/genética , Síndrome de Down/fisiopatología , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Animales , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Síndrome de Down/patología , Genómica , Humanos , Discapacidad Intelectual/patología , Ratones , Proteómica
2.
Int J Dev Neurosci ; 27(4): 393-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19460634

RESUMEN

The brain alterations and mental retardation in Down syndrome are associated with overdosage of chromosome 21 genes. To shed light on the understanding of the molecular effect of this genetic overdosage, gene expression studies have crucial importance to quantify expression variations in Down syndrome tissues compared to normal ones. Herein, an in situ Quantitative Assessment of Gene Expression (QAGE) was used to quantify and statistically analyze, for the first time, DOPEY2 expression variations in different regions of the Down syndrome human fetal brains and to compare them to corresponding normal brains. DOPEY2, which is localized in the Down Syndrome Critical Region (DSCR) and is a candidate gene for neurological alterations in Down syndrome, showed a delimited regional and cellular expression pattern in the cortex, hippocampus and cerebellum, characterized by different transcriptional intensities in both normal and trisomic brains. DOPEY2 is overexpressed more than 50% (1.79-, 1.97- and 2.12-folds in the cortex, cerebellum and hippocampus, respectively), and showed statistically significant differences in the overexpression ratios in the three brain regions expressing DOPEY2. The demonstration of differential DOPEY2 expression and overexpression in human fetal brains suggests that this gene is submitted to a complex transcriptional control and could depend from other human chromosome 21 genes. Moreover, DOPEY2 overexpression in the brain regions, that are altered in Down syndrome patients and involved in learning and memory processes, is in agreement to the hypothesis that this gene plays a potential role in functional brain alterations and in the pathogenesis of mental retardation in Down syndrome. This new in situ QAGE approach allowed quantitative measurements of transcriptional changes and statistical evaluations of the expression and overexpression patterns of DOPEY2 at specific regions of the brain, which is a complementary approach to qRT-PCR and microarray for transcriptome study. Moreover, this approach could be a powerful tool to study the candidate chromosome 21 genes for Down syndrome and other pathologies caused by regionalized quantitative transcriptional alterations, for greater interpretation of functional processes driving gene expression.


Asunto(s)
Encéfalo/metabolismo , Síndrome de Down/genética , Regulación de la Expresión Génica , Discapacidad Intelectual/genética , Proteínas de Transporte Vesicular/genética , Encéfalo/anatomía & histología , Niño , Síndrome de Down/metabolismo , Perfilación de la Expresión Génica , Humanos , Lactante , Discapacidad Intelectual/metabolismo , Proteínas de Transporte Vesicular/metabolismo
3.
Eur J Paediatr Neurol ; 12(3): 168-82, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-17933568

RESUMEN

Down syndrome (DS), affecting 1/700 live births, is the major genetic cause of mental retardation (MR), a cognitive disorder with hard impact on public health. DS brain is characterized by a reduced cerebellar volume and number of granular cells, defective cortical lamination and reduced cortical neurons, malformed dendritic trees and spines, and abnormal synapses. These neurological alterations, also found in trisomic mouse models, result from gene-dosage effects of Human Chromosome 21 (HC21) on the expression of critical developmental genes. HC21 sequencing, mouse ortholog gene identification and DS mouse model generation lead to determine HC21 gene functions and the effects of protein-dosage alterations in neurodevelopmental and metabolic pathways in DS individuals. Trisomic brain transcriptome of DS patients and trisomic mouse models identified some molecular changes determined by gene-overdosage and associated dysregulation of some disomic gene expression in DS brains. These transcriptional variations cause developmental alterations in neural patterning and signal transduction pathways that may lead to defective neuronal circuits responsible for the pathogenesis of MR in DS. Recently, the first altered molecular pathway responsible of some DS phenotypes, including neurological and cognitive disorders has been identified. In this pathway, two critical HC21 genes (DYRK1A and DSCR1) act synergistically to control the phosphorylation levels of NFATc and NFATc-regulated gene expression. Interestingly, the NFATc mice show neurological dysfunctions similar to those seen in DS patients and trisomic mouse models. Treatment of DS mouse model Ts65Dn with GABA(A) antagonists allowed post-drug rescue of cognitive defects, indicating a hopeful direction in clinical therapies for MR in children with DS.


Asunto(s)
Encéfalo/patología , Cromosomas Humanos Par 21 , Síndrome de Down/genética , Discapacidad Intelectual/genética , Transducción de Señal/genética , Animales , Síndrome de Down/complicaciones , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Discapacidad Intelectual/etiología , Ratones , Fenotipo
4.
Biochem Biophys Res Commun ; 364(3): 488-94, 2007 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-17963726

RESUMEN

Down syndrome (DS) is the most frequent genetic cause of mental retardation (MR) associated with neurological alterations. To allow a genetic dissection of DS phenotype, we studied eight transgenic mouse lines carrying YACs containing human DNA fragments covering DS critical region (DCR-1), as an in vivo library. Herein, we found an increased brain size in the 152F7-mice containing DYRK1A gene. We also identified a new cerebellar alteration in two independent lines carrying 230E8-YAC. These mice showed significant elongation of the cerebellar antero-posterior axis (p<0.001), determined by increased length of rostral folia of the vermis (lobule II-V, p<0.0001; lobule VI, p<0.001). In addition, we identified a major neurological defect in culmen and declivus lobules in the 230E8-mice. We analyzed P30, P12, and P9 stages and detected high significant increased lengths of anterior lobules (II-VI) of 230E8-mice at P30 and P12 (lobule II-V, p<0.0001; lobule VI, p<0.05), but not at P9, indicating that this new phenotype appears between P9 and P12. Interestingly, 230E8-mice also present increased cortical cell density and mild learning defects. 230E8-YAC contains seven genes, some of which could be potentially responsible for this phenotype. Between them, we proposed DOPEY2 as potential candidate gene for these cerebellar alterations considering its high expression in the brain and that its homologous genes in yeast, Caenorhabditis elegans and Drosophila are involved in morphogenesis, suggesting a conserved role of DOPEY2 as a patterning gene.


Asunto(s)
Cerebelo/patología , Cerebelo/fisiopatología , Cromosomas Artificiales de Levadura/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Musculares/genética , Animales , Secuencia Conservada , Proteínas de Unión al ADN , Predisposición Genética a la Enfermedad/genética , Ratones , Ratones Transgénicos , Biblioteca de Péptidos , Fenotipo
5.
Neurosci Res ; 59(4): 349-69, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17897742

RESUMEN

Down syndrome (DS), the most frequent genetic disorder leading to mental retardation (MR), is caused by three copies of human chromosome 21 (HC21). Trisomic and transgenic mouse models for DS allow genetic dissection of DS neurological and cognitive disorders in view to identify genes responsible for these phenotypes. The effects of the gene dosage imbalance on DS phenotypes are explained by two hypotheses: the "gene dosage effect" hypothesis claims that a DS critical region, containing a subset of dosage-sensitive genes, determines DS phenotypes, and the "amplified developmental instability" hypothesis holds that HC21 trisomy determines general alteration in developmental homeostasis. Transcriptome and expression studies showed different up- or down-expression levels of genes located on HC21 and the other disomic chromosomes. HC21 genes, characterized by their overexpression in brain regions affected in DS patients and by their contribution to neurological and cognitive defects when overexpressed in mouse models, are proposed herein as good candidates for MR. In this article, we propose a new molecular and cellular mechanism explaining MR pathogenesis in DS. In this model, gene dosage imbalance effects on transcriptional variations are described considering the nature of gene products and their functional relationships. These transcriptional variations may affect different aspects of neuronal differentiation and metabolism and finally, determine the brain neuropathologies and mental retardation in DS.


Asunto(s)
Síndrome de Down/genética , Dosificación de Gen/genética , Predisposición Genética a la Enfermedad/genética , Discapacidad Intelectual/genética , Animales , Química Encefálica/genética , Cromosomas Humanos Par 21/genética , Modelos Animales de Enfermedad , Síndrome de Down/metabolismo , Síndrome de Down/fisiopatología , Regulación de la Expresión Génica/genética , Humanos , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/fisiopatología , Ratones , Elementos Reguladores de la Transcripción/genética
6.
Biochem Biophys Res Commun ; 339(1): 296-304, 2006 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-16307728

RESUMEN

The mouse homeobox gene Barhl1 plays a central role in cerebellum development and its expression is activated by the transcription factor Math1 which is involved in bone morphogenetic protein response pathways. We studied the human ortholog BARHL1 and we found that human, mouse, monkey, rat, and zebrafish orthologs were highly conserved and are members of the BarH homeogene family, containing Drosophila BarH1 and BarH2. The N-terminus of BARHL1 protein presents two FIL domains and an acidic domain rich in serine/threonine and proline, while the C-terminus contains a canonical proline-rich domain. Secondary structure analysis showed that outside the three helixes of the homeodomain, BARHL1 protein has essentially random coil structure. We isolated BARHL1 and defined its expression pattern in human embryonic and fetal central nervous system (CNS) and compared it to the mouse Barhl1 transcription. BARHL1 mRNA was found exclusively in the CNS restricted to p1-p4 prosomeres of the diencephalon, to the dorsal cells of the mesencephalon, to the dorsal dl1 sensory neurons of the spinal cord, and to the rhombic lips yielding the cerebellar anlage. Detailed analysis of BARHL1 expression in fetal cerebellar cell layers using our new optic microscopy technology showed BARHL1 expression in external and internal granular cells and also in mouse adult granular cells, in agreement to Barhl1 null mouse phenotype affecting the differentiation and migration of granular cells. These findings indicate that the regional and cellular specificities of BARHL1 transcriptional control well correspond to the mouse Barhl1 transcription and suggest a potential role of this gene in the differentiation of BARHL1-expressing neuronal progenitors involved in the pattern formation of human cerebral and cerebellar structures.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/citología , Encéfalo/embriología , Cerebelo/citología , Cerebelo/embriología , Cerebelo/metabolismo , Proteínas de Homeodominio/genética , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Estructura Terciaria de Proteína , Proteínas Represoras , Homología de Secuencia de Aminoácido
7.
Int J Dev Neurosci ; 24(1): 35-44, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16384683

RESUMEN

The mouse Barhl1 homeogene, member of the BarH subfamily, play a crucial role in the cerebellum development and its human ortholog BARHL1 has been proposed as a positional and functional candidate gene for the Joubert syndrome, a form of cerebellar ataxia. The Barhl1 expression has been demonstrated to be induced by the transcription factor Math1 involved in BMP responses. We isolated the mouse Barhl1 by screening of a cDNA library with the Xenopus Xvent-2, member of the BarH subfamily, which acts in the BMP4 pathway during embryonic patterning and neural plate differentiation. We studied the detailed Barhl1 expression pattern and showed its transcription in spatio-temporally and functionally restricted domains of the developing central nervous system (CNS). Using our new optical microscopy technology, we compare the transcript steady state level and cell density in the Barhl1-expressing regions. We found that Barhl1 was transcribed in superior and inferior colliculi in the dorsal mesencephalon at a relatively low transcriptional level. In the diencephalon, Barhl1 was found higher expressed first within the basal plate and later in the mammillary region. In the cerebellum, Barhl1 showed the highest transcriptional level restricted to the anterior and posterior rhombic lips giving rise to the external and internal cerebellar granular cells and to the deep nuclei. In the spinal cord, Barhl1 showed similar expression level than in the cerebellum and is delimited to a subset of dorsal interneurons. Therefore, our results indicated that Barhl1 homeodomain gene is exclusively transcribed in restricted CNS domain at differential transcription levels which suggest a highly regulated transcriptional mechanism. In addition, these regional and cellular specificities indicated that Barhl1 may be involved in the differentiation of the specific subsets of neuronal progenitors.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Proteínas de Homeodominio/metabolismo , Morfogénesis , Proteínas del Tejido Nervioso/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Secuencia de Aminoácidos , Animales , Encéfalo/anatomía & histología , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Hibridación in Situ , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Proteínas Represoras/genética , Alineación de Secuencia
8.
DNA Res ; 12(3): 203-10, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16303751

RESUMEN

Availability of the human genome sequence promises important progress in the understanding of human pathologies, particularly for multifactorial diseases. Among these, Down syndrome (DS) is the most frequent genetic cause of mental retardation. A critical region of chromosome 21, the Down syndrome Chromosomal Region-1 (DCR-1), is responsible for many features of the DS phenotype including mental retardation. We studied DCR-1 C21orf5 as a new candidate gene for DS considering its restricted expression in key brain regions altered in DS patients and involved in learning and memory processes. To elucidate C21orf5 molecular function, we performed a comparative study of protein sequences in several species and showed that C21orf5 represents a new member of the Dopey leucine zipper-like family. The C21orf5 C-termini contains two highly conserved leucine-like zipper domains in invertebrate and vertebrate species. Evolution analysis indicated a common ancestral origin of these protein sequences also suggesting a conserved function of this gene throughout phylogenesis. Mutations of the known C21orf5 homologous genes Aspergillus nidulans DopA, Saccharomyces cerevisiae Dop1 and Caenorhabditis elegans pad1, determine morphological abnormalities. We studied transgenic mice carrying the human C21orf5 gene and we showed that this gene is overexpressed in brain regions by in situ hybridization and by real-time RT-PCR experiments. Interestingly, we also showed that these transgenic mice have an increased density of cortical cells overexpressing C21orf5. Similarly, DS patients have an altered lamination pattern in their cortex. Considering together our and previous findings, we suggest that the human dopey family member, C21orf5, could play a role in brain morphogenesis and, when overexpressed, it could participate in neurological features and mental retardation observed in DS patients.


Asunto(s)
Síndrome de Down/genética , Proteínas de Transporte Vesicular/genética , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 21/metabolismo , Síndrome de Down/metabolismo , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Morfogénesis , Filogenia , Homología de Secuencia de Aminoácido , Proteínas de Transporte Vesicular/metabolismo
9.
Int J Dev Neurosci ; 23(5): 475-84, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15946822

RESUMEN

Human SIM2 is the ortholog of Drosophila single-minded (sim), a master regulator of neurogenesis and transcriptional factor controlling midline cell fate determination. We previously localized SIM2 in a chromosome 21 critical region for Down syndrome (DS). Here, we studied SIM2 gene using a new approach to provide insights in understanding of its potential role in human development. For the first time, we showed SIM2 spatial and temporal expression pattern during human central nervous system (CNS) development, from embryonic to fetal stages. Additional investigations were performed using a new optic microscopy technology to compare signal intensity and cell density [M. Rachidi, C. Lopes, S. Gassanova, P.M. Sinet, M. Vekemans, T. Attie, A.L. Delezoide, J.M. Delabar, Regional and cellular specificity of the expression of TPRD, the tetratricopeptide Down syndrome gene, during human embryonic development, Mech. Dev. 93 (2000) 189--193]. In embryonic stages, SIM2 was identified predominantly in restricted regions of CNS, in ventral part of D1/D2 diencephalic neuroepithelium, along the neural tube and in a few cell subsets of dorsal root ganglia. In fetal stages, SIM2 showed differential expression in pyramidal and granular cell layers of hippocampal formation, in cortical cells and in cerebellar external granular and Purkinje cell layers. SIM2 expression in embryonic and fetal brain could suggest a potential role in human CNS development, in agreement with Drosophila and mouse Sim mutant phenotypes and with the conservation of the Sim function in CNS development from Drosophila to Human. SIM2 expression in human fetal brain regions, which correspond to key structures for cognitive processes, correlates well with the behavioral phenotypes of Drosophila Sim mutants and transgenic mice overexpressing Sim2. In addition, SIM2-expressing brain regions correspond to the altered structures in DS patients. All together, these findings suggest a potential role of SIM2 in CNS development and indicate that SIM2 overexpression could participate to the pathogenesis of mental retardation in Down syndrome patients.


Asunto(s)
Encéfalo/embriología , Síndrome de Down/embriología , Factores de Transcripción/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Embrión de Mamíferos/metabolismo , Feto/metabolismo , Edad Gestacional , Humanos , Factores de Tiempo , Distribución Tisular
10.
Biochem Biophys Res Commun ; 305(4): 915-24, 2003 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-12767918

RESUMEN

Mental retardation represents the more invalidating pathological aspect of Down syndrome, DS, and has a hard impact in public health. Modifications in DS brain, concerning abnormal size, neuronal differentiation, and cell density, cause changes in the neurophysiology and behavior of DS patients, and could be determined by dosage imbalance of genes localized in the DS critical region, DCR. Among these genes, C21orf5 showed high homology with Caenorhabditis elegans Pad1 involved in cellular differentiation and patterning. To shed light on C21orf5 role in DS, we performed molecular characterization of human and mouse orthologs, their spatio-temporal expression during development and in adult, and overexpression in DS and transgenic mice. C21orf5 was widely expressed early in embryogenesis in the nervous system. Later, its expression became differential and increased in mesencephalon and rhomboencephalon. This developmental expression profile evolves selectively in adult brain with higher signals in hippocampus, cerebellum, perirhinal, and entorhinal cortex, compared to the other cortical regions. Cellular specificity was detected in hippocampus with higher C21orf5 mRNA level in CA3 cells. Our findings appoint C21orf5 as candidate gene for mental retardation: Its overexpression in DS cells may contribute to gene imbalance in DS.Its specific expression in normal and its mirroring pattern in transgenic mice correspond to abnormal regions in DS patients and to neurological phenotype of transgenic mice. Altered cortical lamination in transgenic mice and the Pad1 ortholog function suggest a potential role of C21orf5 in cell differentiation. Its patterned differential expression in the medial temporal-lobe system, including hippocampal formation and perirhinal cortex involved in memory storage, and learning and memory defects in the transgenic mice suggest a specialized role for C21orf5 in cognitive processes. These evidences suggest that C21orf5 is an attractive candidate gene contributing to neurological alterations responsible for mental retardation in DS patients.


Asunto(s)
Síndrome de Down/etiología , Proteínas de la Membrana/fisiología , Lóbulo Temporal/metabolismo , Animales , Encéfalo/metabolismo , Síndrome de Down/genética , Síndrome de Down/metabolismo , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario y Fetal , Femenino , Humanos , Hibridación in Situ , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Distribución Tisular , Transcripción Genética , Proteínas de Transporte Vesicular
11.
Cell Tissue Res ; 310(2): 237-52, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12397378

RESUMEN

Transgenic flies that can drive GAL4 expression under the control of the 7 kb 5'-region of the Drosophila Ca(2+)/calmodulin-dependent protein kinase II (dCaMKII) gene (dCaMKII-GAL4) were established. Characteristic features of this dCaMKII-GAL4 driven reporter expression were compatible with the endogenous dCaMKII expression pattern: The dCaMKII-GAL4 driven reporter gene was expressed preferentially in the central nervous system of the embryo and larvae. Reporter expression was also observed in the brain, thoracic ganglion, and gut of the adult. The whole-brain distribution and projections of dCaMKII-GAL4-expressing cells in the adults were visualized three-dimensionally by using UAS-linked reporter genes. Prominent signals of nuclear-localized beta-Gal reporter gene expression were found in extensive brain regions, especially in the Kenyon cells of the mushroom body (MB), cells in the pars intercerebralis, and subesophageal ganglion (SOG). tau reporter gene expression highlighting neurite projections was detected in the MB lobes, median bundle, antennal lobe glomeruli, and fibers of clusters in the SOG, ventrolateral protocerebrum and superior lateral protocerebrum. These observations agree with those of a previous study mapping the dCaMKII-dependent memory circuits in courtship conditioning. Interestingly, green fluorescent protein reporter gene expression in adult MB lobes was predominantly observed in the alpha and beta lobes with a core-deficient pattern, but not in the alpha' and beta' lobes, similar to Fasciclin II immunoreactivity.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Drosophila/genética , Regulación del Desarrollo de la Expresión Génica , Genes de Insecto , Proteínas Asociadas a Microtúbulos , Neuropéptidos/metabolismo , Regiones Promotoras Genéticas , Animales , Animales Modificados Genéticamente , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Sistema Nervioso Central/metabolismo , Proteínas de Dominio Doblecortina , Drosophila/embriología , Embrión no Mamífero , Genes Reporteros , Proteínas Fluorescentes Verdes , Inmunohistoquímica , Larva , Proteínas Luminiscentes/metabolismo , Cuerpos Pedunculados/química , Cuerpos Pedunculados/metabolismo , Neuropéptidos/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/genética , Distribución Tisular , Transgenes/genética , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA