Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neural Plast ; 2016: 4235898, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27547454

RESUMEN

One of the most challenging questions in neuroscience is to dissect how learning and memory, the foundational pillars of cognition, are grounded in stable, yet plastic, gene expression states. All known epigenetic mechanisms such as DNA methylation and hydroxymethylation, histone modifications, chromatin remodelling, and noncoding RNAs regulate brain gene expression, both during neurodevelopment and in the adult brain in processes related to cognition. On the other hand, alterations in the various components of the epigenetic machinery have been linked to well-known causes of intellectual disability disorders (IDDs). Two examples are Down Syndrome (DS) and Fragile X Syndrome (FXS), where global and local epigenetic alterations lead to impairments in synaptic plasticity, memory, and learning. Since epigenetic modifications are reversible, it is theoretically possible to use epigenetic drugs as cognitive enhancers for the treatment of IDDs. Epigenetic treatments act in a context specific manner, targeting different regions based on cell and state specific chromatin accessibility, facilitating the establishment of the lost balance. Here, we discuss epigenetic studies of IDDs, focusing on DS and FXS, and the use of epidrugs in combinatorial therapies for IDDs.


Asunto(s)
Cognición/fisiología , Síndrome de Down/genética , Epigénesis Genética/genética , Síndrome del Cromosoma X Frágil/genética , Interacción Gen-Ambiente , Terapia Genética/tendencias , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Catequina/análogos & derivados , Catequina/farmacología , Catequina/uso terapéutico , Cognición/efectos de los fármacos , Metilación de ADN/genética , Síndrome de Down/psicología , Síndrome de Down/terapia , Epigénesis Genética/efectos de los fármacos , Síndrome del Cromosoma X Frágil/psicología , Síndrome del Cromosoma X Frágil/terapia , Terapia Genética/métodos , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/psicología , Discapacidad Intelectual/terapia , Dominios y Motivos de Interacción de Proteínas/genética
2.
Cereb Cortex ; 22(12): 2867-77, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22215728

RESUMEN

Down syndrome (DS) is the most frequent genetic cause of mental retardation. Cognitive dysfunction in these patients is correlated with reduced dendritic branching and complexity, along with fewer spines of abnormal shape that characterize the cortical neuronal profile of DS. DS phenotypes are caused by the disruptive effect of specific trisomic genes. Here, we report that overexpression of dual-specificity tyrosine phosphorylation-regulated kinase 1A, DYRK1A, is sufficient to produce the dendritic alterations observed in DS patients. Engineered changes in Dyrk1A gene dosage in vivo strongly alter the postnatal dendritic arborization processes with a similar progression than in humans. In cultured mammalian cortical neurons, we determined a reduction of neurite outgrowth and synaptogenesis. The mechanism underlying neurite dysgenesia involves changes in the dynamic reorganization of the cytoskeleton.


Asunto(s)
Corteza Cerebral/metabolismo , Citoesqueleto/metabolismo , Síndrome de Down/metabolismo , Neurogénesis , Neuronas/metabolismo , Neuronas/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Corteza Cerebral/patología , Citoesqueleto/patología , Síndrome de Down/patología , Ratones , Ratones Transgénicos , Quinasas DyrK
3.
Dev Neurosci ; 33(5): 442-50, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21865666

RESUMEN

Individuals with Down syndrome (DS) present cognitive deficits that can be improved by early implementation of special care programs. However, they showed limited and temporary cognitive effects. We previously demonstrated that postnatal environmental enrichment (EE) improved clearly, though temporarily, the execution of visuospatial memory tasks in Ts65Dn mice, a DS model bearing a partial trisomy of murine chromosome 16; but in contrast to wild-type littermates, there was a lack of structural plasticity in pyramidal cell structure in the trisomic cerebral cortex. In the present study, we have investigated the impact of EE on the function of adenylyl cyclase and phospholipase C as a possible mechanism underlying the time-limited improvements observed. Basal production of cyclic adenosine monophosphate (cAMP) was not affected, but responses to GTPγS, isoprenaline, noradrenaline, SKF 38393 and forskolin were depressed in the Ts65Dn hippocampus. In EE conditions, cAMP accumulation was not significantly modified in control animals with respect to nonenriched controls. However, EE had a marked effect in Ts65Dn mice, in which cAMP production was significantly increased. Similarly, EE increased phospholipase C activity in Ts65Dn mice, in response to carbachol and calcium. We conclude that EE restores the G-protein-associated signal transduction systems that are altered in Ts65Dn mice.


Asunto(s)
Síndrome de Down/fisiopatología , Ambiente , Proteínas de Unión al GTP/metabolismo , Transducción de Señal/fisiología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Agonistas alfa-Adrenérgicos/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Carbacol/farmacología , Agonistas Colinérgicos/farmacología , Colforsina/farmacología , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Agonistas de Dopamina/farmacología , Femenino , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Norepinefrina/farmacología
4.
Sci Rep ; 10(1): 16023, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32994493

RESUMEN

Down syndrome (DS) is the main genetic cause of intellectual disability due to triplication of human chromosome 21 (HSA21). Although there is no treatment for intellectual disability, environmental enrichment (EE) and the administration of green tea extracts containing epigallocatechin-3-gallate (EGCG) improve cognition in mouse models and individuals with DS. Using proteome, and phosphoproteome analysis in the hippocampi of a DS mouse model (Ts65Dn), we investigated the possible mechanisms underlying the effects of green tea extracts, EE and their combination. Our results revealed disturbances in cognitive-related (synaptic proteins, neuronal projection, neuron development, microtubule), GTPase/kinase activity and chromatin proteins. Green tea extracts, EE, and their combination restored more than 70% of the phosphoprotein deregulation in Ts65Dn, and induced possible compensatory effects. Our downstream analyses indicate that re-establishment of a proper epigenetic state and rescue of the kinome deregulation may contribute to the cognitive rescue induced by green tea extracts.


Asunto(s)
Camellia sinensis/química , Cognición/efectos de los fármacos , Síndrome de Down/psicología , Extractos Vegetales/administración & dosificación , Proteómica/métodos , Animales , Catequina/administración & dosificación , Catequina/análogos & derivados , Catequina/farmacología , Cromatografía Liquida , Modelos Animales de Enfermedad , Síndrome de Down/genética , Epigénesis Genética/efectos de los fármacos , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Extractos Vegetales/química , Extractos Vegetales/farmacología , Mapas de Interacción de Proteínas/efectos de los fármacos , Espectrometría de Masas en Tándem
5.
Neuroscience ; 158(4): 1234-50, 2009 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-19121372

RESUMEN

The involvement of brain-derived neurotrophic factor (BDNF) in cognitive processes and the decrease in its expression in Huntington's disease suggest that this neurotrophin may play a role in learning impairment during the disease progression. We therefore analyzed the onset and severity of cognitive deficits in two different mouse models with the same mutant huntingtin but with different levels of BDNF (R6/1 and R6/1:BDNF+/- mice). We observed that BDNF modulates cognitive function in different learning tasks, even before the onset of motor symptoms. R6/1:BDNF+/- mice showed earlier and more accentuated cognitive impairment than R6/1 mice at 5 weeks of age in discrimination learning; at 5 weeks of age in procedural learning; and at 9 weeks of age in alternation learning. At the earliest age at which cognitive impairment was detected, electrophysiological analysis was performed in the hippocampus. All mutant genotypes showed reduced hippocampal long term potentiation (LTP) with respect to wild type but did not show differences between them. Thus, we evaluated the involvement of BDNF-trkB signaling and glutamate receptor expression in the hippocampus of these mice. We observed a decrease in phospholipaseCgamma activity, but not ERK, in R61, BDNF+/- and R6/1:BDNF+/- hippocampus at the age when LTP was altered. However, a specific decrease in the expression of glutamate receptors NR1, NR2A and GluR1 was detected only in R6/1:BDNF+/- hippocampus. Therefore, these results show that BDNF modulates the learning and memory alterations induced by mutant huntingtin. This interaction leads to intracellular changes, such as specific changes in glutamate receptors and in BDNF-trkB signaling through phospholipaseCgamma.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Trastornos del Conocimiento/fisiopatología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Fosfolipasa C gamma/metabolismo , Receptores de Glutamato/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Biofisica , Factor Neurotrófico Derivado del Encéfalo/genética , Trastornos del Conocimiento/genética , Aprendizaje Discriminativo/fisiología , Modelos Animales de Enfermedad , Estimulación Eléctrica/métodos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Proteína Huntingtina , Técnicas In Vitro , Potenciación a Largo Plazo/genética , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Ratones Transgénicos , Mutación , Técnicas de Placa-Clamp/métodos , Receptores AMPA/genética , Receptores AMPA/metabolismo , Receptores de Glutamato/genética , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Natación
6.
Neurobiol Dis ; 32(3): 377-84, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18773961

RESUMEN

Transgenic mice overexpressing Dyrk1A (TgDyrk1A), a Down syndrome (DS) candidate gene, exhibit motor and cognitive alterations similar to those observed in DS individuals. To gain new insights into the molecular consequences of Dyrk1A overexpression underlying TgDyrk1A and possibly DS motor phenotypes, microarray studies were performed. Transcriptome analysis showed an upregulation of the NR2A subunit of the NMDA type of glutamate receptors in TgDyrk1A cerebellum. NR2A protein overexpression was also detected in TgDyrk1A cerebellar homogenates, in the synaptosome-enriched fraction and in TgDyrk1A primary cerebellar granular neuronal cultures (CGNs). In TgDyrk1A synaptosomes, calcium-imaging experiments showed a higher calcium uptake after NMDA stimulation. Similarly, NMDA administration promoted longer calcium transients in TgDyrk1A CGNs. Taken together, these results show that NMDA-induced calcium rise is altered in TgDyrk1A cerebellar neurons and indicate that calcium signaling is dysregulated in TgDyrk1A mice cerebella. These findings suggest that DYRK1A overexpression might contribute to the dysbalance in the excitatory transmission found in the cerebellum of DS individuals and DS mouse models.


Asunto(s)
Calcio/metabolismo , Cerebelo/metabolismo , Síndrome de Down/genética , N-Metilaspartato/farmacología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Tirosina Quinasas/fisiología , Receptores de N-Metil-D-Aspartato/genética , Regulación hacia Arriba , Animales , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Síndrome de Down/metabolismo , Perfilación de la Expresión Génica , Inmunohistoquímica , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sinaptosomas/metabolismo , Quinasas DyrK
7.
Cytogenet Genome Res ; 121(1): 67-77, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18544929

RESUMEN

Down syndrome (DS), trisomy of human chromosome 21, is the most common genetic cause of intellectual disability. With an incidence in some countries as high as one in approximately 700 live births, and a complex, extensive and variably severe phenotype, Down syndrome is a significant medical and social challenge. In recent years, there has been a rapid increase in information on the functions of the genes of human chromosome 21, as well as in techniques and resources for their analysis. A recent workshop brought together experts on the molecular biology of Down syndrome and chromosome 21 with interested researchers in other fields to discuss advances and potentials for generating gene-phenotype correlations. An additional goal of the workshop was to work towards identification of targets for therapeutics that will correct features of DS. A knowledge-based approach to therapeutics also requires the correlation of chromosome 21 gene function with phenotypic features.


Asunto(s)
Cromosomas Humanos Par 21/genética , Síndrome de Down/genética , Animales , Citogenética , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Síndrome de Down/terapia , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , MicroARNs/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Musculares/genética , Sistema Nervioso/crecimiento & desarrollo , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Proteína de Unión al GTP cdc42/genética , Quinasas DyrK
8.
Amino Acids ; 35(2): 339-43, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18163181

RESUMEN

Down syndrome (DS) is the most common human chromosomal abnormality caused by an extra copy of chromosome 21. The phenotype of DS is thought to result from overexpression of a gene or genes located on the triplicated chromosome or chromosome region. Several reports have shown that the neuropathology of DS comprises developmental abnormalities and Alzheimer-like lesions such as senile plaques. A key component of senile plaques is amyloid beta-peptide which is generated from the amyloid precursor protein (APP) by sequential action of beta-secretases (BACE1 and BACE2) and gamma-secretase. While BACE1 maps to chromosome 11, APP and BACE2 are located on chromosome 21. To challenge the gene dosage effect and gain insight into the expressional relation between beta-secretases and APP in DS brain, we evaluated protein expression levels of BACE1, BACE2 and APP in fetal and adult DS brain compared to controls. In fetal brain, protein expression levels of BACE2 and APP were comparable between DS and controls. BACE1 was increased, but did not reach statistical significance. In adult brain, BACE1 and BACE2 were comparable between DS and controls, but APP was significantly increased. We conclude that APP overexpression seems to be absent during the development of DS brain up to 18-19 weeks of gestational age. However, its overexpression in adult DS brain could lead to disturbance of normal function of APP contributing to neurodegeneration. Comparable expression of BACE1 and BACE2 speaks against the hypothesis that increased beta-secretase results in (or even underlies) increased production of amyloidogenic A beta fragments. Furthermore, current data indicate that the DS phenotype cannot be fully explained by simple gene dosage effect.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/biosíntesis , Precursor de Proteína beta-Amiloide/biosíntesis , Ácido Aspártico Endopeptidasas/biosíntesis , Corteza Cerebral/metabolismo , Síndrome de Down/metabolismo , Secretasas de la Proteína Precursora del Amiloide/análisis , Precursor de Proteína beta-Amiloide/análisis , Ácido Aspártico Endopeptidasas/análisis , Western Blotting , Corteza Cerebral/química , Corteza Cerebral/patología , Síndrome de Down/patología , Femenino , Feto , Humanos , Persona de Mediana Edad
9.
eNeuro ; 5(6)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30637329

RESUMEN

Obesogenic diets lead to overeating and obesity by inducing the expression of genes involved in hedonic and homeostatic responses in specific brain regions. However, how the effects on gene expression are coordinated in the brain so far remains largely unknown. In our study, we provided mice with access to energy-dense diet, which induced overeating and overweight, and we explored the transcriptome changes across the main regions involved in feeding and energy balance: hypothalamus, frontal cortex, and striatum. Interestingly, we detected two regulatory processes: a switch-like regulation with differentially expressed (DE) genes changing over 1.5-fold and "fine-tuned" subtler changes of genes whose levels correlated with body weight and behavioral changes. We found that genes in both categories were positioned within specific topologically associated domains (TADs), which were often differently regulated across different brain regions. These TADs were enriched in genes relevant for the physiological and behavioral observed changes. Our results suggest that chromatin structure coordinates diet-dependent transcriptional regulation.


Asunto(s)
Encéfalo/metabolismo , Cromatina/metabolismo , Regulación de la Expresión Génica/fisiología , Expresión Génica/fisiología , Homeostasis/fisiología , Sobrepeso/patología , Sobrepeso/fisiopatología , Animales , Conducta Compulsiva , Biología Computacional , Correlación de Datos , Dieta/efectos adversos , Conducta Alimentaria/fisiología , Femenino , Aseo Animal , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Modelos Biológicos , Comportamiento de Nidificación/fisiología , Sobrepeso/etiología
10.
Free Radic Biol Med ; 114: 122-130, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28958596

RESUMEN

Down syndrome (DS) is caused by the trisomy of human chromosome 21 and is the most common genetic cause of intellectual disability. In addition to the intellectual deficiencies and physical anomalies, DS individuals present a higher prevalence of obesity and subsequent metabolic disorders than healthy adults. There is increasing evidence from both clinical and experimental studies indicating the association of visceral obesity with a pro-inflammatory status and recent studies have reported that obese people with DS suffer from low-grade systemic inflammation. However, the link between adiposity and inflammation has not been explored in DS. Here we used Ts65Dn mice, a validated DS mouse model, for the study of obesity-related inflammatory markers. Ts65Dn mice presented increased energy intake, and a positive energy balance leading to increased adiposity (fat mass per body weight), but did not show overweight, which only was apparent upon high fat diet induced obesity. Trisomic mice also had fasting hyperglycemia and hypoinsulinemia, and normal incretin levels. Those trisomy-associated changes were accompanied by reduced ghrelin plasma levels and slightly but not significantly increased leptin levels. Upon a glucose load, Ts65Dn mice showed normal increase of incretins accompanied by over-responses of leptin and resistin, while maintaining the hyperglycemic and hypoinsulinemic phenotype. These changes in the adipoinsular axis were accompanied by increased plasma levels of inflammatory biomarkers previously correlated with obesity galectin-3 and HSP72, and reduced IL-6. Taken together, these results suggest that increased adiposity, and pro-inflammatory adipokines leading to low-grade inflammation are important players in the propensity to obesity in DS. We conclude that DS would be a case of impaired metabolic-inflammatory axis.


Asunto(s)
Modelos Animales de Enfermedad , Síndrome de Down/complicaciones , Mediadores de Inflamación/sangre , Obesidad/etiología , Animales , Síndrome de Down/sangre , Síndrome de Down/patología , Ratones , Obesidad/sangre , Obesidad/patología , Factores de Riesgo
11.
Genes Brain Behav ; 6 Suppl 1: 2-23, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17543035

RESUMEN

Panic disorder is a major cause of medical attention with substantial social and health service cost. Based on pharmacological studies, research on its etiopathogenesis has been focused on the possible dysfunction of specific neurotransmitter systems. However, recent work has related the genes involved in development, synaptic plasticity and synaptic remodeling to anxiety disorders. This implies that learning processes and changes in perception, interpretation and behavioral responses to environmental stimuli are essential for development of complex anxiety responses secondary to the building of specific brain neural circuits and to adult plasticity. The focus of this review is on progress achieved in identifying genes that confer increased risk for panic disorder through genetic epidemiology and the use of genetically modified mouse models. The integration of human and animal studies targeting behavioral, systems-level, cellular and molecular levels will most probably help identify new molecules with potential impact on the pathogenetic aspects of the disease.


Asunto(s)
Factores de Crecimiento Nervioso/genética , Neurotransmisores/genética , Trastorno de Pánico/genética , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Modelos Genéticos
12.
Genes Brain Behav ; 6(6): 569-78, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17137466

RESUMEN

The dual-specificity tyrosine-phosphorylated and regulated kinase 1A (DYRK1A) gene encodes a protein kinase known to play a critical role in neurodevelopment. Mice with one functional copy of Dyrk1A (Dyrk1A(+/-)) display a marked hypoactivity and altered gait dynamics in basal conditions and in novel environments. Dopamine (DA) is a key neurotransmitter in motor behavior and genetic deletion of certain genes directly related to the dopaminergic system has a strong impact on motor activity. We have studied the effects of reduced Dyrk1A expression on the function of the nigrostriatal dopaminergic system. To characterize the dopaminergic system in DYRK1A(+/-) mice, we have used behavioral, pharmacological, histological, neurochemical and neuroimaging (microPET) techniques in a multidisciplinary approach. Dyrk1A(+/-) mice exhibited decreased striatal DA levels, reduced number of DA neurons in the substantia nigra pars compacta, as well as altered behavioral responses to dopaminergic agents. Moreover, microdialysis experiments revealed attenuated striatal DA release and positron emission tomography scan display reduced forebrain activation when challenged with amphetamine, in Dyrk1A(+/-) compared with wild-type mice. These data indicate that Dyrk1A is essential for a proper function of nigrostriatal dopaminergic neurons and suggest that Dyrk1A(+/-) mice can be used to study the pathogenesis of motor disorders involving dopaminergic dysfunction.


Asunto(s)
Dopamina/metabolismo , Actividad Motora/fisiología , Neostriado/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Sustancia Negra/enzimología , Animales , Femenino , Heterocigoto , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microdiálisis , Vías Nerviosas/metabolismo , Tomografía de Emisión de Positrones , Prosencéfalo/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Tirosina 3-Monooxigenasa/metabolismo , Quinasas DyrK
13.
Amino Acids ; 33(4): 677-88, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17111100

RESUMEN

Sensitivity to pharmacological challenges has been reported in patients with panic disorder. We have previously validated transgenic mice overexpressing the neurotrophin-3 (NT-3) receptor, TrkC (TgNTRK3), as an engineered murine model of panic disorder. We could determine that TgNTRK3 mice presented increased cellularity in brain regions, such as the locus ceruleus, that are important neural substrates for the expression of anxiety in severe anxiety states. Here, we investigated the sensitivity to induce anxiety and panic-related symptoms by sodium lactate and the effects of various drugs (the alpha2-adrenoceptor antagonist, yohimbine and the adenosine antagonist, caffeine), in TgNTRK3 mice. We found enhanced panicogenic sensitivity to sodium lactate and an increased intensity and a differential pattern of Fos expression after the administration of yohimbine or caffeine in TgNTRK3. Our findings validate the relevance of the NT-3/TrkC system to pathological anxiety and raise the possibility that a specific set of fear-related pathways involved in the processing of anxiety-related information may be differentially activated in panic disorder.


Asunto(s)
Ansiedad/inducido químicamente , Modelos Animales de Enfermedad , Miedo , Proteínas Oncogénicas v-fos/metabolismo , Trastorno de Pánico/metabolismo , Lactato de Sodio/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cafeína/metabolismo , Cafeína/farmacología , Miedo/psicología , Masculino , Ratones , Ratones Transgénicos , Trastorno de Pánico/psicología , Distribución Aleatoria , Yohimbina/farmacología
14.
Genes Brain Behav ; 5 Suppl 2: 48-60, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16681800

RESUMEN

Mental retardation (MR) is a developmental brain disorder characterized by impaired cognitive performance and adaptive skills that affects 1-2% of the population. During the last decade, a large number of genes have been cloned that cause MR upon mutation in humans. The causal role of these genes provides an excellent starting point to investigate the cellular, neurobiological and behavioral alterations and mechanisms responsible for the cognitive impairment in mentally retarded persons. However, studies on Down syndrome (DS) reveal that overexpression of a cluster of genes and various forms of MR that are caused by single-gene mutations, such as fragile X (FraX), Rett, Coffin-Lowry, Rubinstein-Taybi syndrome and non-syndromic forms of MR, causes similar phenotypes. In spite of the many differences in the manifestation of these forms of MR, evidence converges on the proposal that MR is primarily due to deficiencies in neuronal network connectivity in the major cognitive centers in the brain, which secondarily results in impaired information processing. Although MR has been largely regarded as a brain disorder that cannot be cured, our increased understanding of the abnormalities and mechanisms underlying MR may provide an avenue for the development of therapies for MR. In this review, we discuss the neurobiology underlying MR, with a focus on FraX and DS.


Asunto(s)
Dendritas/patología , Síndrome de Down/genética , Síndrome del Cromosoma X Frágil/genética , Discapacidad Intelectual/genética , Vías Nerviosas/patología , Animales , Dendritas/genética , Modelos Animales de Enfermedad , Síndrome de Down/complicaciones , Síndrome de Down/patología , Síndrome del Cromosoma X Frágil/complicaciones , Síndrome del Cromosoma X Frágil/patología , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Discapacidad Intelectual/etiología , Discapacidad Intelectual/patología , Ratones , Proteínas del Tejido Nervioso/genética , Vías Nerviosas/citología , Vías Nerviosas/fisiopatología , Fenotipo
15.
Amino Acids ; 31(4): 449-56, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16583316

RESUMEN

Premature aging and neuropathological features of Alzheimer's disease (AD) are commonly observed in Down syndrome (DS). Based on previous findings in a DS mouse model, the function of signaling pathways associated with adenylyl cyclase (AC) and phospholipase C (PLC) was assessed in cerebral cortex and cerebellum of age-matched adults with DS, AD, and controls. Basal production of cAMP was reduced in DS but not in AD cortex, and in both, DS and AD cerebellum. Responses to GTPgammaS, noradrenaline, SKF 38393 and forskolin were more depressed in DS than in AD cortex and cerebellum. Although no differences in PLC activity among control, DS and AD cortex were observed under basal and GTPgammaS- or Ca-stimulated conditions, the response of DS cortex to serotonergic and cholinergic stimulation was depressed, and that of AD was only impaired at cholinergic stimulation. No differences were documented in cerebellum. Our results demonstrate that PLC and AC were severely disturbed in the aged DS and AD brains, but the alterations in DS were more severe, and differed to some extent from those observed in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Cerebelo/metabolismo , Corteza Cerebral/metabolismo , Síndrome de Down/metabolismo , Proteínas de Unión al GTP/metabolismo , Transducción de Señal , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Adenilil Ciclasas/metabolismo , Anciano , Cerebelo/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Colforsina/farmacología , AMP Cíclico/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Humanos , Masculino , Persona de Mediana Edad , Norepinefrina/farmacología , Fosfolipasas de Tipo C/metabolismo
16.
Prog Neurobiol ; 74(2): 111-26, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15518956

RESUMEN

Since the discovery in the 1970s that dendritic abnormalities in cortical pyramidal neurons are the most consistent pathologic correlate of mental retardation, research has focused on how dendritic alterations are related to reduced intellectual ability. Due in part to obvious ethical problems and in part to the lack of fruitful methods to study neuronal circuitry in the human cortex, there is little data about the microanatomical contribution to mental retardation. The recent identification of the genetic bases of some mental retardation associated alterations, coupled with the technology to create transgenic animal models and the introduction of powerful sophisticated tools in the field of microanatomy, has led to a growth in the studies of the alterations of pyramidal cell morphology in these disorders. Studies of individuals with Down syndrome, the most frequent genetic disorder leading to mental retardation, allow the analysis of the relationships between cognition, genotype and brain microanatomy. In Down syndrome the crucial question is to define the mechanisms by which an excess of normal gene products, in interaction with the environment, directs and constrains neural maturation, and how this abnormal development translates into cognition and behaviour. In the present article we discuss mainly Down syndrome-associated dendritic abnormalities and plasticity and the role of animal models in these studies. We believe that through the further development of such approaches, the study of the microanatomical substrates of mental retardation will contribute significantly to our understanding of the mechanisms underlying human brain disorders associated with mental retardation.


Asunto(s)
Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Dendritas/efectos de los fármacos , Dendritas/patología , Síndrome de Down/patología , Síndrome de Down/fisiopatología , Plasticidad Neuronal/efectos de los fármacos , Animales , Trastornos del Conocimiento/tratamiento farmacológico , Modelos Animales de Enfermedad , Síndrome de Down/tratamiento farmacológico , Síndrome de Down/genética , Terapia Genética/métodos , Humanos , Ratones , Neuronas/efectos de los fármacos , Neuronas/patología
17.
ScientificWorldJournal ; 6: 1911-22, 2006 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-17205196

RESUMEN

DYRKs (dual-specificity tyrosine-regulated kinases) are an emerging family of evolutionarily conserved dual-specificity kinases that play key roles in cell proliferation, survival, and development. The research in the last years suggests a relevant conserved function during neuronal development, related to proliferation and/or differentiation for DYRK1A. It is expressed in neural progenitor cells and has been proposed to participate in the signaling mechanisms that regulate dendrite differentiation. In Drosophila, disruption of the homolog minibrain gene results in flies with reduced neuroblast proliferation, decreased numbers of central brain neurons, and learning/memory deficits. Knockout DYRK1A mice are embryonic lethal, and heterozygotes show decreased viability and region-specific reductions in brain size. In humans, DYRK1A has been proposed to be involved in the neurodevelopmental alterations associated with Down syndrome. The large number of protein interaction and putative substrates described for DYRK1A suggest multiple pathways and functions to be involved in its developmental function. This review focuses on the functional role that DYRK1A plays in brain development.


Asunto(s)
Encéfalo/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Animales , Encéfalo/embriología , Encéfalo/enzimología , Dosificación de Gen , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , Neuronas/citología , Neuronas/enzimología , Neuronas/metabolismo , Organogénesis/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Quinasas DyrK
18.
Transl Psychiatry ; 6: e718, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26784972

RESUMEN

Different neurodegenerative disorders often show similar lesions, such as the presence of amyloid plaques, TAU-neurotangles and synuclein inclusions. The genetically inherited forms are rare, so we wondered whether shared epigenetic aberrations, such as those affecting DNA methylation, might also exist. The studied samples were gray matter samples from the prefrontal cortex of control and neurodegenerative disease-associated cases. We performed the DNA methylation analyses of Alzheimer's disease, dementia with Lewy bodies, Parkinson's disease and Alzheimer-like neurodegenerative profile associated with Down's syndrome samples. The DNA methylation landscapes obtained show that neurodegenerative diseases share similar aberrant CpG methylation shifts targeting a defined gene set. Our findings suggest that neurodegenerative disorders might have similar pathogenetic mechanisms that subsequently evolve into different clinical entities. The identified aberrant DNA methylation changes can be used as biomarkers of the disorders and as potential new targets for the development of new therapies.


Asunto(s)
Metilación de ADN/fisiología , Epigenómica , Enfermedades Neurodegenerativas/metabolismo , Corteza Prefrontal/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Análisis de Matrices Tisulares
19.
Neuropharmacology ; 34(4): 367-75, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7566467

RESUMEN

The mechanism by which 9-amino-1,2,3,4-tetrahydroacridine (THA) inhibits beta-adrenoceptor linked cyclic AMP formation and its possible relationship with the cholinergic system were studied. In addition, the effect of THA on alpha 1-adrenoceptor coupled transduction systems was also investigated. THA was not able to influence the concentration-response curve for forskolin indicating that it is not acting on the catalytic subunit of the adenylate cyclase complex. On the other hand a cholinergic component seems to participate in the action of THA on beta-adrenoceptor stimulated adenylate cyclase activity since the blockade of muscarinic receptors with atropine (10 microM) partially prevented the reduction in cyclic AMP formation attained by THA in the hippocampus, in isoprenaline-stimulated conditions. This effect is not reproducible by another potent anticholinesterase physostigmine. Moreover, THA at concentrations up to micromolar did not affect alpha 1-adrenoceptor stimulated cyclic AMP formation or phosphoinositide hydrolysis. In conclusion, the neuropharmacological profile of THA is not to be restricted to the cholinergic system and its effectiveness in improving age-associated cognitive deterioration may involve an action on the beta-adrenoceptor coupled signal transduction system. Moreover, the action of THA on the beta-adrenergic and cholinergic systems in the brain could be relevant to the amelioration of cognitive deterioration and could lead to the development of new therapeutic strategies.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Hipocampo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tacrina/farmacología , Animales , Metabolismo Basal , Corteza Cerebral/metabolismo , Colforsina/farmacología , AMP Cíclico/metabolismo , Hipocampo/metabolismo , Hidrólisis , Técnicas In Vitro , Masculino , Fosfatidilinositoles/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Receptores Adrenérgicos alfa 1/metabolismo , Estimulación Química
20.
Behav Neurosci ; 118(4): 815-21, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15301607

RESUMEN

DYRK1A is a protein kinase proposed to be involved in neurogenesis. Gene-targeting disruption of Dyrk1A in mice leads to decreased body and brain size, with no severe disturbance of behavior. In this study, the authors focused on the motor profile of Dyrk1A(+/-) mice. These mice presented impairment of neuromotor development with decreased activity, suggesting a physiological role of Dyrk1A in the maturation of the neuromotor system. In the adult, a marked hypoactivity and alteration of specific motor parameters were detected. These results are in agreement with the significant expression of Dyrk1A in structures related to motor function and support a role of Dyrk1A in the control of motor function.


Asunto(s)
Haplotipos , Actividad Motora/fisiología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Tirosina Quinasas/deficiencia , Desempeño Psicomotor/fisiología , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Conducta Exploratoria/fisiología , Fuerza de la Mano/fisiología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/fisiología , Tiempo de Reacción/fisiología , Caminata/fisiología , Quinasas DyrK
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA