Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Proc Natl Acad Sci U S A ; 109(52): 21516-21, 2012 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-23236174

RESUMEN

Mutations in the X-linked cyclin-dependent kinase-like 5 (CDKL5) gene have been identified in neurodevelopmental disorders including atypical Rett syndrome (RTT), autism spectrum disorders (ASDs), and early infantile epileptic encephalopathy. The biological function of CDKL5 and its role in the etiology of these disorders, however, remain unclear. Here we report the development of a unique knockout mouse model of CDKL5-related disorders and demonstrate that mice lacking CDKL5 show autistic-like deficits in social interaction, as well as impairments in motor control and fear memory. Neurophysiological recordings reveal alterations in event-related potentials (ERPs) similar to those observed in RTT and ASDs. Moreover, kinome profiling uncovers disruption of multiple signal transduction pathways, including the AKT-mammalian target of rapamycin (mTOR) cascade, upon Cdkl5 loss-of-function. These data demonstrate that CDKL5 regulates signal transduction pathways and mediates autistic-like phenotypes and together establish a causal role for Cdkl5 loss-of-function in neurodevelopmental disorders.


Asunto(s)
Trastorno Autístico/enzimología , Trastorno Autístico/fisiopatología , Potenciales Evocados/fisiología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteoma/metabolismo , Animales , Ansiedad/complicaciones , Ansiedad/enzimología , Ansiedad/fisiopatología , Trastorno Autístico/complicaciones , Conducta Animal , Electroencefalografía , Hipercinesia/complicaciones , Hipercinesia/enzimología , Hipercinesia/fisiopatología , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Convulsiones/complicaciones , Convulsiones/fisiopatología , Transducción de Señal , Conducta Social , Serina-Treonina Quinasas TOR/metabolismo
2.
Neurobiol Dis ; 59: 257-66, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23948639

RESUMEN

Rett syndrome (RTT) is a neurodevelopmental disorder characterized by developmental regression beginning 6-18months after birth, followed by a lifetime of intellectual disability, stereotyped behaviors, and motor deficits. RTT is caused by mutations in the gene encoding MeCP2, a methyl-CpG binding protein believed to modulate gene transcription. Gene expression studies of individual brain regions have reported that Mecp2 loss-of-function leads to both activation and repression of its gene targets in mice. Conditional deletion of MeCP2 from different brain regions has revealed unique insights into the role of these structures in mediating particular RTT-like phenotypes. However, the function of MeCP2 in the striatum, a major brain region involved in motor control and executive cognitive functions, has yet to be studied. Here, we characterized the gene expression changes in the striatum of Mecp2 mutant mice. We found a number of differentially expressed genes in the striatum of both constitutive Mecp2-null mice and mice lacking MeCP2 only from forebrain GABAergic neurons. These changes only occurred when MeCP2 expression levels had reached mature levels and RTT-like symptoms were manifest, supporting a role for MeCP2 in maintaining proper brain function. Many of the gene expression changes identified in the striatum have not previously been shown to change in the hypothalamus or cerebellum. Bioinformatic analysis of differentially expressed genes in striatum as well as hypothalamus and cerebellum revealed that loss of MeCP2 does not affect the global landscape of gene expression. Additionally, we uncovered a number of differentially expressed genes in the liver of Mecp2-null mice suggesting an important role for MeCP2 in non-neuronal tissues. Collectively, our data suggest that the differential expression of genes following loss of MeCP2 occurs in a tissue- or cell-type specific manner and thus MeCP2 function should be understood in a cellular context.


Asunto(s)
Cuerpo Estriado/crecimiento & desarrollo , Cuerpo Estriado/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Proteína 2 de Unión a Metil-CpG/deficiencia , Animales , Animales Recién Nacidos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ontología de Genes , Redes Reguladoras de Genes/genética , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo
3.
J Neurosci ; 30(8): 2935-50, 2010 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-20181591

RESUMEN

Dopaminergic projections to the striatum, crucial for the correct functioning of this brain region in adulthood, are known to be established early in development, but their role is currently uncharacterized. We demonstrate here that dopamine, by activating D(1)- and/or D(2)-dopamine receptors, decreases the number of functional GABAergic synapses formed between the embryonic precursors of the medium spiny neurons, the principal output neurons of the striatum, with associated changes in spontaneous synaptic activity. Activation of these receptors reduces the size of postsynaptic GABA(A) receptor clusters and their overall cell-surface expression, without affecting the total number of clusters or the size or number of GABAergic nerve terminals. These changes result from an increased internalization of GABA(A) receptors, and are mediated by distinct signaling pathways converging at the level of GABA(A) receptors to cause a transient PP2A/PP1-dependent dephosphorylation. Thus, tonic D(1)- and D(2)-receptor activity limits the extent of collateral inhibitory synaptogenesis between medium spiny neurons, revealing a novel role of dopamine in controlling the development of intrinsic striatal microcircuits.


Asunto(s)
Cuerpo Estriado/embriología , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Inhibición Neural/fisiología , Neurogénesis/fisiología , Sinapsis/metabolismo , Animales , Diferenciación Celular/fisiología , Cuerpo Estriado/citología , Vías Nerviosas/citología , Vías Nerviosas/embriología , Vías Nerviosas/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Agregación de Receptores/fisiología , Receptores Dopaminérgicos/metabolismo , Receptores de GABA-A/metabolismo , Transmisión Sináptica/fisiología , Factores de Tiempo , Ácido gamma-Aminobutírico/metabolismo
4.
J Neurochem ; 107(4): 964-75, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19006818

RESUMEN

The proliferation and differentiation of neural progenitor (NP) cells can be regulated by neurotransmitters including GABA and dopamine. The present study aimed to examine how these two neurotransmitter systems interact to affect post-natal hippocampal NP cell proliferation in vitro. Mouse hippocampal NP cells express functional GABAA receptors, which upon activation led to an increase in intracellular calcium levels via the opening of L-type calcium channels. Activation of these GABAA receptors also caused a significant decrease in proliferation; an effect that required the entry of calcium through L-type calcium channels. Furthermore, while activation of D1-like dopamine receptors had no effect on proliferation, it abrogated the suppressive effects of GABAA receptor activation on proliferation. The effects of D1-like dopamine receptors are associated with a decrease in the ability of GABAA receptors to increase intracellular calcium levels, and a reduction in the surface expression of GABAA receptors. In this way, D1-like dopamine receptor activation can increase the proliferation of NP cells by preventing GABAA receptor-mediated inhibition of proliferation. These results suggest that, in conditions where NP cell proliferation is under the tonic suppression of GABA, agonists which act through D1-like dopamine receptors may increase the proliferation of neural progenitors.


Asunto(s)
Proliferación Celular , Hipocampo/citología , Neuronas/metabolismo , Receptores de Dopamina D1/fisiología , Receptores de GABA-A/fisiología , Células Madre/fisiología , Animales , Animales Recién Nacidos , Bromodesoxiuridina/metabolismo , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Recuento de Células/métodos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Dopaminérgicos/farmacología , Interacciones Farmacológicas , Ensayo de Inmunoadsorción Enzimática/métodos , GABAérgicos/farmacología , Expresión Génica/efectos de los fármacos , Ratones , Muscimol/farmacología , Neuronas/efectos de los fármacos , Nifedipino/farmacología , Piridazinas/farmacología , Receptores de GABA-A/genética , Células Madre/efectos de los fármacos
5.
J Clin Invest ; 127(5): 1889-1904, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28394263

RESUMEN

Mutations in the X-linked gene encoding methyl-CpG-binding protein 2 (MeCP2) cause Rett syndrome (RTT), a neurological disorder affecting cognitive development, respiration, and motor function. Genetic restoration of MeCP2 expression reverses RTT-like phenotypes in mice, highlighting the need to search for therapeutic approaches. Here, we have developed knockin mice recapitulating the most common RTT-associated missense mutation, MeCP2 T158M. We found that the T158M mutation impaired MECP2 binding to methylated DNA and destabilized MeCP2 protein in an age-dependent manner, leading to the development of RTT-like phenotypes in these mice. Genetic elevation of MeCP2 T158M expression ameliorated multiple RTT-like features, including motor dysfunction and breathing irregularities, in both male and female mice. These improvements were accompanied by increased binding of MeCP2 T158M to DNA. Further, we found that the ubiquitin/proteasome pathway was responsible for MeCP2 T158M degradation and that proteasome inhibition increased MeCP2 T158M levels. Together, these findings demonstrate that increasing MeCP2 T158M protein expression is sufficient to mitigate RTT-like phenotypes and support the targeting of MeCP2 T158M expression or stability as an alternative therapeutic approach.


Asunto(s)
Regulación de la Expresión Génica , Proteína 2 de Unión a Metil-CpG , Mutación Missense , Proteolisis , Síndrome de Rett , Sustitución de Aminoácidos , Animales , Humanos , Proteína 2 de Unión a Metil-CpG/biosíntesis , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Transgénicos , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patología , Ubiquitina/genética , Ubiquitina/metabolismo
6.
Nat Med ; 23(10): 1203-1214, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28920956

RESUMEN

Mutations in MECP2 cause Rett syndrome (RTT), an X-linked neurological disorder characterized by regressive loss of neurodevelopmental milestones and acquired psychomotor deficits. However, the cellular heterogeneity of the brain impedes an understanding of how MECP2 mutations contribute to RTT. Here we developed a Cre-inducible method for cell-type-specific biotin tagging of MeCP2 in mice. Combining this approach with an allelic series of knock-in mice carrying frequent RTT-associated mutations (encoding T158M and R106W) enabled the selective profiling of RTT-associated nuclear transcriptomes in excitatory and inhibitory cortical neurons. We found that most gene-expression changes were largely specific to each RTT-associated mutation and cell type. Lowly expressed cell-type-enriched genes were preferentially disrupted by MeCP2 mutations, with upregulated and downregulated genes reflecting distinct functional categories. Subcellular RNA analysis in MeCP2-mutant neurons further revealed reductions in the nascent transcription of long genes and uncovered widespread post-transcriptional compensation at the cellular level. Finally, we overcame X-linked cellular mosaicism in female RTT models and identified distinct gene-expression changes between neighboring wild-type and mutant neurons, providing contextual insights into RTT etiology that support personalized therapeutic interventions.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/genética , Neuronas/metabolismo , Síndrome de Rett/genética , Transcriptoma/genética , Alelos , Animales , Biotina , Biotinilación , Corteza Cerebral/citología , Femenino , Perfilación de la Expresión Génica , Técnicas de Sustitución del Gen , Genotipo , Ratones , Mosaicismo , Mutación , Mutación Missense , Fenotipo
7.
Front Cell Neurosci ; 9: 290, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26300728

RESUMEN

Basal ganglia play an essential role in motor coordination and cognitive functions. The GABAergic medium spiny neurons (MSNs) account for ~95% of all the neurons in this brain region. Central to the normal functioning of MSNs is integration of synaptic activity arriving from the glutamatergic corticostriatal and thalamostriatal afferents, with synaptic inhibition mediated by local interneurons and MSN axon collaterals. In this study we have investigated how the specific types of GABAergic synapses between the MSNs develop over time, and how the activity of GABAA receptors (GABAARs) influences this development. Isolated embryonic (E17) MSNs form a homogenous population in vitro and display spontaneous synaptic activity and functional properties similar to their in vivo counterparts. In dual whole-cell recordings of synaptically connected pairs of MSNs, action potential (AP)-activated synaptic events were detected between 7 and 14 days in vitro (DIV), which coincided with the shift in GABAAR operation from depolarization to hyperpolarization, as detected indirectly by intracellular calcium imaging. In parallel, the predominant subtypes of inhibitory synapses, which innervate dendrites of MSNs and contain GABAAR α1 or α2 subunits, underwent distinct changes in the size of postsynaptic clusters, with α1 becoming smaller and α2 larger over time, while both the percentage and the size of mixed α1/α2-postsynaptic clusters were increased. When activity of GABAARs was under chronic blockade between 4-7 DIV, the structural properties of these synapses remained unchanged. In contrast, chronic inhibition of GABAARs between 7-14 DIV led to reduction in size of α1- and α1/α2-postsynaptic clusters and a concomitant increase in number and size of α2-postsynaptic clusters. Thus, the main subtypes of GABAergic synapses formed by MSNs are regulated by GABAAR activity, but in opposite directions, and thus appear to be driven by different molecular mechanisms.

8.
Nat Neurosci ; 17(6): 804-6, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24777420

RESUMEN

Dysfunction in sensory information processing is a hallmark of many neurological disorders, including autism spectrum disorders, schizophrenia and Rett syndrome (RTT). Using mouse models of RTT, a monogenic disorder caused by mutations in MECP2, we found that the large-scale loss of MeCP2 from forebrain GABAergic interneurons led to deficits in auditory event-related potentials and seizure manifestation, whereas the restoration of MeCP2 in specific classes of interneurons ameliorated these deficits.


Asunto(s)
Potenciales Evocados Auditivos/fisiología , Interneuronas/patología , Proteína 2 de Unión a Metil-CpG/genética , Prosencéfalo/patología , Síndrome de Rett/patología , Síndrome de Rett/fisiopatología , Animales , Interneuronas/fisiología , Masculino , Proteína 2 de Unión a Metil-CpG/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Prosencéfalo/fisiología , Síndrome de Rett/genética
9.
Front Biol (Beijing) ; 7(5): 428-435, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24999353

RESUMEN

Rett syndrome is an Autism Spectrum Disorder caused by mutations in the gene encoding methyl-CpG binding protein (MeCP2). Following a period of normal development, patients lose learned communication and motor skills, and develop a number of symptoms including motor disturbances, cognitive impairments and often seizures. In this review, we discuss the role of MeCP2 in regulating synaptic function and how synaptic dysfunctions lead to neuronal network impairments and alterations in sensory information processing. We propose that Rett syndrome is a disorder of neural circuits as a result of non-linear accumulated dysfunction of synapses at the level of individual cell populations across multiple neurotransmitter systems and brain regions.

10.
Nat Neurosci ; 15(2): 274-83, 2011 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-22119903

RESUMEN

Mutations in the MECP2 gene cause the autism spectrum disorder Rett syndrome (RTT). One of the most common MeCP2 mutations associated with RTT occurs at threonine 158, converting it to methionine (T158M) or alanine (T158A). To understand the role of T158 mutations in the pathogenesis of RTT, we generated knockin mice that recapitulate the MeCP2 T158A mutation. We found a causal role for T158A mutation in the development of RTT-like phenotypes, including developmental regression, motor dysfunction, and learning and memory deficits. These phenotypes resemble those present in Mecp2 null mice and manifest through a reduction in MeCP2 binding to methylated DNA and a decrease in MeCP2 protein stability. The age-dependent development of event-related neuronal responses was disrupted by MeCP2 mutation, suggesting that impaired neuronal circuitry underlies the pathogenesis of RTT and that assessment of event-related potentials (ERPs) may serve as a biomarker for RTT and treatment evaluation.


Asunto(s)
Proteínas de Unión al ADN/genética , Potenciales Evocados Auditivos del Tronco Encefálico/genética , Proteína 2 de Unión a Metil-CpG/genética , Mutación/genética , Treonina/genética , Estimulación Acústica/métodos , Factores de Edad , Alanina/genética , Animales , Células Cultivadas , Corteza Cerebral/citología , Inmunoprecipitación de Cromatina , Condicionamiento Psicológico/fisiología , Análisis Mutacional de ADN , Electroencefalografía , Embrión de Mamíferos , Conducta Exploratoria/fisiología , Miedo/fisiología , Regulación de la Expresión Génica/genética , Humanos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/genética , Neuronas/fisiología , Análisis Espectral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA