Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Hum Mol Genet ; 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39137370

RESUMEN

Mutations in methyl-CpG binding protein 2 (MeCP2), such as the T158M, P152R, R294X, and R306C mutations, are responsible for most Rett syndrome (RTT) cases. These mutations often result in altered protein expression that appears to correlate with changes in the nuclear size; however, the molecular details of these observations are poorly understood. Using a C2C12 cellular system expressing human MeCP2-E1 isoform as well as mouse models expressing these mutations, we show that T158M and P152R result in a decrease in MeCP2 protein, whereas R306C has a milder variation, and R294X resulted in an overall 2.5 to 3 fold increase. We also explored the potential involvement of the MeCP2 PEST domains in the proteasome-mediated regulation of MeCP2. Finally, we used the R294X mutant to gain further insight into the controversial competition between MeCP2 and histone H1 in the chromatin context. Interestingly, in R294X, MeCP2 E1 and E2 isoforms were differently affected, where the E1 isoform contributes to much of the overall protein increase observed, while E2 decreases by half. The modes of MeCP2 regulation, thus, appear to be differently regulated in the two isoforms.

2.
PLoS Genet ; 19(2): e1010659, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36848371

RESUMEN

Copy number variations (CNVs) in the Neurexin 1 (NRXN1) gene, which encodes a presynaptic protein involved in neurotransmitter release, are some of the most frequently observed single-gene variants associated with autism spectrum disorder (ASD). To address the functional contribution of NRXN1 CNVs to behavioral phenotypes relevant to ASD, we carried out systematic behavioral phenotyping of an allelic series of Nrxn1 mouse models: one carrying promoter and exon 1 deletion abolishing Nrxn1α transcription, one carrying exon 9 deletion disrupting Nrxn1α protein translation, and one carrying an intronic deletion with no observable effect on Nrxn1α expression. We found that homozygous loss of Nrxn1α resulted in enhanced aggression in males, reduced affiliative social behaviors in females, and significantly altered circadian activities in both sexes. Heterozygous or homozygous loss of Nrxn1α affected the preference for social novelty in male mice, and notably, enhanced repetitive motor skills and motor coordination in both sexes. In contrast, mice bearing an intronic deletion of Nrxn1 did not display alterations in any of the behaviors assessed. These findings demonstrate the importance of Nrxn1α gene dosage in regulating social, circadian, and motor functions, and the variables of sex and genomic positioning of CNVs in the expression of autism-related phenotypes. Importantly, mice with heterozygous loss of Nrxn1, as found in numerous autistic individuals, show an elevated propensity to manifest autism-related phenotypes, supporting the use of models with this genomic architecture to study ASD etiology and assess additional genetic variants associated with autism.


Asunto(s)
Trastorno del Espectro Autista , Proteínas de Unión al Calcio , Moléculas de Adhesión de Célula Nerviosa , Animales , Femenino , Masculino , Ratones , Trastorno del Espectro Autista/genética , Variaciones en el Número de Copia de ADN/genética , Fenotipo , Conducta Social , Moléculas de Adhesión de Célula Nerviosa/genética , Proteínas de Unión al Calcio/genética
3.
Mol Cell ; 68(5): 1006-1015.e7, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29220646

RESUMEN

Massively parallel single-cell RNA sequencing can precisely resolve cellular diversity in a high-throughput manner at low cost, but unbiased isolation of intact single cells from complex tissues such as adult mammalian brains is challenging. Here, we integrate sucrose-gradient-assisted purification of nuclei with droplet microfluidics to develop a highly scalable single-nucleus RNA-seq approach (sNucDrop-seq), which is free of enzymatic dissociation and nucleus sorting. By profiling ∼18,000 nuclei isolated from cortical tissues of adult mice, we demonstrate that sNucDrop-seq not only accurately reveals neuronal and non-neuronal subtype composition with high sensitivity but also enables in-depth analysis of transient transcriptional states driven by neuronal activity, at single-cell resolution, in vivo.


Asunto(s)
Núcleo Celular/metabolismo , Corteza Cerebral/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Neuronas/metabolismo , ARN/genética , Convulsiones/genética , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Transcripción Genética , Animales , Núcleo Celular/patología , Centrifugación por Gradiente de Densidad , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Células Madre Embrionarias Humanas/metabolismo , Humanos , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas Analíticas Microfluídicas , Células 3T3 NIH , Inhibición Neural , Neuronas/patología , Pentilenotetrazol , ARN/metabolismo , Convulsiones/metabolismo , Convulsiones/patología , Convulsiones/fisiopatología , Transmisión Sináptica , Transfección
4.
Neurobiol Dis ; 148: 105176, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33197557

RESUMEN

CDKL5 deficiency disorder (CDD) is an infantile, epileptic encephalopathy presenting with early-onset seizures, intellectual disability, motor impairment, and autistic features. The disorder has been linked to mutations in the X-linked CDKL5, and mouse models of the disease recapitulate several aspects of CDD symptomology, including learning and memory impairments, motor deficits, and autistic-like features. Although early-onset epilepsy is one of the hallmark features of CDD, evidence of spontaneous seizure activity has only recently been described in Cdkl5-deficient heterozygous female mice, but the etiology, prevalence, and sex-specificity of this phenotype remain unknown. Here, we report the first observation of disturbance-associated seizure-like events in heterozygous female mice across two independent mouse models of CDD: Cdkl5 knockout mice and CDKL5 R59X knock-in mice. We find that both the prevalence and severity of this phenotype increase with aging, with a median onset around 28 weeks of age. Similar seizure-like events are not observed in hemizygous knockout male or homozygous knockout female littermates, suggesting that X-linked cellular mosaicism is a driving factor underlying these seizure-like events. Together, these findings not only contribute to our understanding of the effects of CDKL5 loss on seizure susceptibility, but also document a novel, pre-clinical phenotype for future therapeutic investigation.


Asunto(s)
Síndromes Epilépticos/fisiopatología , Mosaicismo , Proteínas Serina-Treonina Quinasas/genética , Convulsiones/fisiopatología , Espasmos Infantiles/fisiopatología , Factores de Edad , Animales , Modelos Animales de Enfermedad , Síndromes Epilépticos/genética , Femenino , Técnicas de Sustitución del Gen , Heterocigoto , Ratones , Ratones Noqueados , Fenotipo , Convulsiones/genética , Factores Sexuales , Espasmos Infantiles/genética
5.
Genome Res ; 28(7): 933-942, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29848492

RESUMEN

Genetic variants associated with autism spectrum disorders (ASDs) are enriched in genes encoding synaptic proteins and chromatin regulators. Although the role of synaptic proteins in ASDs is widely studied, the mechanism by which chromatin regulators contribute to ASD risk remains poorly understood. Upon profiling and analyzing the transcriptional and epigenomic features of genes expressed in the cortex, we uncovered a unique set of long genes that contain broad enhancer-like chromatin domains (BELDs) spanning across their entire gene bodies. Analyses of these BELD genes show that they are highly transcribed with frequent RNA polymerase II (Pol II) initiation and low Pol II pausing, and they exhibit frequent chromatin-chromatin interactions within their gene bodies. These BELD features are conserved from rodents to humans, are enriched in genes involved in synaptic function, and appear post-natally concomitant with synapse development. Importantly, we find that BELD genes are highly implicated in neurodevelopmental disorders, particularly ASDs, and that their expression is preferentially down-regulated in individuals with idiopathic autism. Finally, we find that the transcription of BELD genes is particularly sensitive to alternations in ASD-associated chromatin regulators. These findings suggest that the epigenomic regulation of BELD genes is important for post-natal cortical development and lend support to a model by which mutations in chromatin regulators causally contribute to ASDs by preferentially impairing BELD gene transcription.


Asunto(s)
Trastorno del Espectro Autista/genética , Cromatina/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Trastorno Autístico/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Neurogénesis/genética , ARN Polimerasa II/genética , Transcripción Genética/genética
6.
Neurobiol Learn Mem ; 165: 106961, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30447288

RESUMEN

Mutations in the methyl-CpG binding protein 2 (MECP2) gene cause Rett syndrome (RTT), a progressive X-linked neurological disorder characterized by loss of developmental milestones, intellectual disability and breathing abnormality. Despite being a monogenic disorder, the pathogenic mechanisms by which mutations in MeCP2 impair neuronal function and underlie the RTT symptoms have been challenging to elucidate. The seemingly simple genetic root and the availability of genetic data from RTT patients have led to the generation and characterization of a series of mouse models recapitulating RTT-associated genetic mutations. This review focuses on the studies of RTT mouse models and describe newly obtained pathogenic insights from these studies. We also highlight the potential of studying pathophysiology using genetics-based modeling approaches in rodents and suggest a future direction to tackle the pathophysiology of intellectual disability with known or complex genetic causes.


Asunto(s)
Síndrome de Rett/genética , Animales , Modelos Animales de Enfermedad , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Ratones , Síndrome de Rett/fisiopatología
7.
Neurobiol Learn Mem ; 165: 106780, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-29307548

RESUMEN

Behavioral neuroscience research incorporates the identical high level of meticulous methodologies and exacting attention to detail as all other scientific disciplines. To achieve maximal rigor and reproducibility of findings, well-trained investigators employ a variety of established best practices. Here we explicate some of the requirements for rigorous experimental design and accurate data analysis in conducting mouse and rat behavioral tests. Novel object recognition is used as an example of a cognitive assay which has been conducted successfully with a range of methods, all based on common principles of appropriate procedures, controls, and statistics. Directors of Rodent Core facilities within Intellectual and Developmental Disabilities Research Centers contribute key aspects of their own novel object recognition protocols, offering insights into essential similarities and less-critical differences. Literature cited in this review article will lead the interested reader to source papers that provide step-by-step protocols which illustrate optimized methods for many standard rodent behavioral assays. Adhering to best practices in behavioral neuroscience will enhance the value of animal models for the multiple goals of understanding biological mechanisms, evaluating consequences of genetic mutations, and discovering efficacious therapeutics.


Asunto(s)
Investigación Conductal/métodos , Ratones/psicología , Ratas/psicología , Animales , Investigación Conductal/normas , Reproducibilidad de los Resultados , Proyectos de Investigación
8.
J Neurosci ; 37(45): 10773-10782, 2017 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-29118205

RESUMEN

Intellectual disability (ID) is a prevailing neurodevelopmental condition associated with impaired cognitive and adaptive behaviors. Many chromatin-modifying enzymes and other epigenetic regulators have been genetically associated with ID disorders (IDDs). Here we review how alterations in the function of histone modifiers, chromatin remodelers, and methyl-DNA binding proteins contribute to neurodevelopmental defects and altered brain plasticity. We also discuss how progress in human genetics has led to the generation of mouse models that unveil the molecular etiology of ID, and outline the direction in which this field is moving to identify therapeutic strategies for IDDs. Importantly, because the chromatin regulators linked to IDDs often target common downstream genes and cellular processes, the impact of research in individual syndromes goes well beyond each syndrome and can also contribute to the understanding and therapy of other IDDs. Furthermore, the investigation of these disorders helps us to understand the role of chromatin regulators in brain development, plasticity, and gene expression, thereby answering fundamental questions in neurobiology.


Asunto(s)
Epigénesis Genética/genética , Discapacidad Intelectual/etiología , Discapacidad Intelectual/genética , Epigenómica , Humanos
9.
J Neurosci ; 37(31): 7420-7437, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28674172

RESUMEN

Cyclin-dependent kinase-like 5 (CDKL5) deficiency is a neurodevelopmental disorder characterized by epileptic seizures, severe intellectual disability, and autistic features. Mice lacking CDKL5 display multiple behavioral abnormalities reminiscent of the disorder, but the cellular origins of these phenotypes remain unclear. Here, we find that ablating CDKL5 expression specifically from forebrain glutamatergic neurons impairs hippocampal-dependent memory in male conditional knock-out mice. Hippocampal pyramidal neurons lacking CDKL5 show decreased dendritic complexity but a trend toward increased spine density. This morphological change is accompanied by an increase in the frequency of spontaneous miniature EPSCs and interestingly, miniature IPSCs. Using voltage-sensitive dye imaging to interrogate the evoked response of the CA1 microcircuit, we find that CA1 pyramidal neurons lacking CDKL5 show hyperexcitability in their dendritic domain that is constrained by elevated inhibition in a spatially and temporally distinct manner. These results suggest a novel role for CDKL5 in the regulation of synaptic function and uncover an intriguing microcircuit mechanism underlying impaired learning and memory.SIGNIFICANCE STATEMENT Cyclin-dependent kinase-like 5 (CDKL5) deficiency is a severe neurodevelopmental disorder caused by mutations in the CDKL5 gene. Although Cdkl5 constitutive knock-out mice have recapitulated key aspects of human symptomatology, the cellular origins of CDKL5 deficiency-related phenotypes are unknown. Here, using conditional knock-out mice, we show that hippocampal-dependent learning and memory deficits in CDKL5 deficiency have origins in glutamatergic neurons of the forebrain and that loss of CDKL5 results in the enhancement of synaptic transmission and disruptions in neural circuit dynamics in a spatially and temporally specific manner. Our findings demonstrate that CDKL5 is an important regulator of synaptic function in glutamatergic neurons and serves a critical role in learning and memory.


Asunto(s)
Glutamatos/metabolismo , Hipocampo/fisiopatología , Trastornos de la Memoria/fisiopatología , Red Nerviosa/fisiopatología , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Masculino , Memoria , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética
10.
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
11.
J Neurodev Disord ; 16(1): 5, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38424476

RESUMEN

X-linked genetic causes of intellectual disability (ID) account for a substantial proportion of cases and remain poorly understood, in part due to the heterogeneous expression of X-linked genes in females. This is because most genes on the X chromosome are subject to random X chromosome inactivation (XCI) during early embryonic development, which results in a mosaic pattern of gene expression for a given X-linked mutant allele. This mosaic expression produces substantial complexity, especially when attempting to study the already complicated neural circuits that underly behavior, thus impeding the understanding of disease-related pathophysiology and the development of therapeutics. Here, we review a few selected X-linked forms of ID that predominantly affect heterozygous females and the current obstacles for developing effective therapies for such disorders. We also propose a genetic strategy to overcome the complexity presented by mosaicism in heterozygous females and highlight specific tools for studying synaptic and circuit mechanisms, many of which could be shared across multiple forms of intellectual disability.


Asunto(s)
Discapacidad Intelectual , Femenino , Humanos , Embarazo , Cromosomas Humanos X , Genes Ligados a X/genética , Discapacidad Intelectual/genética , Mosaicismo , Inactivación del Cromosoma X/genética
12.
Neurobiol Dis ; 58: 3-12, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23659895

RESUMEN

Rett Syndrome (RTT), a progressive neurological disorder characterized by developmental regression and loss of motor and language skills, is caused by mutations in the X-linked gene encoding methyl-CpG binding protein 2 (MECP2). Neurostructural phenotypes including decreased neuronal size, dendritic complexity, and spine density have been reported in postmortem RTT brain tissue and in Mecp2 animal models. How these changes in neuronal morphology are related to RTT-like phenotype and MeCP2 function, and the extent to which restoration of neuronal morphology can be used as a cellular readout in therapeutic studies, however, remain unclear. Here, we systematically examined neuronal morphology in vivo across three Mecp2 mouse models representing Mecp2 loss-of-function, partial loss-of-function, and gain-of-function mutations, at developmental time points corresponding to early- and late-symptomatic RTT-like behavioral phenotypes. We found that in Mecp2 loss-of-function mouse models, dendritic complexity is reduced in a mild, age-dependent, and brain region-specific manner, whereas soma size is reduced consistently throughout development. Neither phenotype, however, is altered in Mecp2 gain-of-function mice. Our results suggest that, in the cell types we examined, the use of dendritic morphology as a cellular readout of RTT phenotype and therapeutic efficacy should be cautioned, as it is intrinsically variable. In contrast, soma size may be a robust and reliable marker for evaluation of MeCP2 function in Mecp2 loss-of-function studies.


Asunto(s)
Envejecimiento/patología , Encéfalo/patología , Proteína 2 de Unión a Metil-CpG/genética , Mutación/genética , Neuronas/patología , Síndrome de Rett , Análisis de Varianza , Animales , Dendritas/genética , Dendritas/patología , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Neuronas/citología , Síndrome de Rett/genética , Síndrome de Rett/patología , Síndrome de Rett/fisiopatología
13.
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
14.
Nat Biotechnol ; 2023 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-37640946

RESUMEN

Oxidative modification of 5-methylcytosine (5mC) by ten-eleven translocation (TET) DNA dioxygenases generates 5-hydroxymethylcytosine (5hmC), the most abundant form of oxidized 5mC. Existing single-cell bisulfite sequencing methods cannot resolve 5mC and 5hmC, leaving the cell-type-specific regulatory mechanisms of TET and 5hmC largely unknown. Here, we present joint single-nucleus (hydroxy)methylcytosine sequencing (Joint-snhmC-seq), a scalable and quantitative approach that simultaneously profiles 5hmC and true 5mC in single cells by harnessing differential deaminase activity of APOBEC3A toward 5mC and chemically protected 5hmC. Joint-snhmC-seq profiling of single nuclei from mouse brains reveals an unprecedented level of epigenetic heterogeneity of both 5hmC and true 5mC at single-cell resolution. We show that cell-type-specific profiles of 5hmC or true 5mC improve multimodal single-cell data integration, enable accurate identification of neuronal subtypes and uncover context-specific regulatory effects on cell-type-specific genes by TET enzymes.

15.
Nat Commun ; 13(1): 55, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013139

RESUMEN

Although the synaptic alterations associated with the stress-related mood disorder major depression has been well-documented, the underlying transcriptional mechanisms remain poorly understood. Here, we perform complementary bulk nuclei- and single-nucleus transcriptome profiling and map locus-specific chromatin interactions in mouse neocortex to identify the cell type-specific transcriptional changes associated with stress-induced behavioral maladaptation. We find that cortical excitatory neurons, layer 2/3 neurons in particular, are vulnerable to chronic stress and acquire signatures of gene transcription and chromatin structure associated with reduced neuronal activity and expression of Yin Yang 1 (YY1). Selective ablation of YY1 in cortical excitatory neurons enhances stress sensitivity in both male and female mice and alters the expression of stress-associated genes following an abbreviated stress exposure. These findings demonstrate how chronic stress impacts transcription in cortical excitatory neurons and identify YY1 as a regulator of stress-induced maladaptive behavior in mice.


Asunto(s)
Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/metabolismo , Animales , Conducta Animal , Cromatina/metabolismo , Epigenómica , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Fisiológico
16.
EMBO Rep ; 10(12): 1327-33, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19820693

RESUMEN

Mutations in the methyl-CpG-binding protein 2 (MeCP2) are associated with Rett syndrome and other neurological disorders. MeCP2 represses transcription mainly by recruiting various co-repressor complexes. Recently, MeCP2 phosphorylation at Ser 80, Ser 229 and Ser 421 was shown to occur in the brain and modulate MeCP2 silencing activities. However, the kinases directly responsible for this are largely unknown. Here, we identify the homeodomain-interacting protein kinase 2 (HIPK2) as a kinase that binds MeCP2 and phosphorylates it at Ser 80 in vitro and in vivo. HIPK2 modulates cell proliferation and apoptosis, and the neurological defects of Hipk2-null mice indicate its role in proper brain functions. We show that MeCP2 cooperates with HIPK2 in induction of apoptosis and that Ser 80 phosphorylation is required together with the DNA binding of MeCP2. These data are, to our knowledge, the first that describe a kinase associating with MeCP2, causing its specific phosphorylation in vivo and, furthermore, they reinforce the role of MeCP2 in regulating cell growth.


Asunto(s)
Apoptosis/genética , Proteínas Portadoras/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Proteína 2 de Unión a Metil-CpG/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Sustitución de Aminoácidos/fisiología , Animales , Apoptosis/efectos de los fármacos , Proteínas Portadoras/antagonistas & inhibidores , Células Cultivadas , ADN/metabolismo , Embrión de Mamíferos , Células HeLa , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Células 3T3 NIH , Fosforilación/efectos de los fármacos , Fosforilación/genética , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Serina/genética , Serina/metabolismo
17.
PLoS Genet ; 4(9): e1000179, 2008 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-18773074

RESUMEN

The levels of methyl-CpG-binding protein 2 (MeCP2) are critical for normal post-natal development and function of the nervous system. Loss of function of MeCP2, a transcriptional regulator involved in chromatin remodeling, causes classic Rett syndrome (RTT) as well as other related conditions characterized by autism, learning disabilities, or mental retardation. Increased dosage of MeCP2 also leads to clinically similar neurological disorders and mental retardation. To identify molecular mechanisms capable of compensating for altered MeCP2 levels, we generated transgenic Drosophila overexpressing human MeCP2. We find that MeCP2 associates with chromatin and is phosphorylated at serine 423 in Drosophila, as is found in mammals. MeCP2 overexpression leads to anatomical (i.e., disorganized eyes, ectopic wing veins) and behavioral (i.e., motor dysfunction) abnormalities. We used a candidate gene approach to identify genes that are able to compensate for abnormal phenotypes caused by MeCP2 increased activity. These genetic modifiers include other chromatin remodeling genes (Additional sex combs, corto, osa, Sex combs on midleg, and trithorax), the kinase tricornered, the UBE3A target pebble, and Drosophila homologues of the MeCP2 physical interactors Sin3a, REST, and N-CoR. These findings demonstrate that anatomical and behavioral phenotypes caused by MeCP2 activity can be ameliorated by altering other factors that might be more amenable to manipulation than MeCP2 itself.


Asunto(s)
Drosophila/embriología , Drosophila/genética , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Animales , Animales Modificados Genéticamente , Cromatina/metabolismo , Drosophila/metabolismo , Humanos , Microscopía Electrónica de Rastreo , Fenotipo , Fosforilación , Serina/genética , Serina/metabolismo
18.
J Clin Invest ; 131(20)2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34651584

RESUMEN

CDKL5 deficiency disorder (CDD) is an early onset, neurodevelopmental syndrome associated with pathogenic variants in the X-linked gene encoding cyclin-dependent kinase-like 5 (CDKL5). CDKL5 has been implicated in neuronal synapse maturation, yet its postdevelopmental necessity and the reversibility of CDD-associated impairments remain unknown. We temporally manipulated endogenous Cdkl5 expression in male mice and found that postdevelopmental loss of CDKL5 disrupts numerous behavioral domains, hippocampal circuit communication, and dendritic spine morphology, demonstrating an indispensable role for CDKL5 in the adult brain. Accordingly, restoration of Cdkl5 after the early stages of brain development using a conditional rescue mouse model ameliorated CDD-related behavioral impairments and aberrant NMDA receptor signaling. These findings highlight the requirement of CDKL5 beyond early development, underscore the potential for disease reversal in CDD, and suggest that a broad therapeutic time window exists for potential treatment of CDD-related deficits.


Asunto(s)
Síndromes Epilépticos/psicología , Proteínas Serina-Treonina Quinasas/fisiología , Espasmos Infantiles/psicología , Animales , Dendritas/patología , Potenciales Evocados/fisiología , Ratones , Ratones Endogámicos C57BL , Receptores de N-Metil-D-Aspartato/fisiología
19.
Neuron ; 52(2): 255-69, 2006 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-17046689

RESUMEN

Mutations or duplications in MECP2 cause Rett and Rett-like syndromes, neurodevelopmental disorders characterized by mental retardation, motor dysfunction, and autistic behaviors. MeCP2 is expressed in many mammalian tissues and functions as a global repressor of transcription; however, the molecular mechanisms by which MeCP2 dysfunction leads to the neural-specific phenotypes of RTT remain poorly understood. Here, we show that neuronal activity and subsequent calcium influx trigger the de novo phosphorylation of MeCP2 at serine 421 (S421) by a CaMKII-dependent mechanism. MeCP2 S421 phosphorylation is induced selectively in the brain in response to physiological stimuli. Significantly, we find that S421 phosphorylation controls the ability of MeCP2 to regulate dendritic patterning, spine morphogenesis, and the activity-dependent induction of Bdnf transcription. These findings suggest that, by triggering MeCP2 phosphorylation, neuronal activity regulates a program of gene expression that mediates nervous system maturation and that disruption of this process in individuals with mutations in MeCP2 may underlie the neural-specific pathology of RTT.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Diferenciación Celular/fisiología , Espinas Dendríticas/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Animales , Encéfalo/citología , Factor Neurotrófico Derivado del Encéfalo/genética , Señalización del Calcio/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Espinas Dendríticas/ultraestructura , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteína 2 de Unión a Metil-CpG/genética , Vías Nerviosas/citología , Vías Nerviosas/crecimiento & desarrollo , Vías Nerviosas/metabolismo , Plasticidad Neuronal/fisiología , Técnicas de Cultivo de Órganos , Especificidad de Órganos/fisiología , Fosforilación , Ratas , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/fisiopatología , Serina/metabolismo , Transmisión Sináptica/fisiología
20.
J Neurosci ; 29(40): 12440-8, 2009 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-19812320

RESUMEN

Rett syndrome, an autism spectrum disorder with prominent motor and cognitive features, results from mutations in the gene for methyl-CpG-binding protein 2 (MeCP2). Here, to identify cortical circuit abnormalities that are specifically associated with MeCP2 deficiency, we used glutamate uncaging and laser scanning photostimulation to survey intracortical networks in mouse brain slices containing motor-frontal cortex. We used in utero transfection of short hairpin RNA constructs to knock down MeCP2 expression in a sparsely distributed subset of layer (L) 2/3 pyramidal neurons in wild-type mice, and compared input maps recorded from transfected-untransfected pairs of neighboring neurons. The effect of MeCP2 deficiency on local excitatory input pathways was severe, with an average reduction in excitatory synaptic input from middle cortical layers (L3/5A) of >30% compared with MeCP2-replete controls. MeCP2 deficiency primarily affected the strength, rather than the topography, of excitatory intracortical pathways. Inhibitory synaptic inputs and intrinsic eletrophysiological properties were unaffected in the MeCP2-knockdown neurons. These studies indicate that MeCP2 deficiency in individual postsynaptic cortical pyramidal neurons is sufficient to induce a pathological synaptic defect in excitatory intracortical circuits.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Corteza Motora/metabolismo , Células Piramidales/anomalías , Células Piramidales/metabolismo , Sinapsis/metabolismo , Animales , Mapeo Encefálico , Femenino , Ratones , Corteza Motora/citología , Corteza Motora/embriología , Técnicas de Placa-Clamp , Estimulación Luminosa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA