Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros












Intervalo de año de publicación
1.
Am J Hum Genet ; 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-39013458

RESUMEN

The shift to a genotype-first approach in genetic diagnostics has revolutionized our understanding of neurodevelopmental disorders, expanding both their molecular and phenotypic spectra. Kleefstra syndrome (KLEFS1) is caused by EHMT1 haploinsufficiency and exhibits broad clinical manifestations. EHMT1 encodes euchromatic histone methyltransferase-1-a pivotal component of the epigenetic machinery. We have recruited 209 individuals with a rare EHMT1 variant and performed comprehensive molecular in silico and in vitro testing alongside DNA methylation (DNAm) signature analysis for the identified variants. We (re)classified the variants as likely pathogenic/pathogenic (molecularly confirming Kleefstra syndrome) in 191 individuals. We provide an updated and broader clinical and molecular spectrum of Kleefstra syndrome, including individuals with normal intelligence and familial occurrence. Analysis of the EHMT1 variants reveals a broad range of molecular effects and their associated phenotypes, including distinct genotype-phenotype associations. Notably, we showed that disruption of the "reader" function of the ankyrin repeat domain by a protein altering variant (PAV) results in a KLEFS1-specific DNAm signature and milder phenotype, while disruption of only "writer" methyltransferase activity of the SET domain does not result in KLEFS1 DNAm signature or typical KLEFS1 phenotype. Similarly, N-terminal truncating variants result in a mild phenotype without the DNAm signature. We demonstrate how comprehensive variant analysis can provide insights into pathogenesis of the disorder and DNAm signature. In summary, this study presents a comprehensive overview of KLEFS1 and EHMT1, revealing its broader spectrum and deepening our understanding of its molecular mechanisms, thereby informing accurate variant interpretation, counseling, and clinical management.

2.
Cell Rep ; 43(3): 113883, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38430517

RESUMEN

Phosphomannomutase 2-congenital disorder of glycosylation (PMM2-CDG) is a rare inborn error of metabolism caused by deficiency of the PMM2 enzyme, which leads to impaired protein glycosylation. While the disorder presents with primarily neurological symptoms, there is limited knowledge about the specific brain-related changes caused by PMM2 deficiency. Here, we demonstrate aberrant neural activity in 2D neuronal networks from PMM2-CDG individuals. Utilizing multi-omics datasets from 3D human cortical organoids (hCOs) derived from PMM2-CDG individuals, we identify widespread decreases in protein glycosylation, highlighting impaired glycosylation as a key pathological feature of PMM2-CDG, as well as impaired mitochondrial structure and abnormal glucose metabolism in PMM2-deficient hCOs, indicating disturbances in energy metabolism. Correlation between PMM2 enzymatic activity in hCOs and symptom severity suggests that the level of PMM2 enzyme function directly influences neurological manifestations. These findings enhance our understanding of specific brain-related perturbations associated with PMM2-CDG, offering insights into the underlying mechanisms and potential directions for therapeutic interventions.


Asunto(s)
Trastornos Congénitos de Glicosilación , Fosfotransferasas (Fosfomutasas)/deficiencia , Humanos , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/metabolismo , Glicosilación
3.
Brain ; 145(12): 4349-4367, 2022 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-36074904

RESUMEN

Parkinson's disease is a common incurable neurodegenerative disease. The identification of genetic variants via genome-wide association studies has considerably advanced our understanding of the Parkinson's disease genetic risk. Understanding the functional significance of the risk loci is now a critical step towards translating these genetic advances into an enhanced biological understanding of the disease. Impaired mitophagy is a key causative pathway in familial Parkinson's disease, but its relevance to idiopathic Parkinson's disease is unclear. We used a mitophagy screening assay to evaluate the functional significance of risk genes identified through genome-wide association studies. We identified two new regulators of PINK1-dependent mitophagy initiation, KAT8 and KANSL1, previously shown to modulate lysine acetylation. These findings suggest PINK1-mitophagy is a contributing factor to idiopathic Parkinson's disease. KANSL1 is located on chromosome 17q21 where the risk associated gene has long been considered to be MAPT. While our data do not exclude a possible association between the MAPT gene and Parkinson's disease, they provide strong evidence that KANSL1 plays a crucial role in the disease. Finally, these results enrich our understanding of physiological events regulating mitophagy and establish a novel pathway for drug targeting in neurodegeneration.


Asunto(s)
Mitofagia , Enfermedad de Parkinson , Humanos , Estudio de Asociación del Genoma Completo , Mitofagia/fisiología , Enfermedades Neurodegenerativas , Enfermedad de Parkinson/metabolismo , Proteínas Quinasas/genética , Proteínas tau/genética
4.
Cell Rep ; 39(8): 110857, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35613587

RESUMEN

Protocadherin-19 (PCDH19) is a synaptic cell-adhesion molecule encoded by X-linked PCDH19, a gene linked with epilepsy. Here, we report a synapse-to-nucleus signaling pathway through which PCDH19 bridges neuronal activity with gene expression. In particular, we describe the NMDA receptor (NMDAR)-dependent proteolytic cleavage of PCDH19, which leads to the generation of a PCDH19 C-terminal fragment (CTF) able to enter the nucleus. We demonstrate that PCDH19 CTF associates with chromatin and with the chromatin remodeler lysine-specific demethylase 1 (LSD1) and regulates expression of immediate-early genes (IEGs). Our results are consistent with a model whereby PCDH19 favors maintenance of neuronal homeostasis via negative feedback regulation of IEG expression and provide a key to interpreting PCDH19-related hyperexcitability.


Asunto(s)
Cadherinas , Epilepsia , Genes Inmediatos-Precoces , Protocadherinas , Cadherinas/genética , Cadherinas/metabolismo , Cromatina/genética , Cromatina/metabolismo , Epilepsia/genética , Epilepsia/metabolismo , Regulación de la Expresión Génica , Humanos , Protocadherinas/genética , Protocadherinas/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal
6.
Stem Cell Reports ; 16(9): 2118-2127, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34358451

RESUMEN

Human neurons engineered from induced pluripotent stem cells (iPSCs) through neurogenin 2 (NGN2) overexpression are widely used to study neuronal differentiation mechanisms and to model neurological diseases. However, the differentiation paths and heterogeneity of emerged neurons have not been fully explored. Here, we used single-cell transcriptomics to dissect the cell states that emerge during NGN2 overexpression across a time course from pluripotency to neuron functional maturation. We find a substantial molecular heterogeneity in the neuron types generated, with at least two populations that express genes associated with neurons of the peripheral nervous system. Neuron heterogeneity is observed across multiple iPSC clones and lines from different individuals. We find that neuron fate acquisition is sensitive to NGN2 expression level and the duration of NGN2-forced expression. Our data reveal that NGN2 dosage can regulate neuron fate acquisition, and that NGN2-iN heterogeneity can confound results that are sensitive to neuron type.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular , Células Cultivadas , Biología Computacional/métodos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Proteínas del Tejido Nervioso/metabolismo , RNA-Seq , Transcriptoma
7.
Neurobiol Learn Mem ; 173: 107265, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32531423

RESUMEN

Kleefstra syndrome is a disorder caused by a mutation in the EHMT1 gene characterized in humans by general developmental delay, mild to severe intellectual disability and autism. Here, we characterized cumulative memory in the Ehmt1+/- mouse model using the Object Space Task. We combined conventional behavioral analysis with automated analysis by deep-learning networks, a session-based computational learning model, and a trial-based classifier. Ehmt1+/- mice showed more anxiety-like features and generally explored objects less, but the difference decreased over time. Interestingly, when analyzing memory-specific exploration, Ehmt1+/- show increased expression of cumulative memory, but a deficit in a more simple, control memory condition. Using our automatic classifier to differentiate between genotypes, we found that cumulative memory features are better suited for classification than general exploration differences. Thus, detailed behavioral classification with the Object Space Task produced a more detailed behavioral phenotype of the Ehmt1+/- mouse model.


Asunto(s)
Conducta Animal/fisiología , Anomalías Craneofaciales/fisiopatología , Conducta Exploratoria/fisiología , Cardiopatías Congénitas/fisiopatología , Discapacidad Intelectual/fisiopatología , Memoria/fisiología , Animales , Deleción Cromosómica , Cromosomas Humanos Par 9/genética , Anomalías Craneofaciales/genética , Aprendizaje Profundo , Modelos Animales de Enfermedad , Cardiopatías Congénitas/genética , N-Metiltransferasa de Histona-Lisina/genética , Discapacidad Intelectual/genética , Masculino , Ratones
8.
Genet Med ; 21(8): 1790-1796, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30607023

RESUMEN

PURPOSE: Identifying and characterizing novel causes of autosomal recessive intellectual disability based on systematic clinical and genetic evaluation, followed by functional experiments. METHODS: Clinical examinations, genome-wide positional mapping, and sequencing were followed by quantitative polymerase chain reaction and western blot of the protein SVBP and its interaction partners. We then knocked down the gene in rat primary hippocampal neurons and evaluated the consequences on synapses. RESULTS: We identified a founder, homozygous stop-gain variant in SVBP (c.82C>T; p.[Gln28*]) in four affected individuals from two independent families with intellectual disability, microcephaly, ataxia, and muscular hypotonia. SVBP encodes a small chaperone protein that transports and stabilizes two angiogenesis regulators, VASH1 and VASH2. The altered protein is unstable and nonfunctional since transfected HeLa cells with mutant SVBP did not reveal evidence for immunoreactive SVBP protein fragments and cotransfection with VASH1 showed a severe reduction of VASH1 in medium and cell lysate. Knocking down Svbp in rat primary hippocampal neurons led to a significant decrease in the number of excitatory synapses. CONCLUSION: SVBP is not only involved in angiogenesis, but also has vital functions in the central nervous system. Biallelic loss-of-function variants in SVBP lead to intellectual disability.


Asunto(s)
Proteínas Portadoras/genética , Genes Recesivos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Discapacidad Intelectual/genética , Proteínas Angiogénicas , Animales , Ataxia/epidemiología , Ataxia/genética , Ataxia/patología , Proteínas de Ciclo Celular , Femenino , Genotipo , Células HeLa , Homocigoto , Humanos , Discapacidad Intelectual/epidemiología , Discapacidad Intelectual/patología , Mutación con Pérdida de Función/genética , Masculino , Microcefalia/epidemiología , Microcefalia/genética , Microcefalia/patología , Hipotonía Muscular/epidemiología , Hipotonía Muscular/genética , Hipotonía Muscular/patología , Linaje , Ratas
10.
Nucleic Acids Res ; 46(10): 4950-4965, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29554304

RESUMEN

Kleefstra syndrome, a disease with intellectual disability, autism spectrum disorders and other developmental defects is caused in humans by haploinsufficiency of EHMT1. Although EHMT1 and its paralog EHMT2 were shown to be histone methyltransferases responsible for deposition of the di-methylated H3K9 (H3K9me2), the exact nature of epigenetic dysfunctions in Kleefstra syndrome remains unknown. Here, we found that the epigenome of Ehmt1+/- adult mouse brain displays a marked increase of H3K9me2/3 which correlates with impaired expression of protocadherins, master regulators of neuronal diversity. Increased H3K9me3 was present already at birth, indicating that aberrant methylation patterns are established during embryogenesis. Interestingly, we found that Ehmt2+/- mice do not present neither the marked increase of H3K9me2/3 nor the cognitive deficits found in Ehmt1+/- mice, indicating an evolutionary diversification of functions. Our finding of increased H3K9me3 in Ehmt1+/- mice is the first one supporting the notion that EHMT1 can quench the deposition of tri-methylation by other Histone methyltransferases, ultimately leading to impaired neurocognitive functioning. Our insights into the epigenetic pathophysiology of Kleefstra syndrome may offer guidance for future developments of therapeutic strategies for this disease.


Asunto(s)
Cadherinas/genética , Disfunción Cognitiva/metabolismo , Anomalías Craneofaciales/metabolismo , Cardiopatías Congénitas/metabolismo , Histonas/metabolismo , Discapacidad Intelectual/metabolismo , Animales , Animales Recién Nacidos , Cadherinas/metabolismo , Deleción Cromosómica , Cromosomas Humanos Par 9/metabolismo , Disfunción Cognitiva/genética , Anomalías Craneofaciales/psicología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Cardiopatías Congénitas/psicología , Hipocampo/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Discapacidad Intelectual/psicología , Lisina/metabolismo , Masculino , Metilación , Ratones Noqueados
11.
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
12.
Sci Rep ; 7: 40284, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28071689

RESUMEN

Heterozygous mutations or deletions of the human Euchromatin Histone Methyltransferase 1 (EHMT1) gene are the main causes of Kleefstra syndrome, a neurodevelopmental disorder that is characterized by impaired memory, autistic features and mostly severe intellectual disability. Previously, Ehmt1+/- heterozygous knockout mice were found to exhibit cranial abnormalities and decreased sociability, phenotypes similar to those observed in Kleefstra syndrome patients. In addition, Ehmt1+/- knockout mice were impaired at fear extinction and novel- and spatial object recognition. In this study, Ehmt1+/- and wild-type mice were tested on several cognitive tests in a touchscreen-equipped operant chamber to further investigate the nature of learning and memory changes. Performance of Ehmt1+/- mice in the Visual Discrimination &Reversal learning, object-location Paired-Associates learning- and Extinction learning tasks was found to be unimpaired. Remarkably, Ehmt1+/- mice showed enhanced performance on the Location Discrimination test of pattern separation. In line with improved Location Discrimination ability, an increase in BrdU-labelled cells in the subgranular zone of the dentate gyrus was observed. In conclusion, reduced levels of EHMT1 protein in Ehmt1+/- mice does not result in general learning deficits in a touchscreen-based battery, but leads to increased adult cell proliferation in the hippocampus and enhanced pattern separation ability.


Asunto(s)
Trastornos del Conocimiento/genética , Anomalías Craneofaciales/genética , Cardiopatías Congénitas/genética , N-Metiltransferasa de Histona-Lisina/genética , Discapacidad Intelectual/genética , Aprendizaje/fisiología , Animales , Proliferación Celular/genética , Deleción Cromosómica , Cromosomas Humanos Par 9/genética , Trastornos del Conocimiento/fisiopatología , Anomalías Craneofaciales/fisiopatología , Haploinsuficiencia/genética , Haploinsuficiencia/fisiología , Cardiopatías Congénitas/fisiopatología , Hipocampo/fisiopatología , Humanos , Discapacidad Intelectual/fisiopatología , Memoria/fisiología , Ratones , Ratones Noqueados , Mutación
13.
Sci Rep ; 6: 34240, 2016 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-27687783

RESUMEN

Schizophrenia is a complex disorder that affects cognitive function and has been linked, both in patients and animal models, to dysfunction of the GABAergic system. However, the pathophysiological consequences of this dysfunction are not well understood. Here, we examined the GABAergic system in an animal model displaying schizophrenia-relevant features, the apomorphine-susceptible (APO-SUS) rat and its phenotypic counterpart, the apomorphine-unsusceptible (APO-UNSUS) rat at postnatal day 20-22. We found changes in the expression of the GABA-synthesizing enzyme GAD67 specifically in the prelimbic- but not the infralimbic region of the medial prefrontal cortex (mPFC), indicative of reduced inhibitory function in this region in APO-SUS rats. While we did not observe changes in basal synaptic transmission onto LII/III pyramidal cells in the mPFC of APO-SUS compared to APO-UNSUS rats, we report reduced paired-pulse ratios at longer inter-stimulus intervals. The GABAB receptor antagonist CGP 55845 abolished this reduction, indicating that the decreased paired-pulse ratio was caused by increased GABAB signaling. Consistently, we find an increased expression of the GABAB1 receptor subunit in APO-SUS rats. Our data provide physiological evidence for increased presynaptic GABAB signaling in the mPFC of APO-SUS rats, further supporting an important role for the GABAergic system in the pathophysiology of schizophrenia.

14.
Neurobiol Learn Mem ; 124: 88-96, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26143996

RESUMEN

Histone post-translational modifications are key epigenetic processes controlling the regulation of gene transcription. In recent years it has become apparent that chromatin modifications contribute to cognition through the modulation of gene expression required for the expression and consolidation of memories. In this review, we focus on the role of histone methylation in the nervous system. Histone methylation is involved in a number of cognitive disturbances, such as intellectual disability, cocaine addiction and age-related cognitive decline. We provide an overview of the dynamic changes in methylation of histone lysine residues during learning and memory. With a special focus on H3K9 histone methyltransferases GLP and G9a, we summarize the effects of deficiencies in writer and eraser enzymes on neuronal plasticity and cognition.


Asunto(s)
Cromatina/genética , Trastornos del Conocimiento/genética , Cognición/fisiología , Epigénesis Genética , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Aprendizaje/fisiología , Animales , Encéfalo/metabolismo , Ensamble y Desensamble de Cromatina , Humanos , Memoria/fisiología , Metilación , Plasticidad Neuronal/genética , Neuronas/metabolismo , Trastornos Relacionados con Sustancias/genética
15.
J Neurosci ; 35(19): 7349-64, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25972165

RESUMEN

The protein NOS1AP/CAPON mediates signaling from a protein complex of NMDA receptor, PSD95 and nNOS. The only stroke trial for neuroprotectants that showed benefit to patients targeted this ternary complex. NOS1AP/nNOS interaction regulates small GTPases, iron transport, p38MAPK-linked excitotoxicity, and anxiety. Moreover, the nos1ap gene is linked to disorders from schizophrenia, post-traumatic stress disorder, and autism to cardiovascular disorders and breast cancer. Understanding protein interactions required for NOS1AP function, therefore, has broad implications for numerous diseases. Here we show that the interaction of NOS1AP with nNOS differs radically from the classical PDZ docking assumed to be responsible. The NOS1AP PDZ motif does not bind nNOS as measured by multiple methods. In contrast, full-length NOS1AP forms an unusually stable interaction with nNOS. We mapped the discrepancy between full-length and C-terminal PDZ motif to a novel internal region we call the ExF motif. The C-terminal PDZ motif, although neither sufficient nor necessary for binding, nevertheless promotes the stability of the complex. It therefore potentially affects signal transduction and suggests that functional interaction of nNOS with NOS1AP might be targetable at two distinct sites. We demonstrate that excitotoxic pathways can be regulated, in cortical neuron and organotypic hippocampal slice cultures from rat, either by the previously described PDZ ligand TAT-GESV or by the ExF motif-bearing region of NOS1AP, even when lacking the critical PDZ residues as long as the ExF motif is intact and not mutated. This previously unrecognized heterodivalent interaction of nNOS with NOS1AP may therefore provide distinct opportunities for pharmacological intervention in NOS1AP-dependent signaling and excitotoxicity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Animales Recién Nacidos , Células COS , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Chlorocebus aethiops , Agonistas de Aminoácidos Excitadores/farmacología , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Técnicas In Vitro , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Mutación/genética , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I/genética , Técnicas de Cultivo de Órganos , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/fisiología , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
J Cell Sci ; 127(Pt 12): 2782-92, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24762814

RESUMEN

The anti-apoptotic B-cell lymphoma-2 (Bcl-2) protein not only counteracts apoptosis at the mitochondria by scaffolding pro-apoptotic Bcl-2-family members, but also acts at the endoplasmic reticulum, thereby controlling intracellular Ca(2+) dynamics. Bcl-2 inhibits Ca(2+) release by targeting the inositol 1,4,5-trisphosphate receptor (IP3R). Sequence analysis has revealed that the Bcl-2-binding site on the IP3R displays strong similarity with a conserved sequence present in all three ryanodine receptor (RyR) isoforms. We now report that Bcl-2 co-immunoprecipitated with RyRs in ectopic expression systems and in native rat hippocampi, indicating that endogenous RyR-Bcl-2 complexes exist. Purified RyR domains containing the putative Bcl-2-binding site bound full-length Bcl-2 in pulldown experiments and interacted with the BH4 domain of Bcl-2 in surface plasmon resonance (SPR) experiments, suggesting a direct interaction. Exogenous expression of full-length Bcl-2 or electroporation loading of the BH4 domain of Bcl-2 dampened RyR-mediated Ca(2+) release in HEK293 cell models. Finally, introducing the BH4-domain peptide into hippocampal neurons through a patch pipette decreased RyR-mediated Ca(2+) release. In conclusion, this study identifies Bcl-2 as a new inhibitor of RyR-based intracellular Ca(2+)-release channels.


Asunto(s)
Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Secuencia de Aminoácidos , Animales , Canales de Calcio/metabolismo , Señalización del Calcio , Células HEK293 , Hipocampo/citología , Humanos , Ratones , Visón , Datos de Secuencia Molecular , Neuronas/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Conejos , Ratas , Canal Liberador de Calcio Receptor de Rianodina/química
17.
Hum Mol Genet ; 22(5): 852-66, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23175442

RESUMEN

Euchromatin histone methyltransferase 1 (EHMT1) is a highly conserved protein that catalyzes mono- and dimethylation of histone H3 lysine 9, thereby epigenetically regulating transcription. Kleefstra syndrome (KS), is caused by haploinsufficiency of the EHMT1 gene, and is an example of an emerging group of intellectual disability (ID) disorders caused by genes encoding epigenetic regulators of neuronal gene activity. Little is known about the mechanisms underlying this disorder, prompting us to study the Euchromatin histone methyltransferase 1 heterozygous knockout (Ehmt1(+/-)) mice as a model for KS. In agreement with the cognitive disturbances observed in patients with KS, we detected deficits in fear extinction learning and both novel and spatial object recognition in Ehmt1(+/-) mice. These learning and memory deficits were associated with a significant reduction in dendritic arborization and the number of mature spines in hippocampal CA1 pyramidal neurons of Ehmt1(+/-) mice. In-depth analysis of the electrophysiological properties of CA3-CA1 synapses revealed no differences in basal synaptic transmission or theta-burst induced long-term potentiation (LTP). However, paired-pulse facilitation (PPF) was significantly increased in Ehmt1(+/-) neurons, pointing to a potential deficiency in presynaptic neurotransmitter release. Accordingly, a reduction in the frequency of miniature excitatory post-synaptic currents (mEPSCs) was observed in Ehmt1(+/-) neurons. These data demonstrate that Ehmt1 haploinsufficiency in mice leads to learning deficits and synaptic dysfunction, providing a possible mechanism for the ID phenotype in patients with KS.


Asunto(s)
Anomalías Craneofaciales/genética , Cardiopatías Congénitas/genética , N-Metiltransferasa de Histona-Lisina/genética , Discapacidad Intelectual/genética , Aprendizaje , Animales , Deleción Cromosómica , Cromosomas Humanos Par 9/genética , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Discapacidad Intelectual/fisiopatología , Ratones , Ratones Noqueados , Células Piramidales/patología , Sinapsis/patología
18.
Am J Hum Genet ; 91(1): 73-82, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22726846

RESUMEN

Intellectual disability (ID) disorders are genetically and phenotypically highly heterogeneous and present a major challenge in clinical genetics and medicine. Although many genes involved in ID have been identified, the etiology is unknown in most affected individuals. Moreover, the function of most genes associated with ID remains poorly characterized. Evidence is accumulating that the control of gene transcription through epigenetic modification of chromatin structure in neurons has an important role in cognitive processes and in the etiology of ID. However, our understanding of the key molecular players and mechanisms in this process is highly fragmentary. Here, we identify a chromatin-modification module that underlies a recognizable form of ID, the Kleefstra syndrome phenotypic spectrum (KSS). In a cohort of KSS individuals without mutations in EHMT1 (the only gene known to be disrupted in KSS until now), we identified de novo mutations in four genes, MBD5, MLL3, SMARCB1, and NR1I3, all of which encode epigenetic regulators. Using Drosophila, we demonstrate that MBD5, MLL3, and NR1I3 cooperate with EHMT1, whereas SMARCB1 is known to directly interact with MLL3. We propose a highly conserved epigenetic network that underlies cognition in health and disease. This network should allow the design of strategies to treat the growing group of ID pathologies that are caused by epigenetic defects.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/genética , Discapacidad Intelectual/genética , Animales , Cromatina , Proteínas Cromosómicas no Histona/genética , Receptor de Androstano Constitutivo , Proteínas de Unión al ADN/genética , Drosophila , Epigénesis Genética , Femenino , Humanos , Recién Nacido , Masculino , Mutación , Proteína SMARCB1 , Síndrome , Factores de Transcripción/genética
19.
Curr Biol ; 19(13): 1133-9, 2009 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-19481455

RESUMEN

Neurons transmit information at chemical synapses by releasing neurotransmitters that are stored in synaptic vesicles (SVs) at the presynaptic site. After release, these vesicles need to be efficiently retrieved in order to maintain synaptic transmission. In concurrence, malfunctions in SV recycling have been associated with cognitive disorders. Oligophrenin-1 (OPHN1) encodes a Rho-GTPase-activating protein (Rho-GAP) whose loss of function causes X-linked mental retardation. OPHN1 is highly expressed in the brain and present both pre- and postsynaptically in neurons. Previous studies report that postsynaptic OPHN1 is important for dendritic spine morphogenesis, but its function at the presynaptic site remains largely unexplored. Here, we present evidence that reduced or defective OPHN1 signaling impairs SV cycling at hippocampal synapses. In particular, we show that OPHN1 knockdown affects the kinetic efficiency of endocytosis. We further demonstrate that OPHN1 forms a complex with endophilin A1, a protein implicated in membrane curvature generation during SV endocytosis and, importantly, that OPHN1's interaction with endophilin A1 and its Rho-GAP activity are important for its function in SV endocytosis. Our findings suggest that defects in efficient SV retrieval may contribute to the pathogenesis of OPHN1-linked cognitive impairment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Citoesqueleto/metabolismo , Endocitosis/fisiología , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Nucleares/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas del Citoesqueleto/genética , Colorantes Fluorescentes/metabolismo , Proteínas Activadoras de GTPasa/genética , Técnicas de Silenciamiento del Gen , Hipocampo/citología , Hipocampo/metabolismo , Ratones , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/genética , Interferencia de ARN , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología
20.
Methods Enzymol ; 439: 255-66, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18374170

RESUMEN

Mutations in regulators and effectors of the Rho GTPases underlie various forms of mental retardation (MR). Among them, oligophrenin-1 (OPHN1), which encodes a Rho-GTPase activating protein, was one of the first Rho-linked MR genes identified. Upon characterization of OPHN1 in hippocampal brain slices, we obtained evidence for the requirement of OPHN1 in dendritic spine morphogenesis and neuronal function of CA1 pyramidal neurons. Organotypic hippocampal brain slice cultures are commonly used as a model system to investigate the morphology and synaptic function of neurons, mainly because they allow for the long-term examination of neurons in a preparation where the gross cellular architecture of the hippocampus is retained. In addition, maintenance of the trisynaptic circuitry in hippocampal slices enables the study of synaptic connections. Today, a multitude of gene transfer methods for postmitotic neurons in brain slices are available to easily manipulate and scrutinize the involvement of signaling molecules, such as Rho GTPases, in specific cellular processes in this system. This chapter covers techniques detailing the preparation and culturing of organotypic hippocampal brain slices, as well as the production and injection of lentivirus into brain slices.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Citoesqueleto/fisiología , Proteínas Activadoras de GTPasa/fisiología , Proteínas Nucleares/fisiología , Animales , Proteínas del Citoesqueleto/aislamiento & purificación , Proteínas Activadoras de GTPasa/aislamiento & purificación , Técnicas de Transferencia de Gen , Vectores Genéticos , Hipocampo/fisiología , Hipocampo/virología , Humanos , Lentivirus , Proteínas Nucleares/aislamiento & purificación , Técnicas de Cultivo de Órganos/métodos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...