Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Psychiatry ; 28(6): 2382-2397, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36991133

RESUMEN

Angelman syndrome (AS) is a rare genetic neurodevelopmental disorder caused by the maternally inherited loss of function of the UBE3A gene. AS is characterized by a developmental delay, lack of speech, motor dysfunction, epilepsy, autistic features, happy demeanor, and intellectual disability. While the cellular roles of UBE3A are not fully understood, studies suggest that the lack of UBE3A function is associated with elevated levels of reactive oxygen species (ROS). Despite the accumulating evidence emphasizing the importance of ROS during early brain development and its involvement in different neurodevelopmental disorders, up to date, the levels of ROS in AS neural precursor cells (NPCs) and the consequences on AS embryonic neural development have not been elucidated. In this study we show multifaceted mitochondrial aberration in AS brain-derived embryonic NPCs, which exhibit elevated mitochondrial membrane potential (ΔΨm), lower levels of endogenous reduced glutathione, excessive mitochondrial ROS (mROS) levels, and increased apoptosis compared to wild-type (WT) littermates. In addition, we report that glutathione replenishment by glutathione-reduced ethyl ester (GSH-EE) corrects the excessive mROS levels and attenuates the enhanced apoptosis in AS NPCs. Studying the glutathione redox imbalance and mitochondrial abnormalities in embryonic AS NPCs provides an essential insight into the involvement of UBE3A in early neural development, information that can serve as a powerful avenue towards a broader view of AS pathogenesis. Moreover, since mitochondrial dysfunction and elevated ROS levels were associated with other neurodevelopmental disorders, the findings herein suggest some potential shared underlying mechanisms for these disorders as well.


Asunto(s)
Síndrome de Angelman , Células-Madre Neurales , Animales , Síndrome de Angelman/genética , Síndrome de Angelman/patología , Especies Reactivas de Oxígeno , Células-Madre Neurales/patología , Neuronas/patología , Glutatión , Ubiquitina-Proteína Ligasas/genética , Modelos Animales de Enfermedad
2.
Int J Mol Sci ; 24(23)2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-38069188

RESUMEN

Transcranial direct current stimulation (tDCS) is a subthreshold neurostimulation technique known for ameliorating neuropsychiatric conditions. The principal mechanism of tDCS is the differential polarization of subcellular neuronal compartments, particularly the axon terminals that are sensitive to external electrical fields. Yet, the underlying mechanism of tDCS is not fully clear. Here, we hypothesized that direct current stimulation (DCS)-induced modulation of presynaptic calcium channel conductance alters axon terminal dynamics with regard to synaptic vesicle release. To examine the involvement of calcium-channel subtypes in tDCS, we recorded spontaneous excitatory postsynaptic currents (sEPSCs) from cortical layer-V pyramidal neurons under DCS while selectively inhibiting distinct subtypes of voltage-dependent calcium channels. Blocking P/Q or N-type calcium channels occluded the effects of DCS on sEPSCs, demonstrating their critical role in the process of DCS-induced modulation of spontaneous vesicle release. However, inhibiting T-type calcium channels did not occlude DCS-induced modulation of sEPSCs, suggesting that despite being active in the subthreshold range, T-type calcium channels are not involved in the axonal effects of DCS. DCS modulates synaptic facilitation by regulating calcium channels in axon terminals, primarily via controlling P/Q and N-type calcium channels, while T-type calcium channels are not involved in this mechanism.


Asunto(s)
Canales de Calcio Tipo T , Estimulación Transcraneal de Corriente Directa , Terminales Presinápticos/metabolismo , Neuronas/metabolismo , Canales de Calcio Tipo N , Calcio/metabolismo , Transmisión Sináptica
3.
Int J Mol Sci ; 23(16)2022 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-36012404

RESUMEN

Angelman syndrome (AS) is a neurodevelopmental disorder caused by the loss of function of the E3-ligase UBE3A. Despite multiple studies, AS pathophysiology is still obscure and has mostly been explored in rodent models of the disease. In recent years, a growing body of studies has utilized omics datasets in the attempt to focus research regarding the pathophysiology of AS. Here, for the first time, we utilized a multi-omics approach at the epigenomic level and the transcriptome level, for human-derived neurons. Using publicly available datasets for DNA methylation and gene expression, we found genome regions in proximity to gene promoters and intersecting with gene-body regions that were differentially methylated and differentially expressed in AS. We found that overall, the genome in AS postmortem brain tissue was hypo-methylated compared to healthy controls. We also found more upregulated genes than downregulated genes in AS. Many of these dysregulated genes in neurons obtained from AS patients are known to be critical for neuronal development and synaptic functioning. Taken together, our results suggest a list of dysregulated genes that may be involved in AS development and its pathological features. Moreover, these genes might also have a role in neurodevelopmental disorders similar to AS.


Asunto(s)
Síndrome de Angelman , Síndrome de Angelman/metabolismo , Biología Computacional , Metilación de ADN , Humanos , Transcriptoma , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
4.
Neurobiol Dis ; 148: 105180, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33212289

RESUMEN

BACKGROUND: Angelman syndrome (AS) is a genetic neurodevelopmental disorder caused by the loss of function of the UBE3A protein in the brain. In a previous study, we showed that activity-dependent calcium dynamics in hippocampal CA1 pyramidal neurons of AS mice is compromised, and its normalization rescues the hippocampal-dependent deficits. Therefore, we expected that the expression profiles of calcium-related genes would be altered in AS mice hippocampi. METHODS: We analyzed mRNA sequencing data from AS model mice and WT controls in light of the newly published CaGeDB database of calcium-related genes. We validated our results in two independent RNA sequencing datasets from two additional different AS models: first one, a human neuroblastoma cell line where UBE3A expression was knocked down by siRNA, and the second, an iPSC-derived neurons from AS patient and healthy donor control. FINDINGS: We found signatures of dysregulated calcium-related genes in AS mouse model hippocampus. Additionally, we show that these calcium-related genes function as signatures for AS in other human cellular models of AS, thus strengthening our findings. INTERPRETATION: Our findings suggest the downstream implications and significance of the compromised calcium signaling in Angelman syndrome. Moreover, since AS share similar features with other autism spectrum disorders, we believe that these findings entail meaningful data and approach for other neurodevelopmental disorders, especially those with known alterations of calcium signaling. FUNDING: This work was supported by the Angelman Syndrome Foundation and by the Israel Science Foundation, Grant Number 248/20.


Asunto(s)
Síndrome de Angelman/genética , Señalización del Calcio/genética , Calcio/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Síndrome de Angelman/metabolismo , Animales , Línea Celular Tumoral , Biología Computacional , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Células Madre Pluripotentes Inducidas , Masculino , Ratones , Neuroblastoma/metabolismo , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Factores Sexuales , Transcriptoma , Ubiquitina-Proteína Ligasas/genética
5.
Int J Mol Sci ; 22(18)2021 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-34576033

RESUMEN

Angelman-like syndromes are a group of neurodevelopmental disorders that entail clinical presentation similar to Angelman Syndrome (AS). In our previous study, we showed that calcium signaling is disrupted in AS, and we identified calcium-target and calcium-regulating gene signatures that are able to differentiate between AS and their controls in different models. In the herein study, we evaluated these sets of calcium-target and calcium-regulating genes as signatures of AS-like and non-AS-like syndromes. We collected a number of RNA-seq datasets of various AS-like and non-AS-like syndromes and performed Principle Component Analysis (PCA) separately on the two sets of signature genes to visualize the distribution of samples on the PC1-PC2 plane. In addition to the evaluation of calcium signature genes, we performed differential gene expression analyses to identify calcium-related genes dysregulated in each of the studied syndromes. These analyses showed that the calcium-target and calcium-regulating signatures differentiate well between AS-like syndromes and their controls. However, in spite of the fact that many of the non-AS-like syndromes have multiple differentially expressed calcium-related genes, the calcium signatures were not efficient classifiers for non-AS-like neurodevelopmental disorders. These results show that features based on clinical presentation are reflected in signatures derived from bioinformatics analyses and suggest the use of bioinformatics as a tool for classification.


Asunto(s)
Síndrome de Angelman/genética , Señalización del Calcio/genética , Biología Computacional , Síndrome de Angelman/patología , Calcio/metabolismo , Deleción Cromosómica , Regulación de la Expresión Génica/genética , Humanos , Análisis de Componente Principal , RNA-Seq
6.
Int J Mol Sci ; 21(11)2020 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-32532103

RESUMEN

The UBE3A gene encodes the ubiquitin E3-ligase protein, UBE3A, which is implicated in severe neurodevelopmental disorders. Lack of UBE3A expression results in Angelman syndrome, while UBE3A overexpression, due to genomic 15q duplication, results in autism. The cellular roles of UBE3A are not fully understood, yet a growing body of evidence indicates that these disorders involve mitochondrial dysfunction and increased oxidative stress. We utilized bioinformatics approaches to delineate the effects of murine Ube3a deletion on the expression of mitochondrial-related genes and pathways. For this, we generated an mRNA sequencing dataset from mouse embryonic fibroblasts (MEFs) in which both alleles of Ube3a gene were deleted and their wild-type controls. Since oxidative stress and mitochondrial dysregulation might not be exhibited in the resting baseline state, we also activated mitochondrial functioning in the cells of these two genotypes using TNFα application. Transcriptomes of the four groups of MEFs, Ube3a+/+ and Ube3a-/-, with or without the application of TNFα, were analyzed using various bioinformatics tools and machine learning approaches. Our results indicate that Ube3a deletion affects the gene expression profiles of mitochondrial-associated pathways. We further confirmed these results by analyzing other publicly available human transcriptome datasets of Angelman syndrome and 15q duplication syndrome.


Asunto(s)
Síndrome de Angelman/genética , Mitocondrias/metabolismo , Ubiquitina-Proteína Ligasas/genética , Síndrome de Angelman/patología , Animales , Células Cultivadas , Biología Computacional , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Factor de Necrosis Tumoral alfa/farmacología , Ubiquitina-Proteína Ligasas/metabolismo
7.
J Neurosci ; 38(3): 648-658, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29196323

RESUMEN

Protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) is one of four known kinases that respond to cellular stress by deactivating the eukaryotic initiation factor 2 α (eIF2α) or other signal transduction cascades. Recently, both eIF2α and its kinases were found to play a role in normal and pathological brain function. Here, we show that reduction of either the amount or the activity of PERK, specifically in the CA1 region of the hippocampus in young adult male mice, enhances neuronal excitability and improves cognitive function. In addition, this manipulation rescues the age-dependent cellular phenotype of reduced excitability and memory decline. Specifically, the reduction of PERK expression in the CA1 region of the hippocampus of middle-aged male mice using a viral vector rejuvenates hippocampal function and improves hippocampal-dependent learning. These results delineate a mechanism for behavior and neuronal aging and position PERK as a promising therapeutic target for age-dependent brain malfunction.SIGNIFICANCE STATEMENT We found that local reduced protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) expression or activity in the hippocampus enhances neuronal excitability and cognitive function in young normal mice, that old CA1 pyramidal cells have reduced excitability and increased PERK expression that can be rescued by reducing PERK expression in the hippocampus, and that reducing PERK expression in the hippocampus of middle-aged mice enhances hippocampal-dependent learning and memory and restores it to normal performance levels of young mice. These findings uncover an entirely new biological link among PERK, neuronal intrinsic properties, aging, and cognitive function. Moreover, our findings propose a new way to fight mild cognitive impairment and aging-related cognitive deterioration.


Asunto(s)
Envejecimiento/fisiología , Cognición/fisiología , Hipocampo/enzimología , Hipocampo/metabolismo , Memoria/fisiología , eIF-2 Quinasa/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Cognición/efectos de los fármacos , Disfunción Cognitiva/enzimología , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Memoria/efectos de los fármacos , Ratones , Células Piramidales/efectos de los fármacos , Células Piramidales/enzimología
8.
Nature ; 493(7432): 411-5, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23263185

RESUMEN

Autism spectrum disorders (ASDs) are an early onset, heterogeneous group of heritable neuropsychiatric disorders with symptoms that include deficits in social interaction skills, impaired communication abilities, and ritualistic-like repetitive behaviours. One of the hypotheses for a common molecular mechanism underlying ASDs is altered translational control resulting in exaggerated protein synthesis. Genetic variants in chromosome 4q, which contains the EIF4E locus, have been described in patients with autism. Importantly, a rare single nucleotide polymorphism has been identified in autism that is associated with increased promoter activity in the EIF4E gene. Here we show that genetically increasing the levels of eukaryotic translation initiation factor 4E (eIF4E) in mice results in exaggerated cap-dependent translation and aberrant behaviours reminiscent of autism, including repetitive and perseverative behaviours and social interaction deficits. Moreover, these autistic-like behaviours are accompanied by synaptic pathophysiology in the medial prefrontal cortex, striatum and hippocampus. The autistic-like behaviours displayed by the eIF4E-transgenic mice are corrected by intracerebroventricular infusions of the cap-dependent translation inhibitor 4EGI-1. Our findings demonstrate a causal relationship between exaggerated cap-dependent translation, synaptic dysfunction and aberrant behaviours associated with autism.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/fisiopatología , Factor 4E Eucariótico de Iniciación/metabolismo , Biosíntesis de Proteínas , Sinapsis/metabolismo , Sinapsis/patología , Animales , Trastorno Autístico/tratamiento farmacológico , Trastorno Autístico/patología , Conducta Animal/efectos de los fármacos , Dendritas/metabolismo , Dendritas/patología , Factor 4E Eucariótico de Iniciación/genética , Factor 4G Eucariótico de Iniciación/metabolismo , Femenino , Hipocampo/metabolismo , Hidrazonas , Infusiones Intraventriculares , Masculino , Ratones , Ratones Transgénicos , Neostriado/metabolismo , Plasticidad Neuronal , Nitrocompuestos/administración & dosificación , Nitrocompuestos/farmacología , Nitrocompuestos/uso terapéutico , Corteza Prefrontal/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/genética , Caperuzas de ARN/metabolismo , Tiazoles/administración & dosificación , Tiazoles/farmacología , Tiazoles/uso terapéutico
9.
Cereb Cortex ; 28(8): 2786-2794, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28655149

RESUMEN

Understanding which cellular compartments are influenced during neuromodulation underpins any rational effort to explain and optimize outcomes. Axon terminals have long been speculated to be sensitive to polarization, but experimentally informed models for CNS stimulation are lacking. We conducted simultaneous intracellular recording from the neuron soma and axon terminal (blebs) during extracellular stimulation with weak sustained (DC) uniform electric fields in mouse cortical slices. Use of weak direct current stimulation (DCS) allowed isolation and quantification of changes in axon terminal biophysics, relevant to both suprathreshold (e.g., deep brain stimulation, spinal cord stimulation, and transcranial magnetic stimulation) and subthreshold (e.g., transcranial DCS and transcranial alternating current stimulation) neuromodulation approaches. Axon terminals polarized with sensitivity (mV of membrane polarization per V/m electric field) 4 times than somas. Even weak polarization (<2 mV) of axon terminals significantly changes action potential dynamics (including amplitude, duration, conduction velocity) in response to an intracellular pulse. Regarding a cellular theory of neuromodulation, we explain how suprathreshold CNS stimulation activates the action potential at terminals while subthreshold approaches modulate synaptic efficacy through axon terminal polarization. We demonstrate that by virtue of axon polarization and resulting changes in action potential dynamics, neuromodulation can influence analog-digital information processing.


Asunto(s)
Corteza Cerebral/citología , Potenciales Evocados/fisiología , Neuronas/citología , Neuronas/fisiología , Terminales Presinápticos/fisiología , Factores de Edad , Animales , Biofisica , Polaridad Celular/fisiología , Estimulación Eléctrica , Femenino , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Neurológicos , Técnicas de Placa-Clamp
10.
J Neurosci ; 35(49): 16213-20, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26658871

RESUMEN

Angelman syndrome (AS) is a neurodevelopmental disorder associated with developmental delay, lack of speech, motor dysfunction, and epilepsy. In the majority of the patients, AS is caused by the deletion of small portions of maternal chromosome 15 harboring the UBE3A gene. This results in a lack of expression of the UBE3A gene because the paternal allele is genetically imprinted. The UBE3A gene encodes an enzyme termed ubiquitin ligase E3A (E6-AP) that targets proteins for degradation by the 26S proteasome. Because neurodegenerative disease and other neurodevelopmental disorders have been linked to oxidative stress, we asked whether mitochondrial reactive oxygen species (ROS) played a role in impaired synaptic plasticity and memory deficits exhibited by AS model mice. We discovered that AS mice have increased levels of superoxide in area CA1 of the hippocampus that is reduced by MitoQ 10-methanesuflonate (MitoQ), a mitochondria-specific antioxidant. In addition, we found that MitoQ rescued impairments in hippocampal synaptic plasticity and deficits in contextual fear memory exhibited by AS model mice. Our findings suggest that mitochondria-derived oxidative stress contributes to hippocampal pathophysiology in AS model mice and that targeting mitochondrial ROS pharmacologically could benefit individuals with AS. SIGNIFICANCE STATEMENT: Oxidative stress has been hypothesized to contribute to the pathophysiology of neurodevelopmental disorders, including autism spectrum disorders and Angelman syndrome (AS). Herein, we report that AS model mice exhibit elevated levels of mitochondria-derived reactive oxygen species in pyramidal neurons in hippocampal area CA1. Moreover, we demonstrate that the administration of MitoQ (MitoQ 10-methanesuflonate), a mitochondria-specific antioxidant, to AS model mice normalizes synaptic plasticity and restores memory. Finally, our findings suggest that antioxidants that target the mitochondria could be used therapeutically to ameliorate synaptic and cognitive deficits in individuals with AS.


Asunto(s)
Síndrome de Angelman/complicaciones , Hipocampo , Mitocondrias/metabolismo , Trastornos del Movimiento/etiología , Trastornos del Movimiento/patología , Superóxidos/metabolismo , Sinapsis/fisiología , Análisis de Varianza , Animales , Condicionamiento Psicológico , Modelos Animales de Enfermedad , Estimulación Eléctrica , Miedo , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/ultraestructura , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Actividad Motora/fisiología , Compuestos Organofosforados/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo
11.
J Neurosci Res ; 94(5): 401-8, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26864654

RESUMEN

Group I metabotropic glutamate receptors (mGluR), including mGluR1 and mGluR 5 (mGluR1/5), are coupled to Gq and modulate activity-dependent synaptic plasticity. Direct activation of mGluR1/5 causes protein translation-dependent long-term depression (LTD). Although it has been established that intracellular Ca(2+) and the Gq-regulated signaling molecules are required for mGluR1/5 LTD, whether and how Ca(2+) regulates Gq signaling and upregulation of protein expression remain unknown. Through pharmacological inhibition, we tested the function of the Ca(2+) sensor calmodulin (CaM) in intracellular signaling triggered by the activation of mGluR1/5. CaM inhibitor N-[4-aminobutyl]-5-chloro-2-naphthalenesulfonamide hydrochloride (W13) suppressed the mGluR1/5-stimulated activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and p70-S6 kinase 1 (S6K1) in hippocampal neurons. W13 also blocked the mGluR1/5 agonist-induced synaptic depression in hippocampal slices and in anesthetized mice. Consistent with the function of CaM, inhibiting the downstream targets Ca(2+) /CaM-dependent protein kinases (CaMK) blocked ERK1/2 and S6K1 activation. Furthermore, disruption of the CaM-CaMK-ERK1/2 signaling cascade suppressed the mGluR1/5-stimulated upregulation of Arc expression. Altogether, our data suggest CaM as a new Gq signaling component for coupling Ca(2+) and protein upregulation and regulating mGluR1/5-mediated synaptic modification.


Asunto(s)
Calmodulina/metabolismo , Depresión Sináptica a Largo Plazo/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Transducción de Señal/fisiología , Animales , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Calmodulina/antagonistas & inhibidores , Células Cultivadas , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología
12.
Learn Mem ; 21(5): 298-304, 2014 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-24741110

RESUMEN

The proper regulation of translation is required for the expression of long-lasting synaptic plasticity. A major site of translational control involves the phosphorylation of eukaryotic initiation factor 2 α (eIF2α) by PKR-like endoplasmic reticulum (ER) kinase (PERK). To determine the role of PERK in hippocampal synaptic plasticity, we used the Cre-lox expression system to selectively disrupt PERK expression in the adult mouse forebrain. Here, we demonstrate that in hippocampal area CA1, metabotropic glutamate receptor (mGluR)-dependent long-term depression (LTD) is associated with increased eIF2α phosphorylation, whereas stimulation of early- and late-phase long-term potentiation (E-LTP and L-LTP, respectively) is associated with decreased eIF2α phosphorylation. Interesting, although PERK-deficient mice exhibit exaggerated mGluR-LTD, both E-LTP and L-LTP remained intact. We also found that mGluR-LTD is associated with a PERK-dependent increase in eIF2α phosphorylation. Our findings are consistent with the notion that eIF2α phosphorylation is a key site for the bidirectional control of persistent forms of synaptic LTP and LTD and suggest a distinct role for PERK in mGluR-LTD.


Asunto(s)
Región CA1 Hipocampal/fisiología , Depresión Sináptica a Largo Plazo/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , eIF-2 Quinasa/metabolismo , Análisis de Varianza , Animales , Fenómenos Biofísicos/efectos de los fármacos , Fenómenos Biofísicos/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteínas de Unión al ADN/metabolismo , Estimulación Eléctrica , Técnicas In Vitro , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Factores de Transcripción/metabolismo , eIF-2 Quinasa/genética
13.
Int J Neuropsychopharmacol ; 17(2): 337-40, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24229565

RESUMEN

Negative symptoms in schizophrenia are associated with decreased dopaminergic activity in the prefrontal cortex (PFC). It is hypothesized that increasing dopamine levels would alleviate negative symptoms. Termination of dopamine activity in the PFC is mainly via catechol-O-methyl tranferase (COMT) activity. Hence, inhibition of COMT activity with entacapone should reverse PFC dopaminergic transmission. To assess the efficacy of entacapone addition to antipsychotic treatment in patients with residual schizophrenia, we conducted a double-blind, randomised, placebo-controlled study for 12 wk of treatment with entacapone or placebo. Clinical measures (PANSS, CGI and QLS) were obtained at baseline and at weeks 4, 8 and 12 and cognitive functions were assessed by the RBANSS. Significant improvement over time in PANSS and QLS scores was observed in both groups. However, entacapone did not demonstrate a beneficial effect compared to placebo. Therefore, this study does not support a therapeutic role for entacapone in residual schizophrenia.


Asunto(s)
Antipsicóticos/administración & dosificación , Catecoles/administración & dosificación , Nitrilos/administración & dosificación , Esquizofrenia/diagnóstico , Esquizofrenia/tratamiento farmacológico , Psicología del Esquizofrénico , Adolescente , Adulto , Catecol O-Metiltransferasa/metabolismo , Inhibidores de Catecol O-Metiltransferasa , Método Doble Ciego , Quimioterapia Combinada , Inhibidores Enzimáticos/administración & dosificación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Esquizofrenia/enzimología , Resultado del Tratamiento , Adulto Joven
14.
iScience ; 27(3): 109230, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38433894

RESUMEN

Transcranial direct current stimulation (tDCS) induces subcellular compartmental-dependent polarization, maximal in the distal portions of axons and dendrites. Using a morphologically realistic neuron model, we simulated tDCS-induced membrane polarization of the apical dendrite. Thus, we investigated the differential dendritic effects of anodal and cathodal tDCS on membrane potential polarization along the dendritic structure and its subsequent effects on dendritic membrane resistance, excitatory postsynaptic potential amplitude, backpropagating action potential amplitude, input/output relations, and long-term synaptic plasticity. We further showed that the effects of anodal and cathodal tDCS on the backpropagating action potential were asymmetric, and explained this asymmetry. Additionally, we showed that the effects on input/output relations were rather weak and limited to the low-mid range of stimulation frequencies, and that synaptic plasticity effects were mostly limited to the distal portion of the dendrite. Thus, we demonstrated how tDCS modifies dendritic physiology due to the dendrite's unique morphology and composition of voltage-gated ion channels.

15.
Mol Autism ; 15(1): 31, 2024 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-39049050

RESUMEN

BACKGROUND: Angelman syndrome (AS) is a rare neurodevelopmental genetic disorder caused by the loss of function of the ubiquitin ligase E3A (UBE3A) gene, affecting approximately 1:15,000 live births. We have recently shown that mitochondrial function in AS is altered during mid to late embryonic brain development leading to increased oxidative stress and enhanced apoptosis of neural precursor cells. However, the overall alterations of metabolic processes are still unknown. Hence, as a follow-up, we aim to investigate the metabolic profiles of wild-type (WT) and AS littermates and to identify which metabolic processes are aberrant in the brain of AS model mice during embryonic development. METHODS: We collected brain tissue samples from mice embryos at E16.5 and performed metabolomic analyses using proton nuclear magnetic resonance (1H-NMR) spectroscopy. Multivariate and Univariate analyses were performed to determine the significantly altered metabolites in AS mice. Pathways associated with the altered metabolites were identified using metabolite set enrichment analysis. RESULTS: Our analysis showed that overall, the metabolomic fingerprint of AS embryonic brains differed from those of their WT littermates. Moreover, we revealed a significant elevation of distinct metabolites, such as acetate, lactate, and succinate in the AS samples compared to the WT samples. The elevated metabolites were significantly associated with the pyruvate metabolism and glycolytic pathways. LIMITATIONS: Only 14 metabolites were successfully identified and investigated in the present study. The effect of unidentified metabolites and their unresolved peaks was not determined. Additionally, we conducted the metabolomic study on whole brain tissue samples. Employing high-resolution NMR studies on different brain regions could further expand our knowledge regarding metabolic alterations in the AS brain. Furthermore, increasing the sample size could reveal the involvement of more significantly altered metabolites in the pathophysiology of the AS brain. CONCLUSIONS: Ube3a loss of function alters bioenergy-related metabolism in the AS brain during embryonic development. Furthermore, these neurochemical changes could be linked to the mitochondrial reactive oxygen species and oxidative stress that occurs during the AS embryonic development.


Asunto(s)
Síndrome de Angelman , Encéfalo , Modelos Animales de Enfermedad , Metabolómica , Espectroscopía de Protones por Resonancia Magnética , Animales , Síndrome de Angelman/metabolismo , Síndrome de Angelman/genética , Encéfalo/metabolismo , Encéfalo/diagnóstico por imagen , Ratones , Metaboloma , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Femenino
16.
Cells ; 13(3)2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38334596

RESUMEN

INTRODUCTION: Transcranial direct current stimulation (tDCS) is an evolving non-invasive neurostimulation technique. Despite multiple studies, its underlying molecular mechanisms are still unclear. Several previous human studies of the effect of tDCS suggest that it generates metabolic effects. The induction of metabolic effects by tDCS could provide an explanation for how it generates its long-term beneficial clinical outcome. AIM: Given these hints of tDCS metabolic effects, we aimed to delineate the metabolic pathways involved in its mode of action. METHODS: To accomplish this, we utilized a broad analytical approach of co-analyzing metabolomics and transcriptomic data generated from anodal tDCS in rat models. Since no metabolomic dataset was available, we performed a tDCS experiment of bilateral anodal stimulation of 200 µA for 20 min and for 5 consecutive days, followed by harvesting the brain tissue below the stimulating electrode and generating a metabolomics dataset using LC-MS/MS. The analysis of the transcriptomic dataset was based on a publicly available dataset. RESULTS: Our analyses revealed that tDCS alters the metabolic profile of brain tissue, affecting bioenergetic-related pathways, such as glycolysis and mitochondrial functioning. In addition, we found changes in calcium-related signaling. CONCLUSIONS: We conclude that tDCS affects metabolism by modulating energy production-related processes. Given our findings concerning calcium-related signaling, we suggest that the immediate effects of tDCS on calcium dynamics drive modifications in distinct metabolic pathways. A thorough understanding of the underlying molecular mechanisms of tDCS has the potential to revolutionize its applicability, enabling the generation of personalized medicine in the field of neurostimulation and thus contributing to its optimization.


Asunto(s)
Estimulación Transcraneal de Corriente Directa , Humanos , Ratas , Animales , Estimulación Transcraneal de Corriente Directa/métodos , Calcio , Cromatografía Liquida , Espectrometría de Masas en Tándem , Perfilación de la Expresión Génica
17.
Digit Health ; 9: 20552076231177132, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37312951

RESUMEN

Objectives: To delineate areas of consensus and disagreements among practicing psychiatrists from various levels of clinical experience, hierarchy and organizations, and to test their ability to converge toward agreement, which will enable better integration of telepsychiatry into mental health services. Methods: To study attitudes of Israeli public health psychiatrists, we utilized a policy Delphi method, during the early stages of the COVID pandemic. In-depth interviews were conducted and analyzed, and a questionnaire was generated. The questionnaire was disseminated amongst 49 psychiatrists, in two succeeding rounds, and areas of consensus and controversies were identified. Results: Psychiatrists showed an overall consensus regarding issues of economic and temporal advantages of telepsychiatry. However, the quality of diagnosis and treatment and the prospect of expanding the usage of telepsychiatry to normal circumstances-beyond situations of pandemic or emergency were disputed. Nonetheless, efficiency and willingness scales slightly improved during the 2nd round of the Delphi process. Prior experience with telepsychiatry had a strong impact on the attitude of psychiatrists, and those who were familiar with this practice were more favorable toward its usage in their clinic. Conclusions: We have delineated experience as a major impact on the attitudes toward telepsychiatry and the willingness for its assimilation in clinical practice as a legitimate and trustworthy method. We have also observed that the organizational affiliation significantly affected psychiatrists' attitude, when those working at local clinics were more positive toward telepsychiatry compared with employees of governmental institutions. This might be related to experience and differences in organizational environment. Taken together, we recommend to include hands-on training of telepsychiatry in medical education curriculum during residency, as well as refresher exercises for attending practitioners.

18.
Front Cell Neurosci ; 17: 1096823, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37020846

RESUMEN

Homeostasis is a precondition for any physiological system of any living organism. Nonetheless, models of learning and memory that are based on processes of synaptic plasticity are unstable by nature according to Hebbian rules, and it is not fully clear how homeostasis is maintained during these processes. This is where theoretical and computational frameworks can help in gaining a deeper understanding of the various cellular processes that enable homeostasis in the face of plasticity. A previous simplistic single compartmental model with a single synapse showed that maintaining input/output response homeostasis and stable synaptic learning could be enabled by introducing a linear relationship between synaptic plasticity and HCN conductance plasticity. In this study, we aimed to examine whether this approach could be extended to a more morphologically realistic model that entails multiple synapses and gradients of various VGICs. In doing so, we found that a linear relationship between synaptic plasticity and HCN conductance plasticity was able to maintain input/output response homeostasis in our morphologically realistic model, where the slope of the linear relationship was dependent on baseline HCN conductance and synaptic permeability values. An increase in either baseline HCN conductance or synaptic permeability value led to a decrease in the slope of the linear relationship. We further show that in striking contrast to the single compartment model, here linear relationship was insufficient in maintaining stable synaptic learning despite maintaining input/output response homeostasis. Additionally, we showed that homeostasis of input/output response profiles was at the expense of decreasing the mutual information transfer due to the increase in noise entropy, which could not be fully rescued by optimizing the linear relationship between synaptic and HCN conductance plasticity. Finally, we generated a place cell model based on theta oscillations and show that synaptic plasticity disrupts place cell activity. Whereas synaptic plasticity accompanied by HCN conductance plasticity through linear relationship maintains the stability of place cell activity. Our study establishes potential differences between a single compartmental model and a morphologically realistic model.

19.
J Neurosci ; 31(48): 17637-48, 2011 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-22131424

RESUMEN

The axon initial segment (AIS) is the site of action potential initiation in neurons. Recent studies have demonstrated activity-dependent regulation of the AIS, including homeostatic changes in AIS length, membrane excitability, and the localization of voltage-gated Na(+) channels. The neurodevelopmental disorder Angelman syndrome (AS) is usually caused by the deletion of small portions of the maternal copy of chromosome 15, which includes the UBE3A gene. A mouse model of AS has been generated and these mice exhibit multiple neurological abnormalities similar to those observed in humans. We examined intrinsic properties of pyramidal neurons in hippocampal area CA1 from AS model mice and observed alterations in resting membrane potential, threshold potential, and action potential amplitude. The altered intrinsic properties in the AS mice were correlated with significant increases in the expression of the α1 subunit of Na/K-ATPase (α1-NaKA), the Na(+) channel NaV1.6, and the AIS anchoring protein ankyrin-G, as well as an increase in length of the AIS. These findings are the first evidence for pathology of intrinsic membrane properties and AIS-specific changes in AS, a neurodevelopmental disorder associated with autism.


Asunto(s)
Potenciales de Acción/fisiología , Síndrome de Angelman/fisiopatología , Axones/fisiología , Potenciales de la Membrana/fisiología , Células Piramidales/fisiología , Potenciales de Acción/genética , Síndrome de Angelman/genética , Animales , Modelos Animales de Enfermedad , Hipocampo/fisiopatología , Potenciales de la Membrana/genética , Ratones
20.
Neurobiol Dis ; 45(3): 1101-10, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22198573

RESUMEN

Tuberous sclerosis complex (TSC) and fragile X syndrome (FXS) are caused by mutations in negative regulators of translation. FXS model mice exhibit enhanced metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD). Therefore, we hypothesized that a mouse model of TSC, ΔRG transgenic mice, also would exhibit enhanced mGluR-LTD. We measured the impact of TSC2-GAP mutations on the mTORC1 and ERK signaling pathways and protein synthesis-dependent hippocampal synaptic plasticity in ΔRG transgenic mice. These mice express a dominant/negative TSC2 that binds to TSC1, but has a deletion and substitution mutation in its GAP-domain, resulting in inactivation of the complex. Consistent with previous studies of several other lines of TSC model mice, we observed elevated S6 phosphorylation in the brains of ΔRG mice, suggesting upregulated translation. Surprisingly, mGluR-LTD was not enhanced, but rather was impaired in the ΔRG transgenic mice, indicating that TSC and FXS have divergent synaptic plasticity phenotypes. Similar to patients with TSC, the ΔRG transgenic mice exhibit elevated ERK signaling. Moreover, the mGluR-LTD impairment displayed by the ΔRG transgenic mice was rescued with the MEK-ERK inhibitor U0126. Our results suggest that the mGluR-LTD impairment observed in ΔRG mice involves aberrant TSC1/2-ERK signaling.


Asunto(s)
Depresión Sináptica a Largo Plazo/genética , Sistema de Señalización de MAP Quinasas/genética , Receptores de Glutamato Metabotrópico/metabolismo , Esclerosis Tuberosa/complicaciones , Esclerosis Tuberosa/genética , Proteínas Supresoras de Tumor/deficiencia , Análisis de Varianza , Animales , Animales Recién Nacidos , Biofisica , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Hipocampo/patología , Técnicas In Vitro , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Placa-Clamp , Receptores de Glutamato Metabotrópico/genética , Proteínas Quinasas S6 Ribosómicas/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Esclerosis Tuberosa/metabolismo , Esclerosis Tuberosa/patología , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA