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











Base de datos
Intervalo de año de publicación
1.
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
2.
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.

3.
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
4.
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
5.
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.

6.
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.

7.
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
8.
Front Cell Neurosci ; 16: 906313, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35936503

RESUMEN

The prerequisites for neurons to function within a circuit and be able to contain and transfer information efficiently and reliably are that they need to be homeostatically stable and fire within a reasonable range, characteristics that are governed, among others, by voltage-gated ion channels (VGICs). Nonetheless, neurons entail large variability in the expression levels of VGICs and their corresponding intrinsic properties, but the role of this variability in information transfer is not fully known. In this study, we aimed to investigate how this variability of VGICs affects information transfer. For this, we used a previously derived population of neuronal model neurons, each with the variable expression of five types of VGICs, fast Na+, delayed rectifier K+, A-type K+, T-type Ca++, and HCN channels. These analyses showed that the model neurons displayed variability in mutual information transfer, measured as the capability of neurons to successfully encode incoming synaptic information in output firing frequencies. Likewise, variability in the expression of VGICs caused variability in EPSPs and IPSPs amplitudes, reflected in the variability of output firing frequencies. Finally, using the virtual knockout methodology, we show that among the ion channels tested, the A-type K+ channel is the major regulator of information processing and transfer.

9.
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
10.
Brain Stimul ; 15(3): 861-869, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35640845

RESUMEN

BACKGROUND: Transcranial direct current stimulation (tDCS) is a non-invasive sub-threshold stimulation, widely accepted for its amelioration of distinct neuropsychiatric disorders. The weak electric field of tDCS modulates the activity of cortical neurons, which in turn modifies brain functioning. However, the underlying mechanisms for that are not fully understood. OBJECTIVE/HYPOTHESIS: Previous studies demonstrated that the axons are the most sensitive subcellular compartment for tDCS-induced polarization. Moreover, it was posited that DCS-induced axonal polarization is amplified by modifying the conductance of ionic channels. We posit that voltage-gated potassium-channels that are highly expressed in axons play a crucial role in DCS-induced modulation of cortical neurons functioning. METHODS: We examined the involvement of voltage-gated potassium-channels in the active modulation of spontaneous vesicle release by DCS. For that, we measured spontaneous excitatory postsynaptic currents (sEPSCs) from layer-V motor cortex during DCS application, while co-applying distinct voltage-gated potassium-channels blockers. Moreover, we examined the role of Kv1 potassium channels in DCS-induced modulation of action potential waveform at axon terminals by recording action potentials at terminal axon blebs during DCS application while locally inhibiting the Kv1 potassium-channels. RESULTS: We demonstrated that inhibiting voltage-gated potassium-channels occluded the DCS-induced modulation of subthreshold presynaptic vesicle release. Moreover, we showed that inhibiting Kv1 voltage-gated potassium-channels also occluded the DCS-induced modulation of action potential waveform at axon terminals. CONCLUSION: We suggest that DCS-induced depolarization inactivates the Kv1 potassium channels thus reducing potassium conductance, which amplifies axonal depolarization, subsequently enhancing the presynaptic component of synaptic transmission. Whereas DCS-induced hyperpolarization induces opposite effects.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Estimulación Transcraneal de Corriente Directa , Potenciales de Acción/fisiología , Axones/fisiología , Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/farmacología , Canales de Potasio de la Superfamilia Shaker/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA