Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.625
Filtrar
1.
CNS Neurosci Ther ; 30(5): e14740, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38715318

RESUMEN

AIMS: γ-aminobutyric acid (GABA) from reactive astrocytes is critical for the dysregulation of neuronal activity in various neuroinflammatory conditions. While Scutellaria baicalensis Georgi (S. baicalensis) is known for its efficacy in addressing neurological symptoms, its potential to reduce GABA synthesis in reactive astrocytes and the associated neuronal suppression remains unclear. This study focuses on the inhibitory action of monoamine oxidase B (MAO-B), the key enzyme for astrocytic GABA synthesis. METHODS: Using a lipopolysaccharide (LPS)-induced neuroinflammation mouse model, we conducted immunohistochemistry to assess the effect of S. baicalensis on astrocyte reactivity and its GABA synthesis. High-performance liquid chromatography was performed to reveal the major compounds of S. baicalensis, the effects of which on MAO-B inhibition, astrocyte reactivity, and tonic inhibition in hippocampal neurons were validated by MAO-B activity assay, qRT-PCR, and whole-cell patch-clamp. RESULTS: The ethanolic extract of S. baicalensis ameliorated astrocyte reactivity and reduced excessive astrocytic GABA content in the CA1 hippocampus. Baicalin and baicalein exhibited significant MAO-B inhibition potential. These two compounds downregulate the mRNA levels of genes associated with reactive astrogliosis or astrocytic GABA synthesis. Additionally, LPS-induced aberrant tonic inhibition was reversed by both S. baicalensis extract and its key compounds. CONCLUSIONS: In summary, baicalin and baicalein isolated from S. baicalensis reduce astrocyte reactivity and alleviate aberrant tonic inhibition of hippocampal neurons during neuroinflammation.


Asunto(s)
Astrocitos , Flavanonas , Flavonoides , Lipopolisacáridos , Neuronas , Extractos Vegetales , Scutellaria baicalensis , Ácido gamma-Aminobutírico , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Flavanonas/farmacología , Scutellaria baicalensis/química , Ratones , Ácido gamma-Aminobutírico/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Masculino , Flavonoides/farmacología , Extractos Vegetales/farmacología , Lipopolisacáridos/toxicidad , Lipopolisacáridos/farmacología , Ratones Endogámicos C57BL , Monoaminooxidasa/metabolismo , Inhibición Neural/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo
2.
Cereb Cortex ; 34(5)2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38771240

RESUMEN

In vitro and ex vivo studies have shown consistent indications of hyperexcitability in the Fragile X Messenger Ribonucleoprotein 1 (Fmr1) knockout mouse model of autism spectrum disorder. We recently introduced a method to quantify network-level functional excitation-inhibition ratio from the neuronal oscillations. Here, we used this measure to study whether the implicated synaptic excitation-inhibition disturbances translate to disturbances in network physiology in the Fragile X Messenger Ribonucleoprotein 1 (Fmr1) gene knockout model. Vigilance-state scoring was used to extract segments of inactive wakefulness as an equivalent behavioral condition to the human resting-state and, subsequently, we performed high-frequency resolution analysis of the functional excitation-inhibition biomarker, long-range temporal correlations, and spectral power. We corroborated earlier studies showing increased high-frequency power in Fragile X Messenger Ribonucleoprotein 1 (Fmr1) knockout mice. Long-range temporal correlations were higher in the gamma frequency ranges. Contrary to expectations, functional excitation-inhibition was lower in the knockout mice in high frequency ranges, suggesting more inhibition-dominated networks. Exposure to the Gamma-aminobutyric acid (GABA)-agonist clonazepam decreased the functional excitation-inhibition in both genotypes, confirming that increasing inhibitory tone results in a reduction of functional excitation-inhibition. In addition, clonazepam decreased electroencephalogram power and increased long-range temporal correlations in both genotypes. These findings show applicability of these new resting-state electroencephalogram biomarkers to animal for translational studies and allow investigation of the effects of lower-level disturbances in excitation-inhibition balance.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Ratones Noqueados , Neuronas , Animales , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Neuronas/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratones , Masculino , Inhibición Neural/fisiología , Inhibición Neural/efectos de los fármacos , Ratones Endogámicos C57BL , Electroencefalografía
3.
J Physiol ; 602(10): 2253-2264, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38638084

RESUMEN

Short- and long-latency afferent inhibition (SAI and LAI respectively) are phenomenon whereby the motor evoked potential induced by transcranial magnetic stimulation (TMS) is inhibited by a sensory afferent volley consequent to nerve stimulation. It remains unclear whether dopamine participates in the genesis or modulation of SAI and LAI. The present study aimed to determine if SAI and LAI are modulated by levodopa (l-DOPA). In this placebo-controlled, double-anonymized study Apo-Levocarb (100 mg l-DOPA in combination with 25 mg carbidopa) and a placebo were administered to 32 adult males (mean age 24 ± 3 years) in two separate sessions. SAI and LAI were evoked by stimulating the median nerve and delivering single-pulse TMS over the motor hotspot corresponding to the first dorsal interosseous muscle of the right hand. SAI and LAI were quantified before and 1 h following ingestion of drug or placebo corresponding to the peak plasma concentration of Apo-Levocarb. The results indicate that Apo-Levocarb increases SAI and does not significantly alter LAI. These findings support literature demonstrating increased SAI following exogenous dopamine administration in neurodegenerative disorders. KEY POINTS: Short- and long-latency afferent inhibition (SAI and LAI respectively) are measures of corticospinal excitability evoked using transcranial magnetic stimulation. SAI and LAI are reduced in conditions such as Parkinson's disease which suggests dopamine may be involved in the mechanism of afferent inhibition. 125 mg of Apo-Levocarb (100 mg dopamine) increases SAI but not LAI. This study increases our understanding of the pharmacological mechanism of SAI and LAI.


Asunto(s)
Carbidopa , Potenciales Evocados Motores , Levodopa , Estimulación Magnética Transcraneal , Humanos , Masculino , Levodopa/farmacología , Adulto , Potenciales Evocados Motores/efectos de los fármacos , Estimulación Magnética Transcraneal/métodos , Carbidopa/farmacología , Adulto Joven , Inhibición Neural/efectos de los fármacos , Método Doble Ciego , Dopaminérgicos/farmacología , Dopamina/farmacología , Combinación de Medicamentos , Nervio Mediano/fisiología , Nervio Mediano/efectos de los fármacos
4.
J Neurosci ; 44(19)2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38553046

RESUMEN

Exercise is known to benefit motor skill learning in health and neurological disease. Evidence from brain stimulation, genotyping, and Parkinson's disease studies converge to suggest that the dopamine D2 receptor, and shifts in the cortical excitation and inhibition (E:I) balance, are prime candidates for the drivers of exercise-enhanced motor learning. However, causal evidence using experimental pharmacological challenge is lacking. We hypothesized that the modulatory effect of the dopamine D2 receptor on exercise-induced changes in the E:I balance would determine the magnitude of motor skill acquisition. To test this, we measured exercise-induced changes in excitation and inhibition using paired-pulse transcranial magnetic stimulation (TMS) in 22 healthy female and male humans, and then had participants learn a novel motor skill-the sequential visual isometric pinch task (SVIPT). We examined the effect of D2 receptor blockade (800 mg sulpiride) on these measures within a randomized, double-blind, placebo-controlled design. Our key result was that motor skill acquisition was driven by an interaction between the D2 receptor and E:I balance. Specifically, poorer skill learning was related to an attenuated shift in the E:I balance in the sulpiride condition, whereas this interaction was not evident in placebo. Our results demonstrate that exercise-primed motor skill acquisition is causally influenced by D2 receptor activity on motor cortical circuits.


Asunto(s)
Ejercicio Físico , Corteza Motora , Destreza Motora , Receptores de Dopamina D2 , Estimulación Magnética Transcraneal , Humanos , Masculino , Femenino , Receptores de Dopamina D2/metabolismo , Adulto , Destreza Motora/fisiología , Destreza Motora/efectos de los fármacos , Estimulación Magnética Transcraneal/métodos , Adulto Joven , Corteza Motora/fisiología , Corteza Motora/efectos de los fármacos , Ejercicio Físico/fisiología , Método Doble Ciego , Inhibición Neural/fisiología , Inhibición Neural/efectos de los fármacos , Aprendizaje/fisiología , Potenciales Evocados Motores/fisiología , Potenciales Evocados Motores/efectos de los fármacos , Sulpirida/farmacología , Antagonistas de Dopamina/farmacología
5.
Proc Natl Acad Sci U S A ; 119(22): e2203680119, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35622887

RESUMEN

Noradrenergic activation of the basolateral amygdala (BLA) by emotional arousal enhances different forms of recognition memory via functional interactions with the insular cortex (IC). Human neuroimaging studies have revealed that the anterior IC (aIC), as part of the salience network, is dynamically regulated during arousing situations. Emotional stimulation first rapidly increases aIC activity but suppresses it in a delayed fashion. Here, we investigated in male Sprague-Dawley rats whether the BLA influence on recognition memory is associated with an increase or suppression of aIC activity during the postlearning consolidation period. We first employed anterograde and retrograde viral tracing and found that the BLA sends dense monosynaptic projections to the aIC. Memory-enhancing norepinephrine administration into the BLA following an object training experience suppressed aIC activity 1 h later, as determined by a reduced expression of the phosphorylated form of the transcription factor cAMP response element-binding (pCREB) protein and neuronal activity marker c-Fos. In contrast, the number of perisomatic γ-aminobutyric acid (GABA)ergic inhibitory synapses per pCREB-positive neuron was significantly increased, suggesting a dynamic up-regulation of GABAergic tone. In support of this possibility, pharmacological inhibition of aIC activity with a GABAergic agonist during consolidation enhanced object recognition memory. Norepinephrine administration into the BLA did not affect neuronal activity within the posterior IC, which receives sparse innervation from the BLA. The evidence that noradrenergic activation of the BLA enhances the consolidation of object recognition memory via a mechanism involving a suppression of aIC activity provides insight into the broader brain network dynamics underlying emotional regulation of memory.


Asunto(s)
Complejo Nuclear Basolateral , Emociones , Corteza Insular , Inhibición Neural , Reconocimiento en Psicología , Percepción Visual , Animales , Nivel de Alerta , Complejo Nuclear Basolateral/efectos de los fármacos , Complejo Nuclear Basolateral/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Emociones/efectos de los fármacos , Emociones/fisiología , Agonistas del GABA/farmacología , Corteza Insular/efectos de los fármacos , Corteza Insular/fisiología , Masculino , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Norepinefrina/administración & dosificación , Norepinefrina/farmacología , Ratas , Ratas Sprague-Dawley , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Percepción Visual/fisiología
6.
Nature ; 602(7895): 112-116, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35046577

RESUMEN

The biological basis of male-female brain differences has been difficult to elucidate in humans. The most notable morphological difference is size, with male individuals having on average a larger brain than female individuals1,2, but a mechanistic understanding of how this difference arises remains unknown. Here we use brain organoids3 to show that although sex chromosomal complement has no observable effect on neurogenesis, sex steroids-namely androgens-lead to increased proliferation of cortical progenitors and an increased neurogenic pool. Transcriptomic analysis and functional studies demonstrate downstream effects on histone deacetylase activity and the mTOR pathway. Finally, we show that androgens specifically increase the neurogenic output of excitatory neuronal progenitors, whereas inhibitory neuronal progenitors are not increased. These findings reveal a role for androgens in regulating the number of excitatory neurons and represent a step towards understanding the origin of sex-related brain differences in humans.


Asunto(s)
Andrógenos/farmacología , Encéfalo/citología , Excitabilidad Cortical/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Organoides/citología , Organoides/efectos de los fármacos , Caracteres Sexuales , Potenciales de Acción/efectos de los fármacos , Andrógenos/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/metabolismo , Recuento de Células , Femenino , Perfilación de la Expresión Génica , Histona Desacetilasas/genética , Humanos , Masculino , Inhibición Neural/efectos de los fármacos , Neuroglía/citología , Neuroglía/efectos de los fármacos , Tamaño de los Órganos/efectos de los fármacos , Organoides/enzimología , Organoides/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética
7.
Cell Rep ; 37(5): 109950, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34731619

RESUMEN

Evidence for prefrontal cortical (PFC) GABAergic dysfunction is one of the most consistent findings in schizophrenia and may contribute to cognitive deficits. Recent studies suggest that the mGlu1 subtype of metabotropic glutamate receptor regulates cortical inhibition; however, understanding the mechanisms through which mGlu1 positive allosteric modulators (PAMs) regulate PFC microcircuit function and cognition is essential for advancing these potential therapeutics toward the clinic. We report a series of electrophysiology, optogenetic, pharmacological magnetic resonance imaging, and animal behavior studies demonstrating that activation of mGlu1 receptors increases inhibitory transmission in the prelimbic PFC by selective excitation of somatostatin-expressing interneurons (SST-INs). An mGlu1 PAM reverses cortical hyperactivity and concomitant cognitive deficits induced by N-methyl-d-aspartate (NMDA) receptor antagonists. Using in vivo optogenetics, we show that prelimbic SST-INs are necessary for mGlu1 PAM efficacy. Collectively, these findings suggest that mGlu1 PAMs could reverse cortical GABAergic deficits and exhibit efficacy in treating cognitive dysfunction in schizophrenia.


Asunto(s)
Antipsicóticos/farmacología , Conducta Animal/efectos de los fármacos , Cognición/efectos de los fármacos , Disfunción Cognitiva/tratamiento farmacológico , Agonistas de Aminoácidos Excitadores/farmacología , Glicina/análogos & derivados , Interneuronas/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Receptores de Glutamato Metabotrópico/agonistas , Resorcinoles/farmacología , Esquizofrenia/tratamiento farmacológico , Psicología del Esquizofrénico , Somatostatina/metabolismo , Animales , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/psicología , Modelos Animales de Enfermedad , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Glicina/farmacología , Interneuronas/metabolismo , Masculino , Memoria a Corto Plazo/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibición Neural/efectos de los fármacos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/metabolismo , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Somatostatina/genética
8.
J Psychopharmacol ; 35(11): 1356-1364, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34694190

RESUMEN

BACKGROUND: Delta-9 tetrahydrocannabinol (THC) is a major exogenous psychoactive agent, which acts as a partial agonist on cannabinoid (CB1) receptors. THC is known to inhibit presynaptic neurotransmission and has been repeatedly linked to acute decrements in cognitive function across multiple domains. Previous electrophysiological studies of sensory gating have shown specific deficits in inhibitory processing in cannabis-users, but to date these findings have been limited to the auditory cortices, and the degree to which these aberrations extend to other brain regions remains largely unknown. METHODS: We used magnetoencephalography (MEG) and a paired-pulse somatosensory stimulation paradigm to probe inhibitory processing in 29 cannabis-users (i.e. at least four times per month) and 41 demographically matched non-user controls. MEG responses to each stimulation were imaged in both the oscillatory and time domain, and voxel time-series data were extracted to quantify the dynamics of sensory gating, oscillatory gamma activity, evoked responses, and spontaneous neural activity. RESULTS: We observed robust somatosensory responses following both stimulations, which were used to compute sensory gating ratios. Cannabis-users exhibited significantly impaired gating relative to non-users in somatosensory cortices, as well as decreased spontaneous neural activity. In contrast, oscillatory gamma activity did not appear to be affected by cannabis use. CONCLUSIONS: We observed impaired gating of redundant somatosensory information and altered spontaneous activity in the same cortical tissue in cannabis-users compared to non-users. These data suggest that cannabis use is associated with a decline in the brain's ability to properly filter repetitive information and impairments in cortical inhibitory processing.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Dronabinol/farmacología , Ritmo Gamma/efectos de los fármacos , Uso de la Marihuana/efectos adversos , Inhibición Neural/efectos de los fármacos , Filtrado Sensorial/efectos de los fármacos , Corteza Somatosensorial/efectos de los fármacos , Adulto , Femenino , Humanos , Magnetoencefalografía , Masculino , Adulto Joven
9.
Nat Commun ; 12(1): 6112, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34671051

RESUMEN

Stroke profoundly disrupts cortical excitability which impedes recovery, but how it affects the function of specific inhibitory interneurons, or subpopulations therein, is poorly understood. Interneurons expressing vasoactive intestinal peptide (VIP) represent an intriguing stroke target because they can regulate cortical excitability through disinhibition. Here we chemogenetically augmented VIP interneuron excitability in a murine model of photothrombotic stroke and show that it enhances somatosensory responses and improves recovery of paw function. Using longitudinal calcium imaging, we discovered that stroke primarily disrupts the fidelity (fraction of responsive trials) and predictability of sensory responses within a subset of highly active VIP neurons. Partial recovery of responses occurred largely within these active neurons and was not accompanied by the recruitment of minimally active neurons. Importantly, chemogenetic stimulation preserved sensory response fidelity and predictability in highly active neurons. These findings provide a new depth of understanding into how stroke and prospective therapies (chemogenetics), can influence subpopulations of inhibitory interneurons.


Asunto(s)
Interneuronas/fisiología , Accidente Cerebrovascular/terapia , Péptido Intestinal Vasoactivo/metabolismo , Animales , Clozapina/análogos & derivados , Clozapina/uso terapéutico , Humanos , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Ratones , Inhibición Neural/efectos de los fármacos , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Recuperación de la Función , Corteza Somatosensorial/citología , Corteza Somatosensorial/efectos de los fármacos , Corteza Somatosensorial/fisiología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología
10.
Cell Rep ; 36(8): 109572, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433032

RESUMEN

The dentate gyrus (DG) of the hippocampus plays a key role in memory formation, and it is known to be modulated by septal projections. By performing electrophysiology and optogenetics, we evaluated the role of cholinergic modulation in the processing of afferent inputs in the DG. We show that mature granule cells (GCs), but not adult-born immature neurons, have increased responses to afferent perforant path stimuli upon cholinergic modulation. This is due to a highly precise reconfiguration of inhibitory circuits, differentially affecting Parvalbumin and Somatostatin interneurons, resulting in a nicotinic-dependent perisomatic disinhibition of GCs. This circuit reorganization provides a mechanism by which mature GCs could escape the strong inhibition they receive, creating a window of opportunity for plasticity. Indeed, coincident activation of perforant path inputs with optogenetic release of acetylcholine produces a long-term potentiated response in GCs, essential for memory formation.


Asunto(s)
Acetilcolina/farmacología , Giro Dentado/metabolismo , Interneuronas/metabolismo , Inhibición Neural/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Ratones , Ratones Transgénicos , Optogenética
11.
Cell Rep ; 36(7): 109563, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34407401

RESUMEN

Overconsumption of highly palatable, energy-dense food is considered a key driver of the obesity pandemic. The orbitofrontal cortex (OFC) is critical for reward valuation of gustatory signals, yet how the OFC adapts to obesogenic diets is poorly understood. Here, we show that extended access to a cafeteria diet impairs astrocyte glutamate clearance, which leads to a heterosynaptic depression of GABA transmission onto pyramidal neurons of the OFC. This decrease in GABA tone is due to an increase in extrasynaptic glutamate, which acts via metabotropic glutamate receptors to liberate endocannabinoids. This impairs the induction of endocannabinoid-mediated long-term plasticity. The nutritional supplement, N-acetylcysteine rescues this cascade of synaptic impairments by restoring astrocytic glutamate transport. Together, our findings indicate that obesity targets astrocytes to disrupt the delicate balance between excitatory and inhibitory transmission in the OFC.


Asunto(s)
Astrocitos/patología , Plasticidad Neuronal , Obesidad/fisiopatología , Corteza Prefrontal/fisiopatología , Acetilcisteína/farmacología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Transporte Biológico/efectos de los fármacos , Dieta , Endocannabinoides/metabolismo , Neuronas GABAérgicas/metabolismo , Ácido Glutámico/metabolismo , Homeostasis/efectos de los fármacos , Hipertrofia , Masculino , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Plasticidad Neuronal/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Ratas Long-Evans , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Transmisión Sináptica/fisiología
12.
J Neurosci ; 41(37): 7831-7847, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34348999

RESUMEN

The principal neurons of the striatum, the spiny projection neurons (SPNs), make inhibitory synaptic connections with each other via collaterals of their main axon, forming a local lateral inhibition network. Serotonin, acting via the 5-HT1B receptor, modulates neurotransmitter release from SPN terminals in striatal output nuclei, but the role of 5-HT1B receptors in lateral inhibition among SPNs in the striatum is unknown. Here, we report the effects of 5-HT1B receptor activation on lateral inhibition in the mouse striatum. Whole-cell recordings were made from SPNs in acute brain slices of either sex, while optogenetically activating presynaptic SPNs or fast-spiking interneurons (FSIs). Activation of 5-HT1B receptors significantly reduced the amplitude of IPSCs evoked by optical stimulation of both direct and indirect pathway SPNs. This reduction was blocked by application of a 5-HT1B receptor antagonist. Activation of 5-HT1B receptors did not reduce the amplitude of IPSCs evoked from FSIs. These results suggest a new role for serotonin as a modulator of lateral inhibition among striatal SPNs. The 5-HT1B receptor may, therefore, be a suitable target for future behavioral experiments investigating the currently unknown role of lateral inhibition in the function of the striatum.SIGNIFICANCE STATEMENT We show that stimulation of serotonin receptors reduces the efficacy of lateral inhibition between spiny projection neurons (SPNs), one of the biggest GABAergic sources in the striatum, by activation of the serotonin 5-HT1B receptor. The striatum receives serotonergic input from the dorsal raphe nuclei and is important in behavioral brain functions like learning and action selection. Our findings suggest a new role for serotonin in modulating the dynamics of neural interactions in the striatum, which extends current knowledge of the mechanisms of the behavioral effects of serotonin.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptor de Serotonina 5-HT1B/metabolismo , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Cuerpo Estriado/metabolismo , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Ratones , Neuronas/metabolismo , Técnicas de Placa-Clamp , Serotonina/metabolismo , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo
13.
J Neurophysiol ; 126(4): 1090-1100, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34406874

RESUMEN

The general anesthetic etomidate, which acts through γ-aminobutyric acid type A (GABAA) receptors, impairs the formation of new memories under anesthesia. This study addresses the molecular and cellular mechanisms by which this occurs. Here, using a new line of genetically engineered mice carrying the GABAA receptor (GABAAR) ß2-N265M mutation, we tested the roles of receptors that incorporate GABAA receptor ß2 versus ß3 subunits to suppression of long-term potentiation (LTP), a cellular model of learning and memory. We found that brain slices from ß2-N265M mice resisted etomidate suppression of LTP, indicating that the ß2-GABAARs are an essential target in this model. As these receptors are most heavily expressed by interneurons in the hippocampus, this finding supports a role for interneuron modulation in etomidate control of synaptic plasticity. Nevertheless, ß2 subunits are also expressed by pyramidal neurons, so they might also contribute. Therefore, using a previously established line of ß3-N265M mice, we also examined the contributions of ß2- versus ß3-GABAARs to GABAA,slow dendritic inhibition, because dendritic inhibition is particularly well suited to controlling synaptic plasticity. We also examined their roles in long-lasting suppression of population activity through feedforward and feedback inhibition. We found that both ß2- and ß3-GABAARs contribute to GABAA,slow inhibition and that both ß2- and ß3-GABAARs contribute to feedback inhibition, whereas only ß3-GABAARs contribute to feedforward inhibition. We conclude that modulation of ß2-GABAARs is essential to etomidate suppression of LTP. Furthermore, to the extent that this occurs through GABAARs on pyramidal neurons, it is through modulation of feedback inhibition.NEW & NOTEWORTHY Etomidate exerts its anesthetic actions through GABAA receptors. However, the mechanism remains unknown. Here, using a hippocampal brain slice model, we show that ß2-GABAARs are essential to this effect. We also show that these receptors contribute to long-lasting dendritic inhibition in feedback but not feedforward inhibition of pyramidal neurons. These findings hold implications for understanding how anesthetics block memory formation and, more generally, how inhibitory circuits control learning and memory.


Asunto(s)
Anestésicos Intravenosos/farmacología , Etomidato/farmacología , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Receptores de GABA-A/efectos de los fármacos , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
14.
Cell Rep ; 36(3): 109411, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34289348

RESUMEN

Oxytocin is a well-known neurohypophysial hormone that plays an important role in behavioral anxiety and nociception. Two major forms of long-term potentiation, presynaptic LTP (pre-LTP) and postsynaptic LTP (post-LTP), have been characterized in the anterior cingulate cortex (ACC). Both pre-LTP and post-LTP contribute to chronic-pain-related anxiety and behavioral sensitization. The roles of oxytocin in the ACC have not been studied. Here, we find that microinjections of oxytocin into the ACC attenuate nociceptive responses and anxiety-like behavioral responses in animals with neuropathic pain. Application of oxytocin selectively blocks the maintenance of pre-LTP but not post-LTP. In addition, oxytocin enhances inhibitory transmission and excites ACC interneurons. Similar results are obtained by using selective optical stimulation of oxytocin-containing projecting terminals in the ACC in animals with neuropathic pain. Our results demonstrate that oxytocin acts on central synapses and reduces chronic-pain-induced anxiety by reducing pre-LTP.


Asunto(s)
Ansiedad/fisiopatología , Emociones , Giro del Cíngulo/patología , Potenciación a Largo Plazo , Neuralgia/patología , Neuralgia/fisiopatología , Oxitocina/farmacología , Terminales Presinápticos/patología , Analgésicos/farmacología , Animales , Ansiolíticos/farmacología , Conducta Animal/efectos de los fármacos , Calcio/metabolismo , Dolor Crónico/patología , Dolor Crónico/fisiopatología , Emociones/efectos de los fármacos , Femenino , Giro del Cíngulo/efectos de los fármacos , Giro del Cíngulo/fisiopatología , Interneuronas/efectos de los fármacos , Luz , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Microinyecciones , Tejido Nervioso/efectos de los fármacos , Tejido Nervioso/patología , Tejido Nervioso/fisiopatología , Inhibición Neural/efectos de los fármacos , Neuralgia/complicaciones , Oxitocina/administración & dosificación , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/patología , Núcleo Hipotalámico Paraventricular/fisiopatología , Terminales Presinápticos/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de GABA-A/metabolismo , Receptores de Oxitocina/genética , Receptores de Oxitocina/metabolismo , Transducción de Señal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
15.
Mol Neurobiol ; 58(10): 4787-4801, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34173171

RESUMEN

Glutamate delta-1 receptor (GluD1) is a member of the ionotropic glutamate receptor family expressed at excitatory synapses and functions as a synaptogenic protein by interacting with presynaptic neurexin. We have previously shown that GluD1 plays a role in the maintenance of excitatory synapses in a region-specific manner. Loss of GluD1 leads to reduced excitatory neurotransmission in medium spiny neurons (MSNs) in the dorsal striatum, but not in the ventral striatum (both core and shell of the nucleus accumbens (NAc)). Here, we found that GluD1 loss leads to reduced inhibitory neurotransmission in MSNs of the NAc core as evidenced by a reduction in the miniature inhibitory postsynaptic current frequency and amplitude. Presynaptic effect of GluD1 loss was further supported by an increase in paired pulse ratio of evoked inhibitory responses indicating reduced release probability. Furthermore, analysis of GAD67 puncta indicated a reduction in the number of putative inhibitory terminals. The changes in mIPSC were independent of cannabinoid or dopamine signaling. A role of feed-forward inhibition was tested by selective ablation of GluD1 from PV neurons which produced modest reduction in mIPSCs. Behaviorally, local ablation of GluD1 from NAc led to hypolocomotion and affected anxiety- and depression-like behaviors. When GluD1 was ablated from the dorsal striatum, several behavioral phenotypes were altered in opposite manner compared to GluD1 ablation from NAc. Our findings demonstrate that GluD1 regulates inhibitory neurotransmission in the NAc by a combination of pre- and postsynaptic mechanisms which is critical for motor control and behaviors relevant to neuropsychiatric disorders.


Asunto(s)
Ansiedad/metabolismo , Glutamato Deshidrogenasa/biosíntesis , Potenciales Postsinápticos Inhibidores/fisiología , Inhibición Neural/fisiología , Núcleo Accumbens/metabolismo , Transmisión Sináptica/fisiología , Animales , Ansiedad/genética , Antagonistas de Aminoácidos Excitadores/farmacología , Glutamato Deshidrogenasa/antagonistas & inhibidores , Glutamato Deshidrogenasa/genética , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Ratones Noqueados , Inhibición Neural/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Interacción Social/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
16.
J Psychiatry Neurosci ; 46(3): E402-E414, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34077150

RESUMEN

Background: Bipolar disorder is characterized by cyclical alternation between mania and depression, often comorbid with psychosis and suicide. Compared with other medications, the mood stabilizer lithium is the most effective treatment for the prevention of manic and depressive episodes. However, the pathophysiology of bipolar disorder and lithium's mode of action are yet to be fully understood. Evidence suggests a change in the balance of excitatory and inhibitory activity, favouring excitation in bipolar disorder. In the present study, we sought to establish a holistic understanding of the neuronal consequences of lithium exposure in mouse cortical neurons, and to identify underlying mechanisms of action. Methods: We used a range of technical approaches to determine the effects of acute and chronic lithium treatment on mature mouse cortical neurons. We combined RNA screening and biochemical and electrophysiological approaches with confocal immunofluorescence and live-cell calcium imaging. Results: We found that only chronic lithium treatment significantly reduced intracellular calcium flux, specifically by activating metabotropic glutamatergic receptor 5. This was associated with altered phosphorylation of protein kinase C and glycogen synthase kinase 3, reduced neuronal excitability and several alterations to synapse function. Consequently, lithium treatment shifts the excitatory­inhibitory balance toward inhibition. Limitations: The mechanisms we identified should be validated in future by similar experiments in whole animals and human neurons. Conclusion: Together, the results revealed how lithium dampens neuronal excitability and the activity of the glutamatergic network, both of which are predicted to be overactive in the manic phase of bipolar disorder. Our working model of lithium action enables the development of targeted strategies to restore the balance of overactive networks, mimicking the therapeutic benefits of lithium but with reduced toxicity.


Asunto(s)
Corteza Cerebral/citología , Compuestos de Litio/uso terapéutico , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Receptor del Glutamato Metabotropico 5/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Animales , Trastorno Bipolar/tratamiento farmacológico , Trastorno Bipolar/metabolismo , Calcio/metabolismo , Células Cultivadas , Compuestos de Litio/administración & dosificación , Compuestos de Litio/farmacología , Ratones , Neuronas/metabolismo , Proteína Quinasa C/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Sinapsis/metabolismo
17.
Neurosci Lett ; 756: 135950, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-33979698

RESUMEN

The mechanisms of general anaesthetics such as propofol have drawn substantial attention. The effects of propofol on inhibitory postsynaptic currents are not exactly the same in different brain nuclei. Recent studies revealed that the paraventricular thalamic nucleus (PVT) is a critical nucleus modulating wakefulness. However, the effects of propofol on PVT neurons and the mechanisms underlying such effects remain unknown. Here, we performed the whole-cell recording of the PVT neurons in acute brain slices and bath application of propofol. We found that propofol hyperpolarized the membrane potentials of the PVT neurons and suppressed the action potentials induced by step-current injection. Propofol did not affect the spontaneous inhibitory postsynaptic currents (sIPSCs) amplitude or frequency, but prolonged the sIPSCs half-width. Besides, propofol increased miniature inhibitory synaptic currents (mIPSCs) frequency and half-width. Furthermore, propofol could induce GABAA receptors-mediated tonic inhibitory currents dose-dependently. Thus, our results demonstrate that propofol hyperpolarizes PVT neurons by modulating inhibitory currents via GABAA receptors in mice.


Asunto(s)
Anestésicos Intravenosos/farmacología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Núcleos Talámicos de la Línea Media/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Propofol/farmacología , Animales , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Técnicas de Placa-Clamp
18.
Sci Rep ; 11(1): 10634, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34017040

RESUMEN

Dravet syndrome (DS) is an intractable form of childhood epilepsy that occurs in infancy. More than 80% of all patients have a heterozygous abnormality in the SCN1A gene, which encodes a subunit of Na+ channels in the brain. However, the detailed pathogenesis of DS remains unclear. This study investigated the synaptic pathogenesis of this disease in terms of excitatory/inhibitory balance using a mouse model of DS. We show that excitatory postsynaptic currents were similar between Scn1a knock-in neurons (Scn1a+/- neurons) and wild-type neurons, but inhibitory postsynaptic currents were significantly lower in Scn1a+/- neurons. Moreover, both the vesicular release probability and the number of inhibitory synapses were significantly lower in Scn1a+/- neurons compared with wild-type neurons. There was no proportional increase in inhibitory postsynaptic current amplitude in response to increased extracellular Ca2+ concentrations. Our study revealed that the number of inhibitory synapses is significantly reduced in Scn1a+/- neurons, while the sensitivity of inhibitory synapses to extracellular Ca2+ concentrations is markedly increased. These data suggest that Ca2+ tethering in inhibitory nerve terminals may be disturbed following the synaptic burst, likely leading to epileptic symptoms.


Asunto(s)
Calcio/farmacología , Epilepsias Mioclónicas/fisiopatología , Espacio Extracelular/química , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Inhibición Neural/efectos de los fármacos , Transmisión Sináptica/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Marcación de Gen , Ratones Endogámicos ICR , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Transmisión Sináptica/efectos de los fármacos , Vesículas Sinápticas/efectos de los fármacos , Vesículas Sinápticas/metabolismo
19.
Int J Neuropsychopharmacol ; 24(7): 580-591, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-33693669

RESUMEN

BACKGROUND: Latent inhibition (LI) reflects an adaptive form of learning impaired in certain forms of mental illness. Glutamate receptor activity is linked to LI, but the potential role of synaptic plasticity remains unspecified. METHODS: Accordingly, the present study examined the possible role of long-term depression (LTD) in LI induced by prior exposure of rats to an auditory stimulus used subsequently as a conditional stimulus to signal a pending footshock. We employed 2 mechanistically distinct LTD inhibitors, the Tat-GluA23Y peptide that blocks endocytosis of the GluA2-containing glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor, or the selective glutamate n-methyl-d-aspartate receptor 2B antagonist, Ro25-6981, administered prior to the acquisition of 2-way conditioned avoidance with or without tone pre-exposure. RESULTS: Systemic LTD blockade with the Tat-GluA23Y peptide strengthened the LI effect by further impairing acquisition of conditioned avoidance in conditional stimulus-preexposed rats compared with normal conditioning in non-preexposed controls. Systemic Ro25-6981 had no significant effects. Brain region-specific microinjections of the Tat-GluA23Y peptide into the nucleus accumbens, medial prefrontal cortex, or central or basolateral amygdala demonstrated that disruption of glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor endocytosis in the central amygdala also potentiated the LI effect. CONCLUSIONS: These data revealed a previously unknown role for central amygdala LTD in LI as a key mediator of cognitive flexibility required to respond to previously irrelevant stimuli that acquire significance through reinforcement. The findings may have relevance both for our mechanistic understanding of LI and its alteration in disease states such as schizophrenia, while further elucidating the role of LTD in learning and memory.


Asunto(s)
Conducta Animal/fisiología , Péptidos de Penetración Celular/farmacología , Núcleo Amigdalino Central/fisiología , Antagonistas de Aminoácidos Excitadores/farmacología , Depresión Sináptica a Largo Plazo/fisiología , Inhibición Neural/fisiología , Animales , Percepción Auditiva/efectos de los fármacos , Percepción Auditiva/fisiología , Conducta Animal/efectos de los fármacos , Núcleo Amigdalino Central/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Clásico/fisiología , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Masculino , Inhibición Neural/efectos de los fármacos , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
20.
Br J Anaesth ; 126(6): 1141-1156, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33641936

RESUMEN

BACKGROUND: Both animal and retrospective human studies have linked extended and repeated general anaesthesia during early development with cognitive and behavioural deficits later in life. However, the neuronal circuit mechanisms underlying this anaesthesia-induced behavioural impairment are poorly understood. METHODS: Neonatal mice were administered one or three doses of propofol, a commonly used i.v. general anaesthetic, over Postnatal days 7-11. Control mice received Intralipid® vehicle injections. At 4 months of age, the mice were subjected to a series of behavioural tests, including motor learning. During the process of motor learning, calcium activity of pyramidal neurones and three classes of inhibitory interneurones in the primary motor cortex were examined in vivo using two-photon microscopy. RESULTS: Repeated, but not a single, exposure of neonatal mice to propofol i.p. caused motor learning impairment in adulthood, which was accompanied by a reduction of pyramidal neurone number and activity in the motor cortex. The activity of local inhibitory interneurone networks was also altered: somatostatin-expressing and parvalbumin-expressing interneurones were hypoactive, whereas vasoactive intestinal peptide-expressing interneurones were hyperactive when the mice were performing a motor learning task. Administration of low-dose pentylenetetrazol to attenuate γ-aminobutyric acid A receptor-mediated inhibition or CX546 to potentiate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-subtype glutamate receptor function during emergence from anaesthesia ameliorated neuronal dysfunction in the cortex and prevented long-term behavioural deficits. CONCLUSIONS: Repeated exposure of neonatal mice to propofol anaesthesia during early development causes cortical circuit dysfunction and behavioural impairments in later life. Potentiation of neuronal activity during recovery from anaesthesia reduces these adverse effects of early-life anaesthesia.


Asunto(s)
Anestésicos Intravenosos/toxicidad , Conducta Animal/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Corteza Motora/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Propofol/toxicidad , Animales , Animales Recién Nacidos , Señalización del Calcio/efectos de los fármacos , Prueba de Laberinto Elevado , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas del GABA/farmacología , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Ratones Transgénicos , Corteza Motora/metabolismo , Corteza Motora/fisiopatología , Inhibición Neural/efectos de los fármacos , Síndromes de Neurotoxicidad/fisiopatología , Síndromes de Neurotoxicidad/prevención & control , Síndromes de Neurotoxicidad/psicología , Prueba de Campo Abierto/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Conducta Social
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA