Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 13(7)2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38607012

RESUMO

Neuronal timing with millisecond precision is critical for many brain functions such as sensory perception, learning and memory formation. At the level of the chemical synapse, the synaptic delay is determined by the presynaptic release probability (Pr) and the waveform of the presynaptic action potential (AP). For instance, paired-pulse facilitation or presynaptic long-term potentiation are associated with reductions in the synaptic delay, whereas paired-pulse depression or presynaptic long-term depression are associated with an increased synaptic delay. Parallelly, the AP broadening that results from the inactivation of voltage gated potassium (Kv) channels responsible for the repolarization phase of the AP delays the synaptic response, and the inactivation of sodium (Nav) channels by voltage reduces the synaptic latency. However, whether synaptic delay is modulated during depolarization-induced analogue-digital facilitation (d-ADF), a form of context-dependent synaptic facilitation induced by prolonged depolarization of the presynaptic neuron and mediated by the voltage-inactivation of presynaptic Kv1 channels, remains unclear. We show here that despite Pr being elevated during d-ADF at pyramidal L5-L5 cell synapses, the synaptic delay is surprisingly unchanged. This finding suggests that both Pr- and AP-dependent changes in synaptic delay compensate for each other during d-ADF. We conclude that, in contrast to other short- or long-term modulations of presynaptic release, synaptic timing is not affected during d-ADF because of the opposite interaction of Pr- and AP-dependent modulations of synaptic delay.


Assuntos
Neurônios , Sinapses , Sinapses/fisiologia , Potenciais de Ação/fisiologia , Células Piramidais/fisiologia , Potenciação de Longa Duração
2.
J Neurosci ; 43(50): 8596-8606, 2023 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-37863654

RESUMO

Leucine-rich glioma inactivated 1 (LGI1) is a glycoprotein secreted by neurons, the deletion of which leads to autosomal dominant lateral temporal lobe epilepsy. We previously showed that LGI1 deficiency in a mouse model (i.e., knock-out for LGI1 or KO-Lgi1) decreased Kv1.1 channel density at the axon initial segment (AIS) and at presynaptic terminals, thus enhancing both intrinsic excitability and glutamate release. However, it is not known whether normal excitability can be restored in epileptic neurons. Here, we show that the selective expression of LGI1 in KO-Lgi1 neurons from mice of both sexes, using single-cell electroporation, reduces intrinsic excitability and restores both the Kv1.1-mediated D-type current and Kv1.1 channels at the AIS. In addition, we show that the homeostatic-like shortening of the AIS length observed in KO-Lgi1 neurons is prevented in neurons electroporated with the Lgi1 gene. Furthermore, we reveal a spatial gradient of intrinsic excitability that is centered on the electroporated neuron. We conclude that expression of LGI1 restores normal excitability through functional Kv1 channels at the AIS.SIGNIFICANCE STATEMENT The lack of leucine-rich glioma inactivated 1 (LGI1) protein induces severe epileptic seizures that leads to death. Enhanced intrinsic and synaptic excitation in KO-Lgi1 mice is because of the decrease in Kv1.1 channels in CA3 neurons. However, the conditions to restore normal excitability profile in epileptic neurons remain to be defined. We show here that the expression of LGI1 in KO-Lgi1 neurons in single neurons reduces intrinsic excitability, and restores both the Kv1.1-mediated D-type current and Kv1.1 channels at the axon initial segment (AIS). Furthermore, the homeostatic shortening of the AIS length observed in KO-Lgi1 neurons is prevented in neurons in which the Lgi1 gene has been rescued. We conclude that LGI1 constitutes a critical factor to restore normal excitability in epileptic neurons.


Assuntos
Epilepsia do Lobo Frontal , Glioma , Neurônios , Animais , Feminino , Masculino , Camundongos , Epilepsia do Lobo Frontal/genética , Epilepsia do Lobo Frontal/metabolismo , Leucina/metabolismo , Neurônios/fisiologia , Convulsões/genética
3.
Heliyon ; 9(6): e16953, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37484233

RESUMO

Channel noise results from rapid transitions of protein channels from closed to open state and is generally considered as the most dominant source of electrical noise causing membrane-potential fluctuations even in the absence of synaptic inputs. The simulation of a realistic channel noise remains a source of possible error. Although the Markovian method is considered as the golden standard for appropriate description of channel noise, its computation time increasing exponentially with the number of channels, it is poorly suitable to simulate realistic features. We describe here a novel algorithm at discrete time unit for simulating ion channel noise based on Markov chains (MC). Although this new algorithm refers to a Monte-Carlo process, it only needs few random numbers whatever the number of channels involved. Our fast MC (FMC) model does not exhibit the drawbacks due to approximations based on stochastic differential equations and the values of spike jitter are comparable to those obtained with the true Markovian method. In fact, we show here, that these drawbacks can be highlighted in the approximation based on stochastic differential equation methods even for a high number of channels (standard deviation of the 5th spike is about two-fold larger than that of MCF or true Markovian method for 5000 sodium channels). The FMC model appears therefore as the most accurate method to simulate channel noise with a fast execution time that does not depend on the channel number.

4.
Curr Opin Neurobiol ; 80: 102707, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36924615

RESUMO

Spike timing-dependent plasticity (STDP) is a bidirectional form of synaptic plasticity discovered about 30 years ago and based on the relative timing of pre- and post-synaptic spiking activity with a millisecond precision. STDP is thought to be involved in the formation of memory but the millisecond-precision spike-timing required for STDP is difficult to reconcile with the much slower timescales of behavioral learning. This review therefore aims to expose and discuss recent findings about i) the multiple STDP learning rules at both excitatory and inhibitory synapses in vitro, ii) the contribution of STDP-like synaptic plasticity in the formation of memory in vivo and iii) the implementation of STDP rules in artificial neural networks and memristive devices.


Assuntos
Plasticidade Neuronal , Neurônios , Redes Neurais de Computação , Sinapses , Aprendizagem , Potenciais de Ação
5.
Proc Natl Acad Sci U S A ; 119(44): e2205264119, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36282913

RESUMO

Brain oscillations have long-lasting effects on synaptic and cellular properties. For instance, synaptic stimulation at theta (θ) frequency induces persistent depression of both excitatory synaptic transmission and intrinsic excitability in CA1 principal neurons. However, the incidence of θ activity on synaptic transmission and intrinsic excitability in hippocampal GABAergic interneurons is unclear. We report here the induction of both synaptic and intrinsic potentiation in oriens-lacunosum moleculare (O-LM) interneurons following stimulation of afferent glutamatergic inputs in the θ frequency range (∼5 Hz). Long-term synaptic potentiation (LTP) is induced by synaptic activation of calcium-permeable AMPA receptors (CP-AMPAR), whereas long-term potentiation of intrinsic excitability (LTP-IE) results from the mGluR1-dependent down-regulation of Kv7 voltage-dependent potassium channel and hyperpolarization activated and cyclic nucleotide-gated (HCN) channel through the depletion of phosphatidylinositol-4,5-biphosphate (PIP2). LTP and LTP-IE are reversible, demonstrating that both synaptic and intrinsic changes are bidirectional in O-LM cells. We conclude that synaptic activity at θ frequency induces both synaptic and intrinsic potentiation in O-LM interneurons, i.e., the opposite of what is typically seen in glutamatergic neurons.


Assuntos
Cálcio , Receptores de AMPA , Receptores de AMPA/metabolismo , Cálcio/metabolismo , Sinapses/metabolismo , Fosfatidilinositol 4,5-Difosfato , Hipocampo/metabolismo , Interneurônios/metabolismo , Potenciação de Longa Duração/fisiologia , Canais de Potássio , Nucleotídeos Cíclicos/farmacologia , Estimulação Elétrica
6.
Cells ; 11(17)2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-36078121

RESUMO

Leucine-rich Glioma-Inactivated protein 1 (LGI1) is expressed in the central nervous system and its genetic loss of function is associated with epileptic disorders. Additionally, patients with LGI1-directed autoantibodies have frequent focal seizures as a key feature of their disease. LGI1 is composed of a Leucine-Rich Repeat (LRR) and an Epitempin (EPTP) domain. These domains are reported to interact with different members of the transsynaptic complex formed by LGI1 at excitatory synapses, including presynaptic Kv1 potassium channels. Patient-derived recombinant monoclonal antibodies (mAbs) are ideal reagents to study whether domain-specific LGI1-autoantibodies induce epileptiform activities in neurons and their downstream mechanisms. We measured the intrinsic excitability of CA3 pyramidal neurons in organotypic cultures from rat hippocampus treated with either an LRR- or an EPTP-reactive patient-derived mAb, or with IgG from control patients. We found an increase in intrinsic excitability correlated with a reduction of the sensitivity to a selective Kv1.1-channel blocker in neurons treated with the LRR mAb, but not in neurons treated with the EPTP mAb. Our findings suggest LRR mAbs are able to modulate neuronal excitability that could account for epileptiform activity observed in patients.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Proteínas do Tecido Nervoso , Animais , Autoanticorpos/metabolismo , Epitopos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Leucina , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Ratos
7.
Brain ; 145(11): 3843-3858, 2022 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-35727946

RESUMO

Autoantibodies against leucine-rich glioma-inactivated 1 (LGI1) occur in patients with encephalitis who present with frequent focal seizures and a pattern of amnesia consistent with focal hippocampal damage. To investigate whether the cellular and subcellular distribution of LGI1 may explain the localization of these features, and hence gain broader insights into LGI1's neurobiology, we analysed the detailed localization of LGI1 and the diversity of its protein interactome, in mouse brains using patient-derived recombinant monoclonal LGI1 antibodies. Combined immunofluorescence and mass spectrometry analyses showed that LGI1 is enriched in excitatory and inhibitory synaptic contact sites, most densely within CA3 regions of the hippocampus. LGI1 is secreted in both neuronal somatodendritic and axonal compartments, and occurs in oligodendrocytic, neuro-oligodendrocytic and astro-microglial protein complexes. Proteomic data support the presence of LGI1-Kv1-MAGUK complexes, but did not reveal LGI1 complexes with postsynaptic glutamate receptors. Our results extend our understanding of regional, cellular and subcellular LGI1 expression profiles and reveal novel LGI1-associated complexes, thus providing insights into the complex biology of LGI1 and its relationship to seizures and memory loss.


Assuntos
Glioma , Peptídeos e Proteínas de Sinalização Intracelular , Animais , Camundongos , Leucina , Proteômica , Autoanticorpos , Convulsões
8.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34799447

RESUMO

Homeostatic plasticity of intrinsic excitability goes hand in hand with homeostatic plasticity of synaptic transmission. However, the mechanisms linking the two forms of homeostatic regulation have not been identified so far. Using electrophysiological, imaging, and immunohistochemical techniques, we show here that blockade of excitatory synaptic receptors for 2 to 3 d induces an up-regulation of both synaptic transmission at CA3-CA3 connections and intrinsic excitability of CA3 pyramidal neurons. Intrinsic plasticity was found to be mediated by a reduction of Kv1.1 channel density at the axon initial segment. In activity-deprived circuits, CA3-CA3 synapses were found to express a high release probability, an insensitivity to dendrotoxin, and a lack of depolarization-induced presynaptic facilitation, indicating a reduction in presynaptic Kv1.1 function. Further support for the down-regulation of axonal Kv1.1 channels in activity-deprived neurons was the broadening of action potentials measured in the axon. We conclude that regulation of the axonal Kv1.1 channel constitutes a major mechanism linking intrinsic excitability and synaptic strength that accounts for the functional synergy existing between homeostatic regulation of intrinsic excitability and synaptic transmission.


Assuntos
Axônios/metabolismo , Hipocampo/metabolismo , Homeostase , Potenciais de Ação/fisiologia , Animais , Plasticidade Neuronal , Neurônios/metabolismo , Células Piramidais/metabolismo , Ratos , Ratos Wistar , Sinapses/metabolismo , Transmissão Sináptica/fisiologia
9.
Cells ; 10(11)2021 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-34831385

RESUMO

Visual plasticity is classically considered to occur essentially in the primary and secondary cortical areas. Subcortical visual areas such as the dorsal lateral geniculate nucleus (dLGN) or the superior colliculus (SC) have long been held as basic structures responsible for a stable and defined function. In this model, the dLGN was considered as a relay of visual information travelling from the retina to cortical areas and the SC as a sensory integrator orienting body movements towards visual targets. However, recent findings suggest that both dLGN and SC neurons express functional plasticity, adding unexplored layers of complexity to their previously attributed functions. The existence of neuronal plasticity at the level of visual subcortical areas redefines our approach of the visual system. The aim of this paper is therefore to review the cellular and molecular mechanisms for activity-dependent plasticity of both synaptic transmission and cellular properties in subcortical visual areas.


Assuntos
Córtex Cerebral/fisiologia , Plasticidade Neuronal/fisiologia , Vias Visuais/fisiologia , Animais , Corpos Geniculados/fisiologia , Humanos , Plasticidade Neuronal/genética , Retina/fisiologia , Sinapses/fisiologia
10.
J Neurosci ; 41(46): 9521-9538, 2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34620719

RESUMO

KCNQ-Kv7 channels are found at the axon initial segment of pyramidal neurons, where they control cell firing and membrane potential. In oriens lacunosum moleculare (O-LM) interneurons, these channels are mainly expressed in the dendrites, suggesting a peculiar function of Kv7 channels in these neurons. Here, we show that Kv7 channel activity is upregulated following induction of presynaptic long-term synaptic depression (LTD) in O-LM interneurons from rats of both sex, thus resulting in a synergistic long-term depression of intrinsic excitability (LTD-IE). Both LTD and LTD-IE involve endocannabinoid (eCB) biosynthesis for induction. However, although LTD is dependent on cannabinoid type 1 receptors, LTD-IE is not. Molecular modeling shows a strong interaction of eCBs with Kv7.2/3 channel, suggesting a persistent action of these lipids on Kv7 channel activity. Our data thus unveil a major role for eCB synthesis in triggering both synaptic and intrinsic depression in O-LM interneurons.SIGNIFICANCE STATEMENT In principal cells, Kv7 channels are essentially located at the axon initial segment. In contrast, in O-LM interneurons, Kv7 channels are highly expressed in the dendrites, suggesting a singular role of these channels in O-LM cell function. Here, we show that LTD of excitatory inputs in O-LM interneurons is associated with an upregulation of Kv7 channels, thus resulting in a synergistic LTD of LTD-IE. Both forms of plasticity are mediated by the biosynthesis of eCBs. Stimulation of CB1 receptors induces LTD, whereas the direct interaction of eCBs with Kv7 channels induces LTD-IE. Our results thus provide a previously unexpected involvement of eCBs in long-lasting plasticity of intrinsic excitability in GABAergic interneurons.


Assuntos
Endocanabinoides/metabolismo , Interneurônios/metabolismo , Canais de Potássio KCNQ/metabolismo , Depressão Sináptica de Longo Prazo/fisiologia , Animais , Feminino , Hipocampo/metabolismo , Masculino , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar
11.
Front Cell Neurosci ; 15: 727336, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34616278

RESUMO

Since its discovery, spike timing-dependent synaptic plasticity (STDP) has been thought to be a primary mechanism underlying the brain's ability to learn and to form new memories. However, despite the enormous interest in both the experimental and theoretical neuroscience communities in activity-dependent plasticity, it is still unclear whether plasticity rules inferred from in vitro experiments apply to in vivo conditions. Among the multiple reasons why plasticity rules in vivo might differ significantly from in vitro studies is that extracellular calcium concentration use in most studies is higher than concentrations estimated in vivo. STDP, like many forms of long-term synaptic plasticity, strongly depends on intracellular calcium influx for its induction. Here, we discuss the importance of considering physiological levels of extracellular calcium concentration to study functional plasticity.

12.
Mol Neurobiol ; 58(12): 6153-6169, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34458961

RESUMO

The axon initial segment (AIS) is essential for maintaining neuronal polarity, modulating protein transport into the axon, and action potential generation. These functions are supported by a distinctive actin and microtubule cytoskeleton that controls axonal trafficking and maintains a high density of voltage-gated ion channels linked by scaffold proteins to the AIS cytoskeleton. However, our knowledge of the mechanisms and proteins involved in AIS cytoskeleton regulation to maintain or modulate AIS structure is limited. In this context, formins play a significant role in the modulation of actin and microtubules. We show that pharmacological inhibition of formins modifies AIS actin and microtubule characteristics in cultured hippocampal neurons, reducing F-actin density and decreasing microtubule acetylation. Moreover, formin inhibition diminishes sodium channels, ankyrinG and ßIV-spectrin AIS density, and AIS length, in cultured neurons and brain slices, accompanied by decreased neuronal excitability. We show that genetic downregulation of the mDia1 formin by interference RNAs also decreases AIS protein density and shortens AIS length. The ankyrinG decrease and AIS shortening observed in pharmacologically inhibited neurons and neuron-expressing mDia1 shRNAs were impaired by HDAC6 downregulation or EB1-GFP expression, known to increase microtubule acetylation or stability. However, actin stabilization only partially prevented AIS shortening without affecting AIS protein density loss. These results suggest that mDia1 maintain AIS composition and length contributing to the stability of AIS microtubules.


Assuntos
Segmento Inicial do Axônio/metabolismo , Citoesqueleto/metabolismo , Forminas/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Animais , Axônios/metabolismo , Células Cultivadas , Camundongos , Microtúbulos/metabolismo
13.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34389672

RESUMO

The position of the axon initial segment (AIS) is thought to play a critical role in neuronal excitability. Previous experimental studies have found that a distal shift in AIS position correlates with a reduction in excitability. Yet theoretical work has suggested the opposite, because of increased electrical isolation. A distal shift in AIS position corresponds to an elevation of axial resistance Ra We therefore examined how changes in Ra at the axon hillock impact the voltage threshold (Vth) of the somatic action potential in L5 pyramidal neurons. Increasing Ra by mechanically pinching the axon between the soma and the AIS was found to lower Vth by ∼6 mV. Conversely, decreasing Ra by substituting internal ions with higher mobility elevated Vth All Ra -dependent changes in Vth could be reproduced in a Hodgkin-Huxley compartmental model. We conclude that in L5 pyramidal neurons, excitability increases with axial resistance and therefore with a distal shift of the AIS.


Assuntos
Potenciais de Ação/fisiologia , Axônios/fisiologia , Células Piramidais/fisiologia , Animais , Simulação por Computador , Microscopia Confocal , Modelos Biológicos , Ratos , Ratos Wistar
14.
Proc Natl Acad Sci U S A ; 117(52): 33639-33648, 2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33328274

RESUMO

Spike-timing-dependent plasticity (STDP) is considered as a primary mechanism underlying formation of new memories during learning. Despite the growing interest in activity-dependent plasticity, it is still unclear whether synaptic plasticity rules inferred from in vitro experiments are correct in physiological conditions. The abnormally high calcium concentration used in in vitro studies of STDP suggests that in vivo plasticity rules may differ significantly from in vitro experiments, especially since STDP depends strongly on calcium for induction. We therefore studied here the influence of extracellular calcium on synaptic plasticity. Using a combination of experimental (patch-clamp recording and Ca2+ imaging at CA3-CA1 synapses) and theoretical approaches, we show here that the classic STDP rule in which pairs of single pre- and postsynaptic action potentials induce synaptic modifications is not valid in the physiological Ca2+ range. Rather, we found that these pairs of single stimuli are unable to induce any synaptic modification in 1.3 and 1.5 mM calcium and lead to depression in 1.8 mM. Plasticity can only be recovered when bursts of postsynaptic spikes are used, or when neurons fire at sufficiently high frequency. In conclusion, the STDP rule is profoundly altered in physiological Ca2+, but specific activity regimes restore a classical STDP profile.


Assuntos
Cálcio/metabolismo , Plasticidade Neuronal/fisiologia , Potenciais de Ação/fisiologia , Animais , Potenciação de Longa Duração , Modelos Neurológicos , Dinâmica não Linear , Ratos Wistar , Fatores de Tempo
16.
Front Cell Neurosci ; 14: 99, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32477066

RESUMO

[This corrects the article DOI: 10.3389/fncel.2020.00040.].

17.
Sci Adv ; 6(19): eaay4313, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32494697

RESUMO

Sensory processing requires mechanisms of fast coincidence detection to discriminate synchronous from asynchronous inputs. Spike threshold adaptation enables such a discrimination but is ineffective in transmitting this information to the network. We show here that presynaptic axonal sodium channels read and transmit precise levels of input synchrony to the postsynaptic cell by modulating the presynaptic action potential (AP) amplitude. As a consequence, synaptic transmission is facilitated at cortical synapses when the presynaptic spike is produced by synchronous inputs. Using dual soma-axon recordings, imaging, and modeling, we show that this facilitation results from enhanced AP amplitude in the axon due to minimized inactivation of axonal sodium channels. Quantifying local circuit activity and using network modeling, we found that spikes induced by synchronous inputs produced a larger effect on network activity than spikes induced by asynchronous inputs. Therefore, this input synchrony-dependent facilitation may constitute a powerful mechanism, regulating synaptic transmission at proximal synapses.

18.
Front Cell Neurosci ; 14: 40, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194377

RESUMO

Analog-digital facilitations (ADFs) have been described in local excitatory brain circuits and correspond to a class of phenomena describing how subthreshold variations of the presynaptic membrane potential influence spike-evoked synaptic transmission. In many brain circuits, ADFs rely on the propagation of somatic membrane potential fluctuations to the presynaptic bouton where they modulate ion channels availability, inducing modifications of the presynaptic spike waveform, the spike-evoked Ca2+ entry, and the transmitter release. Therefore, one major requirement for ADFs to occur is the propagation of subthreshold membrane potential variations from the soma to the presynaptic bouton. To date, reported ADFs space constants are relatively short (250-500 µm) which limits their action to proximal synapses. However, ADFs have been studied either in unmyelinated axons or in juvenile animals in which myelination is incomplete. We examined here the potential gain of ADFs spatial extent caused by myelination using a realistic model of L5 pyramidal cell. Myelination of the axon was found to induce a 3-fold increase in the axonal length constant. As a result, the different forms of ADF were found to display a much longer spatial extent (up to 3,000 µm). In addition, while the internodal length displayed a mild effect, the number of myelin wraps ensheathing the internodes was found to play a critical role in the ADFs spatial extents. We conclude that axonal myelination induces an increase in ADFs spatial extent in our model, thus making ADFs plausible in long-distance connections.

19.
Neurobiol Learn Mem ; 166: 107095, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31539624

RESUMO

Persistent changes that occur in brain circuits are classically thought to be mediated by long-term modifications in synaptic efficacy. Yet, many studies have shown that voltage-gated ion channels located at the input and output side of the neurons are also the subject to persistent modifications. These channels are thus responsible for intrinsic plasticity that is expressed in many different neuronal types including glutamatergic principal neurons and GABAergic interneurons. As for synaptic plasticity, activation of synaptic glutamate receptors initiate persistent modification in neuronal excitability. We review here how synaptic input can be efficiently altered by activity-dependent modulation of ion channels that control EPSP amplification, spike threshold or resting membrane potential. We discuss the nature of the learning rules shared by intrinsic and synaptic plasticity, the mechanisms of ion channel regulation and the impact of intrinsic plasticity on induction of synaptic modifications.


Assuntos
Encéfalo/fisiologia , Canais Iônicos/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Transmissão Sináptica/fisiologia , Potenciais de Ação/fisiologia , Animais , Aprendizagem/fisiologia , Memória/fisiologia
20.
J Pharmacol Toxicol Methods ; 99: 106587, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31207287

RESUMO

INTRODUCTION: Failures in drug development often result from the emergence of unexpected adverse drug reactions. It is clear that adverse drug reactions, including seizure liability, should be assessed earlier. The goal of the present work was to develop a new platform of in vitro assays, NS-PC set (for Neuroservice proconvulsive set), to determine the proconvulsive potential of compounds earlier in preclinical development. METHODS: Assays were based on electrophysiological recordings in acute hippocampal slices performed with multielectrode arrays. 4 reference proconvulsive/seizurogenic compounds (4-aminopyridine, bicuculline, kainate and carbachol) and 4 anti-epileptic drugs (AEDs; phenobarbital, carbamazepine, clonazepam and valproic acid) were evaluated on electrophysiological endpoints involved in seizure risk (neuronal excitability, balance of excitatory/inhibitory synaptic transmission, occurrence of neuronal synchronization mechanisms materialized by epileptiform discharges). RESULTS: The reference compounds increased the number and area under the curve of population spikes, triggered epileptiform discharges and enhanced the firing rate of CA1 neurons. The effects of the 4 antiepileptic drugs were assessed on these 3 parameters. They were able to partially of completely reverse the effects of proconvulsive compounds. DISCUSSION: The use of reference proconvulsive compounds and AEDs validated the electrophysiological parameters to detect proconvulsive risk. Systematic evaluation of compounds with the 3 complementary endpoints increase the probability to detect seizure liability in vitro. Depending on the compound mechanism of action, only one or two of the identified parameters might be modified.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...