Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
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.

3.
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
4.
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
5.
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
6.
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
7.
Sci Rep ; 7(1): 14418, 2017 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-29089586

RESUMO

The polarity of excitability changes associated with induction of Long-Term synaptic Depression (LTD) in CA1 pyramidal neurons is a contentious issue. Postsynaptic neuronal excitability after LTD induction is found to be reduced in certain cases (i.e. synergistic changes) but enhanced in others (i.e. compensatory or homeostatic). We examined here whether these divergent findings could result from the activation of two separate mechanisms converging onto a single learning rule linking synergistic and homeostatic plasticity. We show that the magnitude of LTD induced with low frequency stimulation (LFS) of the Schaffer collaterals determines the polarity of intrinsic changes in CA1 pyramidal neurons. Apparent input resistance (Rin) is reduced following induction of moderate LTD (<20-30%). In contrast, Rin is increased after induction of large LTD (>40%) induced by repetitive episodes of LFS. The up-regulation of I h observed after moderate LTD results from the activation of NMDA receptors whereas the down-regulation of I h is due to activation of mGluR1 receptors. These changes in Rin were associated with changes in intrinsic excitability. In conclusion, our study indicates that changes in excitability after LTD induction follow a learning rule describing a continuum linking synergistic and compensatory changes in excitability.


Assuntos
Potenciação de Longa Duração/fisiologia , Depressão Sináptica de Longo Prazo/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Região CA1 Hipocampal/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/fisiologia , Células Piramidais/fisiologia , Ratos , Receptores de Glutamato Metabotrópico , Receptores de N-Metil-D-Aspartato , Transmissão Sináptica/fisiologia
8.
Nat Commun ; 6: 10163, 2015 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-26657943

RESUMO

In the mammalian brain, synaptic transmission usually depends on presynaptic action potentials (APs) in an all-or-none (or digital) manner. Recent studies suggest, however, that subthreshold depolarization in the presynaptic cell facilitates spike-evoked transmission, thus creating an analogue modulation of a digital process (or analogue-digital (AD) modulation). At most synapses, this process is slow and not ideally suited for the fast dynamics of neural networks. We show here that transmission at CA3-CA3 and L5-L5 synapses can be enhanced by brief presynaptic hyperpolarization such as an inhibitory postsynaptic potential (IPSP). Using dual soma-axon patch recordings and live imaging, we find that this hyperpolarization-induced AD facilitation (h-ADF) is due to the recovery from inactivation of Nav channels controlling AP amplitude in the axon. Incorporated in a network model, h-ADF promotes both pyramidal cell synchrony and gamma oscillations. In conclusion, cortical excitatory synapses in local circuits display hyperpolarization-induced facilitation of spike-evoked synaptic transmission that promotes network synchrony.


Assuntos
Axônios/fisiologia , Potenciais da Membrana/fisiologia , Terminações Pré-Sinápticas , Canais de Sódio/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Encéfalo/fisiologia , Cálcio/metabolismo , Simulação por Computador , Feminino , Masculino , Modelos Biológicos , Rede Nervosa/fisiologia , Neurônios , Ratos
9.
Eur J Neurosci ; 41(3): 293-304, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25394682

RESUMO

Synaptic transmission usually depends on action potentials (APs) in an all-or-none (digital) fashion. Recent studies indicate, however, that subthreshold presynaptic depolarization may facilitate spike-evoked transmission, thus creating an analog modulation of spike-evoked synaptic transmission, also called analog-digital (AD) synaptic facilitation. Yet, the underlying mechanisms behind this facilitation remain unclear. We show here that AD facilitation at rat CA3-CA3 synapses is time-dependent and requires long presynaptic depolarization (5-10 s) for its induction. This depolarization-induced AD facilitation (d-ADF) is blocked by the specific Kv1.1 channel blocker dendrotoxin-K. Using fast voltage-imaging of the axon, we show that somatic depolarization used for induction of d-ADF broadened the AP in the axon through inactivation of Kv1.1 channels. Somatic depolarization enhanced spike-evoked calcium signals in presynaptic terminals, but not basal calcium. In conclusion, axonal Kv1.1 channels determine glutamate release in CA3 neurons in a time-dependent manner through the control of the presynaptic spike waveform.


Assuntos
Potenciais de Ação/fisiologia , Região CA3 Hipocampal/fisiologia , Canal de Potássio Kv1.1/metabolismo , Transmissão Sináptica/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Região CA3 Hipocampal/efeitos dos fármacos , Cálcio/metabolismo , Quelantes de Cálcio/farmacologia , Ácido Egtázico/farmacologia , Ácido Glutâmico/metabolismo , Canal de Potássio Kv1.1/antagonistas & inibidores , Modelos Neurológicos , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Ratos Wistar , Sódio/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Tempo , Técnicas de Cultura de Tecidos
10.
Neuron ; 77(4): 712-22, 2013 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-23439123

RESUMO

The dynamics of inhibitory circuits in the cortex is thought to rely mainly on synaptic modifications. We challenge this view by showing that hippocampal parvalbumin-positive basket cells (PV-BCs) of the CA1 region express long-term (>30 min) potentiation of intrinsic neuronal excitability (LTP-IE(PV-BC)) upon brief repetitive stimulation of the Schaffer collaterals. LTP-IE(PV-BC) is induced by synaptic activation of metabotropic glutamate receptor subtype 5 (mGluR5) and mediated by the downregulation of Kv1 channel activity. LTP-IE(PV-BC) promotes spiking activity at the gamma frequency (∼35 Hz) and facilitates recruitment of PV-BCs to balance synaptic and intrinsic excitation in pyramidal neurons. In conclusion, activity-dependent modulation of intrinsic neuronal excitability in PV-BCs maintains excitatory-inhibitory balance and thus plays a major role in the dynamics of hippocampal circuits.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Rede Nervosa/fisiologia , Neurônios/fisiologia , Potenciais de Ação/fisiologia , Animais , Animais Recém-Nascidos , Hipocampo/citologia , Parvalbuminas/metabolismo , Células Piramidais/fisiologia , Ratos , Ratos Wistar , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/fisiologia
11.
ACS Nano ; 5(10): 8080-8, 2011 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-21932787

RESUMO

We report on parallel high-resolution electrical single-molecule analysis on a chip-based nanopore microarray. Lipid bilayers of <20 µm diameter containing single alpha-hemolysin pores were formed on arrays of subpicoliter cavities containing individual microelectrodes (microelectrode cavity array, MECA), and ion conductance-based single molecule mass spectrometry was performed on mixtures of poly(ethylene glycol) molecules of different length. We thereby demonstrate the function of the MECA device as a chip-based platform for array-format nanopore recordings with a resolution at least equal to that of established single microbilayer supports. We conclude that devices based on MECAs may enable more widespread analytical use of nanopores by providing the high throughput and ease of operation of a high-density array format while maintaining or exceeding the precision of state-of-the-art microbilayer recordings.


Assuntos
Membrana Celular/química , Bicamadas Lipídicas/química , Espectrometria de Massas/instrumentação , Análise em Microsséries/instrumentação , Nanoporos , Condutividade Elétrica , Proteínas Hemolisinas/química , Microeletrodos , Polietilenoglicóis/química
12.
J Physiol ; 589(Pt 15): 3753-73, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21624967

RESUMO

Hyperpolarization-activated cyclic nucleotide modulated current (I(h)) sets resonance frequency within the θ-range (5­12 Hz) in pyramidal neurons. However, its precise contribution to the temporal fidelity of spike generation in response to stimulation of excitatory or inhibitory synapses remains unclear. In conditions where pharmacological blockade of I(h) does not affect synaptic transmission, we show that postsynaptic h-channels improve spike time precision in CA1 pyramidal neurons through two main mechanisms. I(h) enhances precision of excitatory postsynaptic potential (EPSP)--spike coupling because I(h) reduces peak EPSP duration. I(h) improves the precision of rebound spiking following inhibitory postsynaptic potentials (IPSPs) in CA1 pyramidal neurons and sets pacemaker activity in stratum oriens interneurons because I(h) accelerates the decay of both IPSPs and after-hyperpolarizing potentials (AHPs). The contribution of h-channels to intrinsic resonance and EPSP waveform was comparatively much smaller in CA3 pyramidal neurons. Our results indicate that the elementary mechanisms by which postsynaptic h-channels control fidelity of spike timing at the scale of individual neurons may account for the decreased theta-activity observed in hippocampal and neocortical networks when h-channel activity is pharmacologically reduced.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciais Pós-Sinápticos Inibidores/fisiologia , Neocórtex/fisiologia , Neurônios/fisiologia , Canais de Potássio/fisiologia , Células Piramidais/fisiologia , Animais , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Estimulação Elétrica/métodos , Eletrofisiologia/métodos , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Neocórtex/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Canais de Potássio/metabolismo , Células Piramidais/efeitos dos fármacos , Pirimidinas/farmacologia , Ratos , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
13.
J Neurosci ; 28(34): 8635-43, 2008 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-18716222

RESUMO

Hyperpolarization-activated (h)-channels occupy a central position in dendritic function. Although it has been demonstrated that these channels are upregulated after large depolarizations to reduce dendritic excitation, it is not clear whether they also support other forms of long-term plasticity. We show here that nearly maximal long-term potentiation (LTP) induced by theta-burst pairing produced upregulation in h-channel activity in CA1 pyramidal neurons. In contrast, moderate LTP induced by spike-timing-dependent plasticity or high-frequency stimulation (HFS) downregulated the h-current (I(h)) in the dendrites. After HFS-induced LTP, the h-conductance (G(h)) was reduced without changing its activation. Pharmacological blockade of I(h) had no effect on LTP induction, but occluded EPSP-to-spike potentiation, an input-specific facilitation of dendritic integration. Dynamic-clamp reduction of G(h) locally in the dendrite mimicked the effects of HFS and enhanced synaptic integration in an input-selective way. We conclude that dendritic I(h) is locally downregulated after induction of nonmaximal LTP, thus facilitating integration of the potentiated input.


Assuntos
Dendritos/fisiologia , Canais Iônicos/fisiologia , Potenciação de Longa Duração/fisiologia , Células Piramidais/fisiologia , Animais , Animais Recém-Nascidos , Regulação para Baixo , Estimulação Elétrica/métodos , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores , Técnicas In Vitro , Canais Iônicos/metabolismo , Modelos Neurológicos , Plasticidade Neuronal , Ratos , Sinapses/fisiologia , Ritmo Teta , Regulação para Cima
14.
Neuron ; 56(6): 1048-60, 2007 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-18093526

RESUMO

In the cortex, synaptic latencies display small variations ( approximately 1-2 ms) that are generally considered to be negligible. We show here that the synaptic latency at monosynaptically connected pairs of L5 and CA3 pyramidal neurons is determined by the presynaptic release probability (Pr): synaptic latency being inversely correlated with the amplitude of the postsynaptic current and sensitive to manipulations of Pr. Changes in synaptic latency were also observed when Pr was physiologically regulated in short- and long-term synaptic plasticity. Paired-pulse depression and facilitation were respectively associated with increased and decreased synaptic latencies. Similarly, latencies were prolonged following induction of presynaptic LTD and reduced after LTP induction. We show using the dynamic-clamp technique that the observed covariation in latency and synaptic strength is a synergistic combination that significantly affects postsynaptic spiking. In conclusion, amplitude-related variation in latency represents a putative code for short- and long-term synaptic dynamics in cortical networks.


Assuntos
Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Dinâmica não Linear , Tempo de Reação/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Animais , Animais Recém-Nascidos , Córtex Cerebral/citologia , Simulação por Computador , Relação Dose-Resposta à Radiação , Estimulação Elétrica , Antagonistas de Aminoácidos Excitatórios/farmacologia , Técnicas In Vitro , Masculino , Modelos Neurológicos , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/efeitos da radiação , Técnicas de Patch-Clamp , Quinoxalinas/farmacologia , Ratos , Ratos Wistar , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/efeitos da radiação , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/efeitos da radiação
15.
J Physiol ; 577(Pt 1): 141-54, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16931548

RESUMO

Brain sodium channels (NaChs) are regulated by various neurotransmitters such as acetylcholine, serotonin and dopamine. However, it is not known whether NaCh activity is regulated by glutamate, the principal brain neurotransmitter. We show here that activation of metabotropic glutamate receptor (mGluR) subtype 1 regulates fast transient (I(NaT)) and persistent Na(+) currents (I(NaP)) in cortical pyramidal neurons. A selective agonist of group I mGluR, (S)-3,5-dihydroxyphenylglycine (DHPG), reduced action potential amplitude and decreased I(NaT). This reduction was blocked when DHPG was applied in the presence of selective mGluR1 antagonists. The DHPG-induced reduction of the current was accompanied by a shift of both the inactivation curve of I(NaT) and the activation curve of I(NaP). These effects were dependent on the activation of PKC. The respective role of these two regulatory processes on neuronal excitability was determined by simulating transient and persistent Na(+) conductances (G(NaT) and G(NaP)) with fast dynamic-clamp techniques. The facilitated activation of G(NaP) increased excitability near the threshold, but, when combined with the down-regulation of G(NaT), repetitive firing was strongly decreased. Consistent with this finding, the mGluR1 antagonist LY367385 increased neuronal excitability when glutamatergic synaptic activity was stimulated with high external K(+). We conclude that mGluR1-dependent regulation of Na(+) current depresses neuronal excitability, which thus might constitute a novel mechanism of homeostatic regulation acting during intense glutamatergic synaptic activity.


Assuntos
Ativação do Canal Iônico/fisiologia , Neocórtex/fisiologia , Células Piramidais/fisiologia , Receptores de Glutamato Metabotrópico/metabolismo , Canais de Sódio/fisiologia , Sódio/metabolismo , Potenciais de Ação/fisiologia , Animais , Células Cultivadas , Ratos , Ratos Wistar , Tempo de Reação/fisiologia
16.
J Neurosci ; 23(32): 10238-48, 2003 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-14614082

RESUMO

The cellular substrate for memory is generally attributed to long-lasting changes in synaptic strength. We report here that synaptic or pharmacological activation of the metabotropic glutamate receptor subtype 5 (mGluR5) induces long-term potentiation of intrinsic excitability (LTP-IE) in layer V pyramidal neurons. mGluR5-dependent LTP-IE was associated with a persistent reduction of the afterhyperpolarization (AHP) outward current (IAHP), resulting in the potentiation of EPSP-spike coupling. Apamin occluded induction of LTP-IE, indicating that downregulation of small conductance calcium-dependent potassium (SK) channels mediates this process. In addition to the improved reliability of the input-output function, LTP-IE led to increased temporal precision. The induced reduction of IAHP accelerated the rate of membrane depolarization preceding each action potential and subsequently decreased the jitter of the neuronal discharge. We conclude that mGluR5-dependent LTP-IE not only promotes the spread of excitation in the cortical network but also persistently enhances the temporal fidelity of the neuronal message.


Assuntos
Potenciação de Longa Duração/fisiologia , Neurônios/fisiologia , Receptores de Glutamato Metabotrópico/metabolismo , Potenciais de Ação/fisiologia , Animais , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Técnicas In Vitro , Neocórtex/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Ratos , Tempo de Reação/fisiologia , Receptor de Glutamato Metabotrópico 5 , Reprodutibilidade dos Testes , Sinapses/fisiologia
17.
J Physiol ; 549(Pt 1): 21-36, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12651919

RESUMO

During postnatal development, profound changes take place in the excitability of nerve cells, including modification in the distribution and properties of receptor-operated channels and changes in the density and nature of voltage-gated channels. We studied here the firing properties of abducens motoneurons (aMns) in transverse brainstem slices from postnatal day (P) 1-13 rats. Recordings were made from aMNs in the whole-cell configuration of the patch-clamp technique. Two main types of aMn could be distinguished according to their firing profile during prolonged depolarizations. Both types were identified as aMns by their fluorescence following retrograde labelling with the lipophilic carbocyanine DiI in the rectus lateralis muscle. The first type (BaMns) exhibited a burst of action potentials (APs) followed by an adaptation of discharge and were encountered in approximately 70 % of aMns. Their discharge profile resembled that of adult aMns and was encountered in all aMns after P9. BaMns exhibited a hyperpolarization-induced rebound potential that was blocked by low concentrations of Ni2+ or by Ca2+-free external solution. This current had the properties of the T-type current. Action potentials of BaMns showed a complex afterhyperpolarization (AHP). An inward rectification was evidenced following hyperpolarization and was blocked by external application of caesium or ZD7288, indicating the presence of the hyperpolarization-activated cationic current (IH). Blocking the IH current almost doubled the input resistance of BaMns. The second class of aMns (DaMns) displayed a delayed excitation that was mediated by A-type K+ currents and was observed only between P4 and P9. DaMns exhibited immature characteristics: an action potential with a simple AHP, a linear current-voltage relation and a large input resistance. The number of aMns remained unchanged when both types were present (P5-P6) and later in development when only BaMns were encountered (P19), suggesting that DaMns mature into BaMns during postnatal development. We conclude that aMns display profound reorganization in their intrinsic excitability during postnatal development.


Assuntos
Nervo Abducente/crescimento & desenvolvimento , Nervo Abducente/fisiologia , Canais de Cálcio Tipo T/fisiologia , Neurônios Motores/fisiologia , Canais de Potássio/fisiologia , Nervo Abducente/citologia , Potenciais de Ação/fisiologia , Animais , Cátions/metabolismo , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Ratos , Ratos Wistar
18.
J Physiol ; 541(Pt 1): 123-37, 2002 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12015425

RESUMO

Whole-cell patch clamp recordings of miniature inhibitory postsynaptic currents (mIPSCs) were obtained in identified abducens motoneurons (aMns) from young rats (P5-P13). Three types of mIPSC were distinguished according to their kinetics and their sensitivity to receptor antagonists: faster decaying events mediated by glycine receptors (glyRs), slower decaying events mediated by GABA(A) receptors (GABA(A)Rs), and mIPSCs displaying two components corresponding to GABA and glycine co-release. Dual component events accounted for approximately 30 % of mIPSCs, independently of the rat's age and were also identified during evoked transmitter release. In contrast, the kinetics of glyR- and GABA(A)R-mediated mIPSCs became faster during development. Monosynaptic inhibitory postsynaptic potentials (IPSPs) were able to fully inhibit motoneuron discharge elicited by current pulses. When the GABA(A)R-mediated component or the glyR-mediated component of the IPSP was blocked, the inhibition of motoneuron firing was reduced. The 20-80 % rise time and duration of GABA(A)R-mediated IPSPs were significantly longer than those mediated by glyRs. The time window of inhibition for each component was determined using single postsynaptic action potentials elicited with various delays from the onset of the IPSP. GlyR-mediated IPSPs induced fast transient inhibition whereas GABA(A)R-mediated IPSPs induced slow sustained suppression of firing. Using a modelling approach, we found that the two components summated non-linearly. We conclude that in developing aMns, co-release of GABA and glycine determines the strength and timing of inhibition through non-linear interactions between the two components, thus optimizing inhibition of motoneuron function.


Assuntos
Tronco Encefálico/metabolismo , Glicina/metabolismo , Neurônios Motores/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Tronco Encefálico/citologia , Eletrofisiologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Antagonistas de Receptores de GABA-A , Glicina/antagonistas & inibidores , Técnicas In Vitro , Cinética , Potenciais da Membrana/fisiologia , Microscopia de Vídeo , Modelos Neurológicos , Vias Neurais/fisiologia , Dinâmica não Linear , Técnicas de Patch-Clamp , Canais de Potássio/metabolismo , Ratos , Ratos Wistar , Reflexo Vestíbulo-Ocular/fisiologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...