Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Neurosci ; 42(30): 5843-5859, 2022 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-35732494

RESUMO

Temporal lobe epilepsy (TLE), the most common focal seizure disorder in adults, can be instigated in experimental animals by convulsant-induced status epilepticus (SE). Principal hippocampal neurons from SE-experienced epileptic male rats (post-SE neurons) display markedly augmented spike output compared with neurons from nonepileptic animals (non-SE neurons). This enhanced firing results from a cAMP-dependent protein kinase A-mediated inhibition of KCa3.1, a subclass of Ca2+-gated K+ channels generating the slow afterhyperpolarizing Ca2+-gated K+ current (IsAHP). The inhibition of KCa3.1 in post-SE neurons leads to a marked reduction in amplitude of the IsAHP that evolves during repetitive firing, as well as in amplitude of the associated Ca2+-dependent component of the slow afterhyperpolarization potential (KCa-sAHP). Here we show that KCa3.1 inhibition in post-SE neurons is induced by corticotropin releasing factor (CRF) through its Type 1 receptor (CRF1R). Acute application of CRF1R antagonists restores KCa3.1 activity in post-SE neurons, normalizing KCa-sAHP/IsAHP amplitudes and neuronal spike output, without affecting these variables in non-SE neurons. Moreover, pharmacological antagonism of CRF1Rs in vivo reduces the frequency of spontaneous recurrent seizures in post-SE chronically epileptic rats. These findings may provide a new vista for treating TLE.SIGNIFICANCE STATEMENT Epilepsy, a common neurologic disorder, often develops following a brain insult. Identifying key cellular mechanisms underlying acquired epilepsy is critical for developing effective antiepileptic therapies. In an experimental model of acquired epilepsy, principal hippocampal neurons manifest hyperexcitability because of downregulation of KCa3.1, a subtype of Ca2+-gated K+ ion channels. We show that KCa3.1 downregulation is mediated by corticotropin releasing factor (CRF) acting through its Type 1 receptor (CRF1R). Congruently, acute application of selective CRF1R antagonists restores KCa3.1 channel activity, leading to normalization of neuronal excitability. In the same model, injection of a CRF1R antagonist to epileptic animals markedly decreases the frequency of electrographic seizures. Therefore, targeting CRF1Rs may provide a new strategy in the treatment of acquired epilepsy.


Assuntos
Hormônio Liberador da Corticotropina , Epilepsia do Lobo Temporal , Epilepsia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária , Estado Epiléptico , Animais , Hormônio Liberador da Corticotropina/metabolismo , Modelos Animais de Doenças , Epilepsia/tratamento farmacológico , Epilepsia/metabolismo , Epilepsia do Lobo Temporal/metabolismo , Hipocampo/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Masculino , Neurônios/fisiologia , Ratos , Estado Epiléptico/metabolismo
2.
J Neurosci ; 40(5): 974-995, 2020 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-31959697

RESUMO

Multiple insults to the brain lead to neuronal cell death, thus raising the question to what extent can lost neurons be replenished by adult neurogenesis. Here we focused on the hippocampus and especially the dentate gyrus (DG), a vulnerable brain region and one of the two sites where adult neuronal stem cells (NSCs) reside. While adult hippocampal neurogenesis was extensively studied with regard to its contribution to cognitive enhancement, we focused on their underestimated capability to repair a massively injured, nonfunctional DG. To address this issue, we inflicted substantial DG-specific damage in mice of either sex either by diphtheria toxin-based ablation of >50% of mature DG granule cells (GCs) or by prolonged brain-specific VEGF overexpression culminating in extensive, highly selective loss of DG GCs (thereby also reinforcing the notion of selective DG vulnerability). The neurogenic system promoted effective regeneration by increasing NSCs proliferation/survival rates, restoring a nearly original DG mass, promoting proper rewiring of regenerated neurons to their afferent and efferent partners, and regaining of lost spatial memory. Notably, concomitantly with the natural age-related decline in the levels of neurogenesis, the regenerative capacity of the hippocampus also subsided with age. The study thus revealed an unappreciated regenerative potential of the young DG and suggests hippocampal NSCs as a critical reservoir enabling recovery from catastrophic DG damage.SIGNIFICANCE STATEMENT Adult hippocampal neurogenesis has been extensively studied in the context of its role in cognitive enhancement, but whether, and to what extent can dentate gyrus (DG)-resident neural stem cells drive regeneration of an injured DG has remained unclear. Here we show that DG neurogenesis acts to replace lost neurons and restore lost functions even following massive (>50%) neuronal loss. Age-related decline of neurogenesis is paralleled by a progressive decline of regenerative capacity. Considering also the exceptional vulnerability of the DG to insults, these findings provide a further rationale for maintaining DG neurogenesis in adult life.


Assuntos
Giro Denteado/fisiopatologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Animais , Proliferação de Células , Sobrevivência Celular , Giro Denteado/lesões , Giro Denteado/patologia , Feminino , Masculino , Camundongos Transgênicos
3.
J Physiol ; 599(15): 3735-3754, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34148230

RESUMO

KEY POINTS: Stimulation of postsynaptic muscarinic receptors was shown to excite principal hippocampal neurons by modulating several membrane ion conductances. We show here that activation of postsynaptic muscarinic receptors also causes neuronal excitation by inhibiting Na+ /K+ -ATPase activity. Muscarinic Na+ /K+ -ATPase inhibition is mediated by two separate signalling pathways that lead downstream to enhanced Na+ /K+ -ATPase phosphorylation by activating protein kinase C and protein kinase G. Muscarinic excitation through Na+ /K+ -ATPase inhibition is probably involved in cholinergic modulation of hippocampal activity and may turn out to be a widespread mechanism of neuronal excitation in the brain. ABSTRACT: Stimulation of muscarinic cholinergic receptors on principal hippocampal neurons enhances intrinsic neuronal excitability by modulating several membrane ion conductances. The electrogenic Na+ /K+ -ATPase (NKA; the 'Na+ pump') is a ubiquitous regulator of intrinsic neuronal excitability, generating a hyperpolarizing current to thwart excessive neuronal firing. Using electrophysiological and pharmacological methodologies in rat hippocampal slices, we show that neuronal NKA pumping activity is also subjected to cholinergic regulation. Stimulation of postsynaptic muscarinic, but not nicotinic, cholinergic receptors activates membrane-bound phospholipase C and hydrolysis of membrane-integral phosphatidylinositol 4,5-bisphosphate into diacylglycerol (DAG) and inositol 1,4,5-triphosphate (IP3 ). Along one signalling pathway, DAG activates protein kinase C (PKC). Along a second signalling pathway, IP3 causes Ca2+ release from the endoplasmic reticulum, facilitating nitric oxide (NO) production. The rise in NO levels stimulates cGMP synthesis by guanylate-cyclase, activating protein kinase G (PKG). The two pathways converge to cause partial NKA inhibition through enzyme phosphorylation by PKC and PKG, leading to a marked increase in intrinsic neuronal excitability. This novel mechanism of neuronal NKA regulation probably contributes to the cholinergic modulation of hippocampal activity in spatial navigation, learning and memory.


Assuntos
Hipocampo , ATPase Trocadora de Sódio-Potássio , Animais , Colinérgicos , Proteínas Quinases Dependentes de GMP Cíclico , Hipocampo/metabolismo , Neurônios/metabolismo , Ratos , ATPase Trocadora de Sódio-Potássio/metabolismo
4.
J Neurosci ; 39(50): 9914-9926, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31672789

RESUMO

Brain insults, such as trauma, stroke, anoxia, and status epilepticus (SE), cause multiple changes in synaptic function and intrinsic properties of surviving neurons that may lead to the development of epilepsy. Experimentally, a single SE episode, induced by the convulsant pilocarpine, initiates the development of an epileptic condition resembling human temporal lobe epilepsy (TLE). Principal hippocampal neurons from such epileptic animals display enhanced spike output in response to excitatory stimuli compared with neurons from nonepileptic animals. This enhanced firing is negatively related to the size of the slow afterhyperpolarization (sAHP), which is reduced in the epileptic neurons. The sAHP is an intrinsic neuronal negative feedback mechanism consisting normally of two partially overlapping components produced by disparate mechanisms. One component is generated by activation of Ca2+-gated K+ (KCa) channels, likely KCa3.1, consequent to spike Ca2+ influx (the KCa-sAHP component). The second component is generated by enhancement of the electrogenic Na+/K+ ATPase (NKA) by spike Na+ influx (NKA-sAHP component). Here we show that the KCa-sAHP component is markedly reduced in male rat epileptic neurons, whereas the NKA-sAHP component is not altered. The KCa-sAHP reduction is due to the downregulation of KCa3.1 channels, mediated by cAMP-dependent protein kinase A (PKA). This sustained effect can be acutely reversed by applying PKA inhibitors, leading also to normalization of the spike output of epileptic neurons. We propose that the novel "acquired channelopathy" described here, namely, PKA-mediated downregulation of KCa3.1 activity, provides an innovative target for developing new treatments for TLE, hopefully overcoming the pharmacoresistance to traditional drugs.SIGNIFICANCE STATEMENT Epilepsy, a common neurological disorder, often develops following a brain insult. Identifying key molecular and cellular mechanisms underlying acquired epilepsy is critical for developing effective antiepileptic therapies. In an experimental model of acquired epilepsy, we show that principal hippocampal neurons become intrinsically hyperexcitable. This alteration is due predominantly to the downregulation of a ubiquitous class of potassium ion channels, KCa3.1, whose main function is to dampen neuronal excitability. KCa3.1 downregulation is mediated by the cAMP-dependent protein kinase A (PKA) signaling pathway. Most importantly, it can be acutely reversed by PKA inhibitors, leading to recovery of KCa3.1 function and normalization of neuronal excitability. The discovery of this novel epileptogenic mechanism hopefully will facilitate the development of more efficient pharmacotherapy for acquired epilepsy.


Assuntos
Potenciais de Ação/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Epilepsia do Lobo Temporal/fisiopatologia , Hipocampo/fisiopatologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Neurônios/fisiologia , Animais , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/metabolismo , Hipocampo/metabolismo , Masculino , Ratos , Ratos Wistar
5.
J Neurosci ; 39(28): 5440-5451, 2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31085608

RESUMO

The Na+/K+-ATPase (NKA) is a ubiquitous membrane-bound enzyme responsible for generating and maintaining the Na+ and K+ electrochemical gradients across the plasmalemma of living cells. Numerous studies in non-neuronal tissues have shown that this transport mechanism is reversibly regulated by phosphorylation/dephosphorylation of the catalytic α subunit and/or associated proteins. In neurons, Na+/K+ transport by NKA is essential for almost all neuronal operations, consuming up to two-thirds of the neuron's energy expenditure. However, little is known about its cellular regulatory mechanisms. Here we have used an electrophysiological approach to monitor NKA transport activity in male rat hippocampal neurons in situ We report that this activity is regulated by a balance between serine/threonine phosphorylation and dephosphorylation. Phosphorylation by the protein kinases PKG and PKC inhibits NKA activity, whereas dephosphorylation by the protein phosphatases PP-1 and PP-2B (calcineurin) reverses this effect. Given that these kinases and phosphatases serve as downstream effectors in key neuronal signaling pathways, they may mediate the coupling of primary messengers, such as neurotransmitters, hormones, and growth factors, to the NKAs, through which multiple brain functions can be regulated or dysregulated.SIGNIFICANCE STATEMENT The Na+/K+-ATPase (NKA), known as the "Na+ pump," is a ubiquitous membrane-bound enzyme responsible for generating and maintaining the Na+ and K+ electrochemical gradients across the plasma membrane of living cells. In neurons, as in most types of cells, the NKA generates the negative resting membrane potential, which is the basis for almost all aspects of cellular function. Here we used an electrophysiological approach to monitor physiological NKA transport activity in single hippocampal pyramidal cells in situ We have found that neuronal NKA activity is oppositely regulated by phosphorylation and dephosphorylation, and we have identified the main protein kinases and phosphatases mediating this regulation. This fundamental form of NKA regulation likely plays a role in multiple brain functions.


Assuntos
Calcineurina/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Proteína Quinase C/metabolismo , Proteína Fosfatase 1/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Hipocampo/metabolismo , Hipocampo/fisiologia , Masculino , Potenciais da Membrana , Neurônios/metabolismo , Neurônios/fisiologia , Fosforilação , Ratos , Ratos Wistar
6.
Hippocampus ; 28(5): 338-357, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29431274

RESUMO

In many types of CNS neurons, repetitive spiking produces a slow afterhyperpolarization (sAHP), providing sustained, intrinsically generated negative feedback to neuronal excitation. Changes in the sAHP have been implicated in learning behaviors, in cognitive decline in aging, and in epileptogenesis. Despite its importance in brain function, the mechanisms generating the sAHP are still controversial. Here we have addressed the roles of M-type K+ current (IM ), Ca2+ -gated K+ currents (ICa(K) 's) and Na+ /K+ -ATPases (NKAs) current to sAHP generation in adult rat CA1 pyramidal cells maintained at near-physiological temperature (35 °C). No evidence for IM contribution to the sAHP was found in these neurons. Both ICa(K) 's and NKA current contributed to sAHP generation, the latter being the predominant generator of the sAHP, particularly when evoked with short trains of spikes. Of the different NKA isoenzymes, α1 -NKA played the key role, endowing the sAHP a steep voltage-dependence. Thus normal and pathological changes in α1 -NKA expression or function may affect cognitive processes by modulating the inhibitory efficacy of the sAHP.


Assuntos
Região CA1 Hipocampal/metabolismo , Potenciais da Membrana/fisiologia , Canais de Potássio Cálcio-Ativados/metabolismo , Células Piramidais/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/farmacologia , Retroalimentação Fisiológica/efeitos dos fármacos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Canais de Potássio Cálcio-Ativados/antagonistas & inibidores , Células Piramidais/efeitos dos fármacos , Ratos Wistar , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Técnicas de Cultura de Tecidos
7.
J Physiol ; 595(3): 713-738, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27506492

RESUMO

KEY POINTS: Neuroinflammation associated with CNS insults leads to neuronal hyperexcitability, which may culminate in epileptiform discharges. Application of the endotoxin lipopolysaccharide (LPS) to brain tissue initiates a neuroinflammatory cascade, providing an experimental model to study the mechanisms of neuroinflammatory neuronal hyperexcitability. Here we show that LPS application to hippocampal slices markedly enhances the excitability of CA1 pyramidal cells by inhibiting a specific potassium current, the M-current, generated by KV 7/M channels, which controls the excitability of almost every neuron in the CNS. The LPS-induced M-current inhibition is triggered by sequential activation of microglia, astrocytes and pyramidal cells, mediated by metabotropic purinergic and glutamatergic transmission, leading to blockade of KV 7/M channels by calcium released from intracellular stores. The identification of the downstream molecular target of neuroinflammation, namely the KV 7/M channel, potentially has far reaching implications for the understanding and treatment of many acute and chronic brain disorders. ABSTRACT: Acute brain insults and many chronic brain diseases manifest an innate inflammatory response. The hallmark of this response is glia activation, which promotes repair of damaged tissue, but also induces structural and functional changes that may lead to an increase in neuronal excitability. We have investigated the mechanisms involved in the modulation of neuronal activity by acute inflammation. Initiating inflammatory responses in hippocampal tissue rapidly led to neuronal depolarization and repetitive firing even in the absence of active synaptic transmission. This action was mediated by a complex metabotropic purinergic and glutamatergic glia-to-neuron signalling cascade, leading to the blockade of neuronal KV 7/M channels by Ca2+ released from internal stores. These channels generate the low voltage-activating, non-inactivating M-type K+ current (M-current) that controls intrinsic neuronal excitability, and its inhibition was the predominant cause of the inflammation-induced hyperexcitability. Our discovery that the ubiquitous KV 7/M channels are the downstream target of the inflammation-induced cascade, has far reaching implications for the understanding and treatment of many acute and chronic brain disorders.


Assuntos
Canais de Potássio KCNQ/fisiologia , Lipopolissacarídeos/farmacologia , Células Piramidais/efeitos dos fármacos , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Região CA1 Hipocampal/citologia , Cálcio/fisiologia , Masculino , Células Piramidais/fisiologia , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5/fisiologia , Receptores de Glutamato Metabotrópico/fisiologia , Receptores Purinérgicos P2Y1/fisiologia
8.
J Neurosci ; 35(46): 15240-53, 2015 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-26586813

RESUMO

Dendritic voltage-gated ion channels profoundly shape the integrative properties of neuronal dendrites. In epilepsy, numerous changes in dendritic ion channels have been described, all of them due to either their altered transcription or phosphorylation. In pilocarpine-treated chronically epileptic rats, we describe a novel mechanism that causes an increased proximal dendritic persistent Na(+) current (INaP). We demonstrate using a combination of electrophysiology and molecular approaches that the upregulation of dendritic INaP is due to a relief from polyamine-dependent inhibition. The polyamine deficit in hippocampal neurons is likely caused by an upregulation of the degrading enzyme spermidine/spermine acetyltransferase. Multiphoton glutamate uncaging experiments revealed that the increase in dendritic INaP causes augmented dendritic summation of excitatory inputs. These results establish a novel post-transcriptional modification of ion channels in chronic epilepsy and may provide a novel avenue for treatment of temporal lobe epilepsy. SIGNIFICANCE STATEMENT: In this paper, we describe a novel mechanism that causes increased dendritic persistent Na(+) current. We demonstrate using a combination of electrophysiology and molecular approaches that the upregulation of persistent Na(+) currents is due to a relief from polyamine-dependent inhibition. The polyamine deficit in hippocampal neurons is likely caused by an upregulation of the degrading enzyme spermidine/spermine acetyltransferase. Multiphoton glutamate uncaging experiments revealed that the increase in dendritic persistent Na current causes augmented dendritic summation of excitatory inputs. We believe that these results establish a novel post-transcriptional modification of ion channels in chronic epilepsy.


Assuntos
Região CA1 Hipocampal/patologia , Dendritos/fisiologia , Regulação para Baixo/fisiologia , Canais de Sódio/fisiologia , Espermina/metabolismo , Estado Epiléptico/patologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Análise de Variância , Animais , Dendritos/efeitos dos fármacos , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Humanos , Técnicas In Vitro , Masculino , Agonistas Muscarínicos/toxicidade , Pilocarpina/toxicidade , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Estatísticas não Paramétricas , Estado Epiléptico/induzido quimicamente , Sinaptofisina/metabolismo , Tetrodotoxina/farmacologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
9.
J Neurosci ; 34(24): 8219-30, 2014 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-24920626

RESUMO

Small-conductance Ca(2+)-activated K(+) (SK or K(Ca)2) channels are widely expressed in the CNS. In several types of neurons, these channels were shown to become activated during repetitive firing, causing early spike frequency adaptation. In CA1 pyramidal cells, SK channels in dendritic spines were shown to regulate synaptic transmission. However, the presence of functional SK channels in the somata and their role in controlling the intrinsic firing of these neurons has been controversial. Using whole-cell voltage-clamp and current-clamp recordings in acute hippocampal slices and focal applications of irreversible and reversible SK channel blockers, we provide evidence that functional SK channels are expressed in the somata and proximal dendrites of adult rat CA1 pyramidal cells. Although these channels can generate a medium duration afterhyperpolarizing current, they play only an auxiliary role in controlling the intrinsic excitability of these neurons, secondary to the low voltage-activating, noninactivating K(V)7/M channels. As long as K(V)7/M channels are operative, activation of SK channels during repetitive firing does not notably affect the spike output of CA1 pyramidal cells. However, when K(V)7/M channel activity is compromised, SK channel activation significantly and uniquely reduces spike output of these neurons. Therefore, proximal SK channels provide a "second line of defense" against intrinsic hyperexcitability, which may play a role in multiple conditions in which K(V)7/M channels activity is compromised, such as hyposmolarity.


Assuntos
Potenciais de Ação/fisiologia , Região CA1 Hipocampal/citologia , Dendritos/metabolismo , Células Piramidais/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , 4-Aminopiridina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Análise de Variância , Animais , Antracenos/farmacologia , Apamina/farmacologia , Biofísica , Dendritos/efeitos dos fármacos , Estimulação Elétrica , Técnicas In Vitro , Masculino , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Cloreto de Potássio/farmacologia , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Ratos , Fatores de Tempo
10.
J Biol Chem ; 287(19): 15489-501, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22431737

RESUMO

The pore-forming Ca(2+) channel subunit Ca(V)3.2 mediates a low voltage-activated (T-type) Ca(2+) current (I(CaT)) that contributes pivotally to neuronal and cardiac pacemaker activity. Despite the importance of tightly regulated Ca(V)3.2 levels, the mechanisms regulating its transcriptional dynamics are not well understood. Here, we have identified two key factors that up- and down-regulate the expression of the gene encoding Ca(V)3.2 (Cacna1h). First, we determined the promoter region and observed several stimulatory and inhibitory clusters. Furthermore, we found binding sites for the transcription factor early growth response 1 (Egr1/Zif268/Krox-24) to be highly overrepresented within the Ca(V)3.2 promoter region. mRNA expression analyses and dual-luciferase promoter assays revealed that the Ca(V)3.2 promoter was strongly activated by Egr1 overexpression in vitro and in vivo. Subsequent chromatin immunoprecipitation assays in NG108-15 cells and mouse hippocampi confirmed specific Egr1 binding to the Ca(V)3.2 promoter. Congruently, whole-cell I(CaT) values were significantly larger after Egr1 overexpression. Intriguingly, Egr1-induced activation of the Ca(V)3.2 promoter was effectively counteracted by the repressor element 1-silencing transcription factor (REST). Thus, Egr1 and REST can bi-directionally regulate Ca(V)3.2 promoter activity and mRNA expression and, hence, the size of I(CaT). This mechanism has critical implications for the regulation of neuronal and cardiac Ca(2+) homeostasis under physiological conditions and in episodic disorders such as arrhythmias and epilepsy.


Assuntos
Canais de Cálcio Tipo T/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica , Regiões Promotoras Genéticas/genética , Proteínas Repressoras/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Encéfalo/metabolismo , Canais de Cálcio Tipo T/fisiologia , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Proteína 1 de Resposta de Crescimento Precoce/genética , Células HEK293 , Humanos , Potenciais da Membrana , Camundongos , Dados de Sequência Molecular , Técnicas de Patch-Clamp , Ligação Proteica , Ratos , Sequências Reguladoras de Ácido Nucleico/genética , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Transfecção
11.
Nat Rev Neurosci ; 9(5): 357-69, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18425090

RESUMO

The input-output relationship of neuronal networks depends both on their synaptic connectivity and on the intrinsic properties of their neuronal elements. In addition to altered synaptic properties, profound changes in intrinsic neuronal properties are observed in many CNS disorders. These changes reflect alterations in the functional properties of dendritic and somatic voltage- and Ca2+-gated ion channels. The molecular mechanisms underlying this intrinsic plasticity comprise the highly specific transcriptional or post-transcriptional regulation of ion-channel expression, trafficking and function. The studies reviewed here show that intrinsic plasticity, in conjunction with synaptic plasticity, can fundamentally alter the input-output properties of neuronal networks in CNS disorders.


Assuntos
Doenças do Sistema Nervoso Central/patologia , Doenças do Sistema Nervoso Central/fisiopatologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Animais , Humanos , Ativação do Canal Iônico/fisiologia , Canais Iônicos/fisiologia , Transporte Proteico/fisiologia , Sinapses/fisiologia
12.
J Physiol ; 590(22): 5895-905, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22930274

RESUMO

Extracellular zinc can induce numerous acute and persistent physiological and toxic effects in neurons by acting at their plasma membrane or intracellularly following permeation or uptake into them. Zinc acutely and reversibly blocks T-type voltage-gated calcium current (I(CaT)), but the long-term effect of zinc on this current has not been studied. Because chemically induced status epilepticus (SE) results in the release of zinc into the extracellular space, as well as in a long-lasting increase in I(CaT) in CA1 pyramidal cells, we hypothesized that zinc may play a causative role in I(CaT) upregulation. We tested this hypothesis by monitoring for 18 days the effects of zinc and ibotenic acid (a neurotoxic agent serving as control for zinc), injected into the right lateral ventricle, on I(CaT) in rat CA1 pyramidal cells. Both zinc and ibotenic acid caused marked hippocampal lesions on the side of injection, but only minor damage to contralateral hippocampi. Zinc, but not ibotenic acid, caused upregulation of a nickel-sensitive I(CaT) in a subset of contralateral CA1 pyramidal cells, appearing 2 days after injection and lasting for about 2 weeks thereafter. In contrast, acute application of zinc to CA1 pyramidal cells promptly blocked I(CaT). These data indicate that extracellular zinc has a dual effect on I(CaT), blocking it acutely while causing its long-term upregulation. Through the latter effect, zinc may regulate the intrinsic excitability of principal neurons, particularly in pathological conditions associated with enhanced release of zinc, such as SE.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Canais de Cálcio Tipo T/efeitos dos fármacos , Hipocampo/fisiologia , Células Piramidais/fisiologia , Zinco/farmacologia , Potenciais de Ação/fisiologia , Animais , Canais de Cálcio Tipo T/fisiologia , Morte Celular , Ácido Ibotênico/farmacologia , Masculino , Níquel/farmacologia , Células Piramidais/efeitos dos fármacos , Ratos , Zinco/toxicidade
13.
J Neurophysiol ; 105(1): 117-29, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20980543

RESUMO

Brain damage causes multiple changes in synaptic function and intrinsic properties of surviving neurons, leading to the development of chronic epilepsy. In the widely used pilocarpine-status epilepticus (SE) rat model of temporal lobe epilepsy (TLE), a major alteration is the marked increase in the fraction of intrinsically bursting CA1 pyramidal cells. Here we have differentiated between two types of bursting phenotypes: 1) bursting in response to threshold-straddling excitatory current pulses (low-threshold bursting) and 2) bursting only in response to suprathreshold stimuli (high-threshold bursting). Low-threshold bursting prevailed in 46.5% of SE-experienced neurons sampled 1-4 wk after pilocarpine-SE, but was rarely seen in control neurons (1.9%). As previously shown, it appeared to be driven predominantly by a T-type Ca(2+) current (I(CaT)) in the apical dendrites. After blocking low-threshold bursting with Ni(2+), the same neurons still manifested a high-threshold bursting phenotype. Another 40.1% of SE-experienced neurons displayed only a high-threshold bursting phenotype and the remaining 13.4% of these neurons were nonbursters. Altogether, high-threshold bursting prevailed in 86.6% of SE-experienced neurons, but only in 33.0% of control neurons. Several lines of evidence indicated that high-threshold bursting is driven by persistent Na(+) current (I(NaP)) at or near the soma. Congruently, I(NaP) was 1.5-fold larger in SE-experienced versus control neurons. We conclude that an increase in I(NaP), conjointly with an increase in I(CaT), strongly contributes to the predominance of bursting phenotypes in CA1 pyramidal cells early after pilocarpine-SE and thus likely plays a role in the development of a chronic epileptic condition in this TLE model.


Assuntos
Região CA1 Hipocampal/fisiopatologia , Neurônios/fisiologia , Canais de Sódio/fisiologia , Estado Epiléptico/fisiopatologia , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Canais de Cálcio Tipo T/fisiologia , Masculino , Modelos Animais , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Pilocarpina/efeitos adversos , Ratos , Ratos Endogâmicos , Ratos Wistar , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Estado Epiléptico/induzido quimicamente , Tetrodotoxina/farmacologia , Fatores de Tempo
14.
J Neurosci ; 29(36): 11098-111, 2009 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-19741116

RESUMO

Modest decreases in extracellular osmolarity induce brain hyperexcitability that may culminate in epileptic seizures. At the cellular level, moderate hyposmolarity markedly potentiates the intrinsic neuronal excitability of principal cortical neurons without significantly affecting their volume. The most conspicuous cellular effect of hyposmolarity is converting regular firing neurons to burst-firing mode. This effect is underlain by hyposmotic facilitation of the spike afterdepolarization (ADP), but its ionic mechanism is unknown. Because blockers of K(V)7 (KCNQ) channels underlying neuronal M-type K(+) currents (K(V)7/M channels) also cause spike ADP facilitation and bursting, we hypothesized that lowering osmolarity inhibits these channels. Using current- and voltage-clamp recordings in CA1 pyramidal cells in situ, we have confirmed this hypothesis. Furthermore, we show that hyposmotic inhibition of K(V)7/M channels is mediated by an increase in intracellular Ca(2+) concentration via release from internal stores but not via influx of extracellular Ca(2+). Finally, we show that interfering with internal Ca(2+)-mediated inhibition of K(V)7/M channels entirely protects against hyposmotic ADP facilitation and bursting, indicating the exclusivity of this novel mechanism in producing intrinsic neuronal hyperexcitability in hyposmotic conditions.


Assuntos
Canais de Potássio KCNQ/fisiologia , Neurônios/metabolismo , Osmose/fisiologia , Potenciais de Ação/fisiologia , Animais , Ativação do Canal Iônico/fisiologia , Masculino , Neurônios/fisiologia , Técnicas de Patch-Clamp , Células Piramidais/metabolismo , Ratos
15.
J Neurosci ; 28(49): 13341-53, 2008 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-19052226

RESUMO

In both humans and animals, an insult to the brain can lead, after a variable latent period, to the appearance of spontaneous epileptic seizures that persist for life. The underlying processes, collectively referred to as epileptogenesis, include multiple structural and functional neuronal alterations. We have identified the T-type Ca(2+) channel Ca(v)3.2 as a central player in epileptogenesis. We show that a transient and selective upregulation of Ca(v)3.2 subunits on the mRNA and protein levels after status epilepticus causes an increase in cellular T-type Ca(2+) currents and a transitional increase in intrinsic burst firing. These functional changes are absent in mice lacking Ca(v)3.2 subunits. Intriguingly, the development of neuropathological hallmarks of chronic epilepsy, such as subfield-specific neuron loss in the hippocampal formation and mossy fiber sprouting, was virtually completely absent in Ca(v)3.2(-/-) mice. In addition, the appearance of spontaneous seizures was dramatically reduced in these mice. Together, these data establish transcriptional induction of Ca(v)3.2 as a critical step in epileptogenesis and neuronal vulnerability.


Assuntos
Canais de Cálcio Tipo T/genética , Sinalização do Cálcio/genética , Epilepsia do Lobo Temporal/genética , Hipocampo/metabolismo , Neurônios/metabolismo , Regulação para Cima/genética , Animais , Canais de Cálcio Tipo T/metabolismo , Canalopatias/genética , Canalopatias/metabolismo , Canalopatias/fisiopatologia , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/fisiopatologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Predisposição Genética para Doença/genética , Hipocampo/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Fibras Musgosas Hipocampais/metabolismo , Fibras Musgosas Hipocampais/fisiopatologia , Agonistas Muscarínicos/farmacologia , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Neurônios/efeitos dos fármacos , Pilocarpina/farmacologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos , Ratos Wistar , Ativação Transcricional/genética
16.
J Physiol ; 586(5): 1351-63, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18187471

RESUMO

In principal brain neurons, activation of Ca(2+) channels during an action potential, or spike, causes Ca(2+) entry into the cytosol within a millisecond. This in turn causes rapid activation of large conductance Ca(2+)-gated channels, which enhances repolarization and abbreviates the spike. Here we describe another remarkable consequence of spike Ca(2+) entry: enhancement of the spike afterdepolarization. This action is also mediated by intracellular modulation of a particular class of K(+) channels, namely by inhibition of K(V)7 (KCNQ) channels. These channels generate the subthreshold, non-inactivating M-type K(+) current, whose activation curtails the spike afterdepolarization. Inhibition of K(V)7/M by spike Ca(2+) entry allows the spike afterdepolarization to grow and can convert solitary spikes into high-frequency bursts of action potentials. Through this novel intracellular modulatory action, Ca(2+) spike entry regulates the discharge mode and the signalling capacity of principal brain neurons.


Assuntos
Potenciais de Ação/fisiologia , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Canais de Potássio KCNQ/antagonistas & inibidores , Difosfato de Adenosina/metabolismo , Animais , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Canais de Potássio KCNQ/efeitos dos fármacos , Canais de Potássio KCNQ/metabolismo , Masculino , Neurônios/metabolismo , Ratos , Ratos Endogâmicos
17.
J Physiol ; 586(22): 5437-53, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18801845

RESUMO

Early in development, network activity in the hippocampus is characterized by recurrent synchronous bursts, whose cellular correlates are giant depolarizing potentials (GDPs). The propensity for generating GDPs is attributed to GABAergic synaptic transmission being depolarizing and excitatory in neonatal neurons. However, developmental regulation of intrinsic conductances may also influence GDPs generation. A likely candidate is the non-inactivating, low-threshold, muscarinic-sensitive K(+) current (M current; I(m)), which down-regulates intrinsic bursting activity in adult hippocampal pyramidal neurons. Western blot analysis of homogenates of the CA3 hippocampal region showed that expression of the Kv7.2 subunit, one of the constituents of neuronal M channels, is weak in neonatal neurons, and markedly increases after the first postnatal week. Likewise, the density of I(m) was very low in neonatal CA3 pyramidal cells and increased later on. Spontaneously occurring intrinsic bursts in neonatal neurons were longer and more robust, and recurred more regularly, than in juvenile neurons. The I(m) blocker linopirdine only mildly affected intrinsic bursting in neonatal neurons, but strongly facilitated and regularized it in juvenile neurons. We conclude that the low expression of Kv7/M channels and the depolarizing action of GABA early after birth enhance intrinsic bursting and neuronal synchronization leading to generation of GDPs within the hippocampal network.


Assuntos
Hipocampo/metabolismo , Canal de Potássio KCNQ2/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Carbamatos/farmacologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/crescimento & desenvolvimento , Técnicas In Vitro , Indóis/farmacologia , Canal de Potássio KCNQ2/agonistas , Canal de Potássio KCNQ2/antagonistas & inibidores , Cinética , Rede Nervosa/citologia , Rede Nervosa/crescimento & desenvolvimento , Rede Nervosa/metabolismo , Fenilenodiaminas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Piridinas/farmacologia , Ratos , Ratos Wistar , Inibidores de Simportadores de Cloreto de Sódio e Potássio , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Membro 2 da Família 12 de Carreador de Soluto , Transmissão Sináptica/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Ácido gama-Aminobutírico/farmacologia
18.
Neuroscience ; 357: 325-337, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28624573

RESUMO

OBJECTIVE: In experimental models of temporal lobe epilepsy (TLE), brain neurons manifest multiple changes in intrinsic excitability that contribute to neuronal network hyperexcitability. We have investigated whether the intrinsic firing response gain, quantified by the slope of the function relating the number of evoked spikes (Ns) to input excitatory current intensity (I), is modified in principal rat hippocampal neurons in the pilocarpine-status epilepticus (SE) model of TLE. METHODS: Intracellular recordings were made in CA3 and CA1 pyramidal cells (PCs) and dentate granule cells (GCs) in acute hippocampal slices obtained 7-36days after pilocarpine-SE. Firing response gains were determined empirically from Ns/I relationships and compared to other measured neuronal properties. RESULTS: The firing response gain in all three types of principal neurons, particularly in CA3 PCs, was markedly multiplied following pilocarpine-SE. Analyses of persistent changes in active and passive properties of CA3 PCs suggested that this increase is multifactorial in origin, the major factors being a reduction in amplitude of the slow afterhyperpolarization and an increase in the fraction of bursting neurons. SIGNIFICANCE: Here we show that pilocarpine-SE causes multiplication of the firing response gain in the three principal neurons in the hippocampal trisynaptic pathway. This alteration undoubtedly would contribute to hippocampal hyperexcitability in SE-induced TLE.


Assuntos
Potenciais de Ação/fisiologia , Hipocampo/fisiopatologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Estado Epiléptico/fisiopatologia , Animais , Modelos Animais de Doenças , Masculino , Pilocarpina , Ratos Wistar , Técnicas de Cultura de Tecidos
19.
J Neurosci ; 25(42): 9704-20, 2005 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-16237175

RESUMO

In many principal brain neurons, the fast, all-or-none Na+ spike initiated at the proximal axon is followed by a slow, graded after depolarization (ADP). The spike ADP is critically important in determining the firing mode of many neurons; large ADPs cause neurons to fire bursts of spikes rather than solitary spikes. Nonetheless, not much is known about how and where spike ADPs are initiated. We addressed these questions in adult CA1 pyramidal cells, which manifest conspicuous somatic spike ADPs and an associated propensity for bursting, using sharp and patch microelectrode recordings in acutely isolated hippocampal slices and single neurons. Voltage-clamp commands mimicking spike waveforms evoked transient Na+ spike currents that declined quickly after the spike but were followed by substantial sustained Na+ spike after currents. Drugs that blocked the persistent Na+ current (INaP), markedly suppressed the sustained Na+ spike after currents, as well as spike ADPs and associated bursting. Ca2+ spike after currents were much smaller, and reducing them had no noticeable effect on the spike ADPs. Truncating the apical dendrites affected neither spike ADPs nor the firing modes of these neurons. Application of INaP blockers to truncated neurons, or their focal application to the somatic region of intact neurons, suppressed spike ADPs and associated bursting, whereas their focal application to distal dendrites did not. We conclude that the somatic spike ADPs are generated predominantly by persistent Na+ channels located at or near the soma. Through this action, proximal INaP critically determines the firing mode and spike output of adult CA1 pyramidal cells.


Assuntos
Potenciais de Ação/fisiologia , Células Piramidais/fisiologia , Canais de Sódio/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Masculino , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Wistar , Bloqueadores dos Canais de Sódio/farmacologia
20.
J Neurosci ; 24(19): 4614-24, 2004 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-15140933

RESUMO

KCNQ channel subunits are widely expressed in peripheral and central neurons, where they give rise to a muscarinic-sensitive, subthreshold, and noninactivating K+ current (M-current). It is generally agreed that activation of KCNQ/M channels contributes to spike frequency adaptation during sustained depolarizations but is too slow to influence the repolarization of solitary spikes. This concept, however, is based mainly on experiments with muscarinic agonists, the multiple effects on membrane conductances of which may overshadow the distinctive effects of KCNQ/M channel block. Here, we have used selective modulators of KCNQ/M channels to investigate their role in spike electrogenesis in CA1 pyramidal cells. Solitary spikes were evoked by brief depolarizing current pulses injected into the neurons. The KCNQ/M channel blockers linopirdine and XE991 markedly enhanced the spike afterdepolarization (ADP) and, in most neurons, converted solitary ("simple") spikes to high-frequency bursts of three to seven spikes ("complex" spikes). Conversely, the KCNQ/M channel opener retigabine reduced the spike ADP and induced regular firing in bursting neurons. Selective block of BK or SK channels had no effect on the spike ADP or firing mode in these neurons. We conclude that KCNQ/M channels activate during the spike ADP and limit its duration, thereby precluding its escalation to a burst. Consequently, down-modulation of KCNQ/M channels converts the neuronal firing pattern from simple to complex spiking, whereas up-modulation of these channels exerts the opposite effect.


Assuntos
Potenciais de Ação/fisiologia , Hipocampo/fisiologia , Neurônios/fisiologia , Canais de Potássio/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Antracenos/farmacologia , Carbamatos/farmacologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Técnicas In Vitro , Indóis/farmacologia , Neurônios/efeitos dos fármacos , Fenilenodiaminas/farmacologia , Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Piridinas/farmacologia , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA