Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(31): e2204901119, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35881790

RESUMO

Although a wide variety of genetic tools has been developed to study learning and memory, the molecular basis of memory encoding remains incompletely understood. Here, we undertook an unbiased approach to identify novel genes critical for memory encoding. From a large-scale, in vivo mutagenesis screen using contextual fear conditioning, we isolated in mice a mutant, named Clueless, with spatial learning deficits. A causative missense mutation (G434V) was found in the voltage-gated potassium channel, subfamily C member 3 (Kcnc3) gene in a region that encodes a transmembrane voltage sensor. Generation of a Kcnc3G434V CRISPR mutant mouse confirmed this mutation as the cause of the learning defects. While G434V had no effect on transcription, translation, or trafficking of the channel, electrophysiological analysis of the G434V mutant channel revealed a complete loss of voltage-gated conductance, a broadening of the action potential, and decreased neuronal firing. Together, our findings have revealed a role for Kcnc3 in learning and memory.


Assuntos
Hipocampo , Deficiências da Aprendizagem , Memória , Mutação de Sentido Incorreto , Canais de Potássio Shaw , Potenciais de Ação/fisiologia , Animais , Hipocampo/fisiopatologia , Deficiências da Aprendizagem/genética , Camundongos , Camundongos Endogâmicos C57BL , Canais de Potássio Shaw/genética , Canais de Potássio Shaw/fisiologia
2.
J Physiol ; 597(23): 5707-5722, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31612994

RESUMO

As in mammals, Drosophila circadian clock neurons display rhythms of activity with higher action potential firing rates and more positive resting membrane potentials during the day. This rhythmic excitability has been widely observed but, critically, its regulation remains unresolved. We have characterized and modelled the changes underlying these electrical activity rhythms in the lateral ventral clock neurons (LNvs). We show that currents mediated by the voltage-gated potassium channels Shaw (Kv3) and Shal (Kv4) oscillate in a circadian manner. Disruption of these channels, by expression of dominant negative (DN) subunits, leads to changes in circadian locomotor activity and shortens lifespan. LNv whole-cell recordings then show that changes in Shaw and Shal currents drive changes in action potential firing rate and that these rhythms are abolished when the circadian molecular clock is stopped. A whole-cell biophysical model using Hodgkin-Huxley equations can recapitulate these changes in electrical activity. Based on this model and by using dynamic clamp to manipulate clock neurons directly, we can rescue the pharmacological block of Shaw and Shal, restore the firing rhythm, and thus demonstrate the critical importance of Shaw and Shal. Together, these findings point to a key role for Shaw and Shal in controlling circadian firing of clock neurons and show that changes in clock neuron currents can account for this. Moreover, with dynamic clamp we can switch the LNvs between morning-like and evening-like states of electrical activity. We conclude that changes in Shaw and Shal underlie the daily oscillation in LNv firing rate.


Assuntos
Relógios Circadianos/fisiologia , Proteínas de Drosophila/fisiologia , Neurônios/fisiologia , Canais de Potássio Shal/fisiologia , Canais de Potássio Shaw/fisiologia , Animais , Ritmo Circadiano , Drosophila , Feminino , Locomoção , Masculino , Modelos Biológicos
3.
Ann Clin Transl Neurol ; 6(7): 1319-1326, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31353862

RESUMO

A recurrent de novo missense variant in KCNC1, encoding a voltage-gated potassium channel expressed in inhibitory neurons, causes progressive myoclonus epilepsy and ataxia, and a nonsense variant is associated with intellectual disability. We identified three new de novo missense variants in KCNC1 in five unrelated individuals causing different phenotypes featuring either isolated nonprogressive myoclonus (p.Cys208Tyr), intellectual disability (p.Thr399Met), or epilepsy with myoclonic, absence and generalized tonic-clonic seizures, ataxia, and developmental delay (p.Ala421Val, three patients). Functional analyses demonstrated no measurable currents for all identified variants and dominant-negative effects for p.Thr399Met and p.Ala421Val predicting neuronal disinhibition as the underlying disease mechanism.


Assuntos
Estudos de Associação Genética , Mutação de Sentido Incorreto , Canais de Potássio Shaw/genética , Animais , Ataxia/genética , Criança , Códon sem Sentido , Humanos , Deficiência Intelectual/genética , Masculino , Epilepsias Mioclônicas Progressivas , Convulsões/genética , Canais de Potássio Shaw/fisiologia , Xenopus laevis
4.
Neuron ; 98(1): 156-165.e6, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29621485

RESUMO

Fast-spiking, parvalbumin-expressing GABAergic interneurons (PV+-BCs) express a complex machinery of rapid signaling mechanisms, including specialized voltage-gated ion channels to generate brief action potentials (APs). However, short APs are associated with overlapping Na+ and K+ fluxes and are therefore energetically expensive. How the potentially vicious combination of high AP frequency and inefficient spike generation can be reconciled with limited energy supply is presently unclear. To address this question, we performed direct recordings from the PV+-BC axon, the subcellular structure where active conductances for AP initiation and propagation are located. Surprisingly, the energy required for the AP was, on average, only ∼1.6 times the theoretical minimum. High energy efficiency emerged from the combination of fast inactivation of Na+ channels and delayed activation of Kv3-type K+ channels, which minimized ion flux overlap during APs. Thus, the complementary tuning of axonal Na+ and K+ channel gating optimizes both fast signaling properties and metabolic efficiency.


Assuntos
Potenciais de Ação/fisiologia , Axônios/fisiologia , Neurônios GABAérgicos/fisiologia , Interneurônios/fisiologia , Canais de Potássio Shaw/fisiologia , Canais de Sódio/fisiologia , Animais , Metabolismo Energético/fisiologia , Feminino , Hipocampo/fisiologia , Ativação do Canal Iônico/fisiologia , Masculino , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar
5.
J Neurosci ; 37(40): 9705-9714, 2017 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-28877968

RESUMO

Action potential (AP) shape is a key determinant of cellular electrophysiological behavior. We found that in small-diameter, capsaicin-sensitive dorsal root ganglia neurons corresponding to nociceptors (from rats of either sex), stimulation at frequencies as low as 1 Hz produced progressive broadening of the APs. Stimulation at 10 Hz for 3 s resulted in an increase in AP width by an average of 76 ± 7% at 22°C and by 38 ± 3% at 35°C. AP clamp experiments showed that spike broadening results from frequency-dependent reduction of potassium current during spike repolarization. The major current responsible for frequency-dependent reduction of overall spike-repolarizing potassium current was identified as Kv3 current by its sensitivity to low concentrations of 4-aminopyridine (IC50 <100 µm) and block by the peptide inhibitor blood depressing substance I (BDS-I). There was a small component of Kv1-mediated current during AP repolarization, but this current did not show frequency-dependent reduction. In a small fraction of cells, there was a component of calcium-dependent potassium current that showed frequency-dependent reduction, but the contribution to overall potassium current reduction was almost always much smaller than that of Kv3-mediated current. These results show that Kv3 channels make a major contribution to spike repolarization in small-diameter DRG neurons and undergo frequency-dependent reduction, leading to spike broadening at moderate firing frequencies. Spike broadening from frequency-dependent reduction in Kv3 current could mitigate the frequency-dependent decreases in conduction velocity typical of C-fiber axons.SIGNIFICANCE STATEMENT Small-diameter dorsal root ganglia (DRG) neurons mediating nociception and other sensory modalities express many types of potassium channels, but how they combine to control firing patterns and conduction is not well understood. We found that action potentials of small-diameter rat DRG neurons showed spike broadening at frequencies as low as 1 Hz and that spike broadening resulted predominantly from frequency-dependent inactivation of Kv3 channels. Spike width helps to control transmitter release, conduction velocity, and firing patterns and understanding the role of particular potassium channels can help to guide new pharmacological strategies for targeting pain-sensing neurons selectively.


Assuntos
Potenciais de Ação/fisiologia , Capsaicina/farmacologia , Gânglios Espinais/fisiologia , Neurônios/fisiologia , Canais de Potássio Shaw/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Feminino , Gânglios Espinais/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Long-Evans , Canais de Potássio Shaw/antagonistas & inibidores
6.
Amino Acids ; 49(11): 1895-1906, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28900735

RESUMO

The effects of 4-chloro-3-nitro-N-butylbenzenesulfonamide (SMD2) on KV3.1 channels, heterologous expressed in L-929 cells, were studied with the whole cell patch-clamp technique. SMD2 blocks KV3.1 in a reversible and use-dependent manner, with IC50 around 10 µM, and a Hill coefficient around 2. Although the conductance vs. voltage relationship in control condition can be described by a single Boltzmann function, two terms are necessary to describe the data in the presence of SMD2. The activation and deactivation time constants are weakly voltage dependent both for control and in the presence of SMD2. SMD2 does not change the channel selectivity and tail currents show a typical crossover phenomenon. The time course of inactivation has a fast and a slow component, and SMD2 significantly decreased their values. Steady-state inactivation is best described by a Boltzmann equation with V 1/2 (the voltage where the probability to find the channels in the inactivated state is 50%) and K (slope factor) equals to -22.9 ± 1.5 mV and 5.3 ± 0.9 mV for control, and -30.3 ± 1.3 mV and 6 ± 0.8 mV for SMD2, respectively. The action of SMD2 is enhanced by high frequency stimulation, and by the time the channel stays open. Taken together, our results suggest that SMD2 blocks the open conformation of KV3.1. From a pharmacological and therapeutic point of view, N-alkylsulfonamides may constitute a new class of pharmacological modulators of KV3.1.


Assuntos
Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Shaw/efeitos dos fármacos , Sulfonamidas/farmacologia , Potenciais de Ação/fisiologia , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Fibroblastos , Camundongos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacocinética , Canais de Potássio Shaw/metabolismo , Canais de Potássio Shaw/fisiologia , Sulfonamidas/metabolismo , Sulfonamidas/farmacocinética
7.
Neurobiol Aging ; 54: 187-198, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28390823

RESUMO

Astrocyte dysfunction emerges early in Alzheimer's disease (AD) and may contribute to its pathology and progression. Recently, the voltage gated potassium channel KV3.4 subunit, which underlies the fast-inactivating K+ currents, has been recognized to be relevant for AD pathogenesis and is emerging as a new target candidate for AD. In the present study, we investigated both in in vitro and in vivo models of AD the expression and functional activity of KV3.4 potassium channel subunits in astrocytes. In primary astrocytes our biochemical, immunohistochemical, and electrophysiological studies demonstrated a time-dependent upregulation of KV3.4 expression and functional activity after exposure to amyloid-ß (Aß) oligomers. Consistently, astrocytic KV3.4 expression was upregulated in the cerebral cortex, hippocampus, and cerebellum of 6-month-old Tg2576 mice. Further, confocal triple labeling studies revealed that in 6-month-old Tg2576 mice, KV3.4 was intensely coexpressed with Aß in nonplaque associated astrocytes. Interestingly, in the cortical and hippocampal regions of 12-month-old Tg2576 mice, plaque-associated astrocytes much more intensely expressed KV3.4 subunits, but not Aß. More important, we evidenced that the selective knockdown of KV3.4 expression significantly downregulated both glial fibrillary acidic protein levels and Aß trimers in the brain of 6-month-old Tg2576 mice. Collectively, our results demonstrate that the expression and function of KV3.4 channel subunits are precociously upregulated in cultured astrocytes exposed to Aß oligomers and in reactive astrocytes of AD Tg2576 mice.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/efeitos adversos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Expressão Gênica , Canais de Potássio Shaw/genética , Canais de Potássio Shaw/metabolismo , Regulação para Cima , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fragmentos de Peptídeos/efeitos adversos , Fragmentos de Peptídeos/metabolismo , Ratos Wistar , Canais de Potássio Shaw/fisiologia , Regulação para Cima/efeitos dos fármacos
8.
Neural Plast ; 2016: 8782518, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27379187

RESUMO

In the last years it has been increasingly clear that KV-channel activity modulates neurotransmitter release. The subcellular localization and composition of potassium channels are crucial to understanding its influence on neurotransmitter release. To investigate the role of KV in corticostriatal synapses modulation, we combined extracellular recording of population-spike and pharmacological blockage with specific and nonspecific blockers to identify several families of KV channels. We induced paired-pulse facilitation (PPF) and studied the changes in paired-pulse ratio (PPR) before and after the addition of specific KV blockers to determine whether particular KV subtypes were located pre- or postsynaptically. Initially, the presence of KV channels was tested by exposing brain slices to tetraethylammonium or 4-aminopyridine; in both cases we observed a decrease in PPR that was dose dependent. Further experiments with tityustoxin, margatoxin, hongotoxin, agitoxin, dendrotoxin, and BDS-I toxins all rendered a reduction in PPR. In contrast heteropodatoxin and phrixotoxin had no effect. Our results reveal that corticostriatal presynaptic KV channels have a complex stoichiometry, including heterologous combinations KV1.1, KV1.2, KV1.3, and KV1.6 isoforms, as well as KV3.4, but not KV4 channels. The variety of KV channels offers a wide spectrum of possibilities to regulate neurotransmitter release, providing fine-tuning mechanisms to modulate synaptic strength.


Assuntos
Córtex Cerebral/fisiologia , Corpo Estriado/fisiologia , Terminações Pré-Sinápticas/fisiologia , Superfamília Shaker de Canais de Potássio/fisiologia , Canais de Potássio Shaw/fisiologia , Sinapses/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Córtex Cerebral/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Masculino , Bloqueadores dos Canais de Potássio/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/fisiologia , Ratos , Ratos Wistar , Superfamília Shaker de Canais de Potássio/antagonistas & inibidores , Canais de Potássio Shaw/antagonistas & inibidores , Sinapses/efeitos dos fármacos
9.
J Physiol ; 594(16): 4677-84, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-26442672

RESUMO

The voltage-dependent potassium channel subunit Kv3.3 is expressed at high levels in cerebellar Purkinje cells, in auditory brainstem nuclei and in many other neurons capable of firing at high rates. In the cerebellum, it helps to shape the very characteristic complex spike of Purkinje cells. Kv3.3 differs from other closely related channels in that human mutations in the gene encoding Kv3.3 (KCNC3) result in a unique neurodegenerative disease termed spinocerebellar ataxia type 13 (SCA13). This primarily affects the cerebellum, but also results in extracerebellar symptoms. Different mutations produce either early onset SCA13, associated with delayed motor and impaired cognitive skill acquisition, or late onset SCA13, which typically produces cerebellar degeneration in middle age. This review covers the localization and physiological function of Kv3.3 in the central nervous system and how the normal function of the channel is altered by the disease-causing mutations. It also describes experimental approaches that are being used to understand how Kv3.3 mutations are linked to neuronal survival, and to develop strategies for treatment.


Assuntos
Canais de Potássio Shaw/fisiologia , Ataxias Espinocerebelares/congênito , Animais , Humanos , Canais de Potássio Shaw/genética , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/fisiopatologia
10.
J. physiol. biochem ; 71(4): 601-610, dic. 2015.
Artigo em Inglês | IBECS | ID: ibc-145714

RESUMO

TREK-2 (TWIK-related K+ channel-2), a member of two-pore domain potassium (K2P) channel family, tunes cellular excitability via conducting leak or background currents. In TREK-2, the isoforms generated by alternative translation initiation (ATI) mechanism exhibit large divergence in unitary conductance, but similar in selectivity to K+. Up to now, the structural basis for this similarity in ion selectivity is unknown. Here, we report that externally applied Ba2+ inhibits the currents of TREK-2 in a concentration- and time-dependent manner. The blocking effect is blunted by elevated extracellular K+ or mutation of S4 K+ binding site, which suggests that the inhibitory mechanism of Ba2+ is due to its competitive docking properties within the selectivity filter (SF). Next, we demonstrate that all the ATI isoforms exhibit analogous behaviors upon the application of Ba2+ and alteration of extracellular pH (pHo), which acts on the outer position of the SF. These results strongly support the notion that all the ATI isoforms of TREK-2 possess resembled SF conformation in S4 site and the position defined by pHo, which implicates that neither the role of N-terminus (Nt) nor the unitary conductance is associated with SF conformation. Our findings might help to understand the detail gating mechanism of TREK-2 and K2P channels


Assuntos
Isoformas de Proteínas/análise , Iniciação Traducional da Cadeia Peptídica/fisiologia , Canais de Potássio Shaw/fisiologia
11.
Channels (Austin) ; 9(4): 209-17, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26039360

RESUMO

Recently, we reported the isolation of the Kv3.4 current in dorsal root ganglion (DRG) neurons and described dysregulation of this current in a spinal cord injury (SCI) model of chronic pain. These studies strongly suggest that rat Kv3.4 channels are major regulators of excitability in DRG neurons from pups and adult females, where they help determine action potential (AP) repolarization and spiking properties. Here, we characterized the Kv3.4 current in rat DRG neurons from adult males and show that it transfers 40-70% of the total repolarizing charge during the AP across all ages and sexes. Following SCI, we also found remodeling of the repolarizing currents during the AP. In the light of these studies, homomeric Kv3.4 channels expressed in DRG nociceptors are emerging novel targets that may help develop new approaches to treat neuropathic pain.


Assuntos
Gânglios Espinais/fisiologia , Neurônios/fisiologia , Nociceptores/fisiologia , Canais de Potássio Shaw/fisiologia , Potenciais de Ação/genética , Potenciais de Ação/fisiologia , Animais , Células Cultivadas , Feminino , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Expressão Gênica , Masculino , Neurônios/metabolismo , Nociceptores/metabolismo , Técnicas de Patch-Clamp , Interferência de RNA , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Potássio Shaw/genética
12.
Eur J Pharmacol ; 758: 164-70, 2015 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-25845309

RESUMO

The effect of two positive modulators, RE1 and EX15, on the voltage-gated K(+) channel Kv3.1 was investigated using the whole-cell patch-clamp technique on HEK293 cells expressing Kv3.1a. RE1 and EX15 increased the Kv3.1 currents in a concentration-dependent manner with an EC50 value of 4.5 and 1.3µM, respectively. However, high compound concentrations caused an inhibition of the Kv3.1 current. The compound-induced activation of Kv3.1 channels showed a profound hyperpolarized shift in activation kinetics. 30µM RE1 shifted V1/2 from 5.63±0.31mV to -9.71±1.00mV and 10µM EX15 induced a shift from 10.77±0.32mV to -15.11±1.57mV. The activation time constant (Tauact) was reduced for both RE1 and EX15, with RE1 being the fastest activator. The deactivation time constant (Taudeact) was also markedly reduced for both RE1 and EX15, with EX15 inducing the most prominent effect. Furthermore, subjected to depolarizing pulses at 30Hz, both compounds were showing a use-dependent effect resulting in a reduction of the compound-mediated effect. However, during these conditions, RE1- and EX15-modified current amplitudes still exceeded the control condition amplitudes by up to 200%. In summary, the present study introduces the first detailed biophysical characterization of two new Kv3.1 channel modifying compounds with different modulating properties.


Assuntos
Canais de Potássio Shaw/agonistas , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Hidantoínas/farmacologia , Cinética , Piridinas/farmacologia , Canais de Potássio Shaw/fisiologia
13.
Nat Genet ; 47(1): 39-46, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25401298

RESUMO

Progressive myoclonus epilepsies (PMEs) are a group of rare, inherited disorders manifesting with action myoclonus, tonic-clonic seizures and ataxia. We sequenced the exomes of 84 unrelated individuals with PME of unknown cause and molecularly solved 26 cases (31%). Remarkably, a recurrent de novo mutation, c.959G>A (p.Arg320His), in KCNC1 was identified as a new major cause for PME. Eleven unrelated exome-sequenced (13%) and two affected individuals in a secondary cohort (7%) had this mutation. KCNC1 encodes KV3.1, a subunit of the KV3 voltage-gated potassium ion channels, which are major determinants of high-frequency neuronal firing. Functional analysis of the Arg320His mutant channel showed a dominant-negative loss-of-function effect. Ten cases had pathogenic mutations in known PME-associated genes (NEU1, NHLRC1, AFG3L2, EPM2A, CLN6 and SERPINI1). Identification of mutations in PRNP, SACS and TBC1D24 expand their phenotypic spectra to PME. These findings provide insights into the molecular genetic basis of PME and show the role of de novo mutations in this disease entity.


Assuntos
Mutação de Sentido Incorreto , Epilepsias Mioclônicas Progressivas/genética , Mutação Puntual , Canais de Potássio Shaw/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sequência de Bases , Proteínas de Transporte/genética , Sequência Conservada , Exoma , Feminino , Proteínas Ativadoras de GTPase , Genes Dominantes , Proteínas de Choque Térmico/genética , Humanos , Masculino , Proteínas de Membrana , Dados de Sequência Molecular , Proteínas do Tecido Nervoso , Linhagem , Proteínas Priônicas , Príons/genética , Conformação Proteica , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Canais de Potássio Shaw/fisiologia , Especificidade da Espécie
14.
J Neurosci ; 34(19): 6611-23, 2014 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24806686

RESUMO

The discrete arrangement of voltage-gated K(+) (Kv) channels in axons may impart functional advantages in action potential (AP) signaling yet, in compact cell types, the organization of Kv channels is poorly understood. We find that in cerebellar stellate cell interneurons of mice, the composition and influence of Kv channels populating the axon is diverse and depends on location allowing axonal compartments to differentially control APs in a local manner. Kv1 channels determine AP repolarization at the spike initiation site but not at more distal sites, limiting the expression of use-dependent spike broadening to the most proximal axon region, likely a key attribute informing spiking phenotype. Local control of AP repolarization at presynaptic boutons depends on Kv3 channels keeping APs brief, thus limiting Ca(2+) influx and synaptic strength. These observations suggest that AP repolarization is tuned by the local influence of distinct Kv channel types, and this organization enhances the functional segregation of axonal compartments.


Assuntos
Axônios/fisiologia , Cerebelo/fisiologia , Interneurônios/fisiologia , Canais de Potássio/fisiologia , Terminações Pré-Sinápticas/fisiologia , Receptores Pré-Sinápticos/fisiologia , Animais , Cerebelo/citologia , Fenômenos Eletrofisiológicos , Feminino , Corantes Fluorescentes , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Técnicas de Patch-Clamp , Receptores de AMPA/fisiologia , Receptores de GABA-A/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Superfamília Shaker de Canais de Potássio/fisiologia , Canais de Potássio Shaw/fisiologia , Transmissão Sináptica/fisiologia
15.
FEBS Lett ; 588(1): 86-91, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24291260

RESUMO

The Kv3.1 channel plays a crucial role in regulating the high-frequency firing properties of neurons. Here, we determined whether Src regulates the subcellular distributions of the Kv3.1b channel. Co-expression of active Src induced a dramatic redistribution of Kv3.1b to the endoplasmic reticulum. Furthermore, co-expression of the Kv3.1b channel with active Src induced a remarkable decrease in the pool of Kv3.1b at the cell surface. Moreover, the co-expression of active Src results in a significant decrease in the peak current densities of the Kv3.1b channel, and a substantial alteration in the voltage dependence of its steady-state inactivation. Taken together, these results indicate that Src kinase may play an important role in regulating membrane trafficking of Kv3.1b channels.


Assuntos
Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Canais de Potássio Shaw/metabolismo , Animais , Western Blotting , Células COS , Membrana Celular/fisiologia , Chlorocebus aethiops , Células HEK293 , Humanos , Potenciais da Membrana/fisiologia , Camundongos , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Técnicas de Patch-Clamp , Transporte Proteico , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Ratos , Canais de Potássio Shaw/genética , Canais de Potássio Shaw/fisiologia
16.
PLoS One ; 8(10): e77105, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24204745

RESUMO

Computational modeling has emerged as an indispensable approach to resolve and predict the intricate interplay among the many ion channels underlying neuronal excitability. However, simulation results using the classic formula-based Hodgkin-Huxley (H-H) model or the superior Markov kinetic model of ion channels often deviate significantly from native cellular signals despite using carefully measured parameters. Here we found that the filters of patch-clamp amplifier not only delayed the signals, but also introduced ringing, and that the residual series resistance in experiments altered the command voltages, which had never been fully eliminated by improving the amplifier itself. To remove all the above errors, a virtual device with the parameters exactly same to that of amplifier was introduced into Markov kinetic modeling so as to establish a null-deviation model. We demonstrate that our novel null-deviation approach fully restores the native gating-kinetics of ion-channels with the data recorded at any condition, and predicts spike waveform and firing patterns clearly distinctive from those without correction.


Assuntos
Ativação do Canal Iônico/fisiologia , Canais Iônicos/fisiologia , Modelos Neurológicos , Neurônios/fisiologia , Potenciais de Ação/fisiologia , Algoritmos , Animais , Células CHO , Simulação por Computador , Cricetinae , Cricetulus , Células HEK293 , Humanos , Canais Iônicos/metabolismo , Potenciais da Membrana/fisiologia , Neurônios/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio Shaw/metabolismo , Canais de Potássio Shaw/fisiologia
17.
PLoS One ; 8(10): e76749, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24116147

RESUMO

Normal sound localization requires precise comparisons of sound timing and pressure levels between the two ears. The primary localization cues are interaural time differences, ITD, and interaural level differences, ILD. Voltage-gated potassium channels, including Kv3.3, are highly expressed in the auditory brainstem and are thought to underlie the exquisite temporal precision and rapid spike rates that characterize brainstem binaural pathways. An autosomal dominant mutation in the gene encoding Kv3.3 has been demonstrated in a large Filipino kindred manifesting as spinocerebellar ataxia type 13 (SCA13). This kindred provides a rare opportunity to test in vivo the importance of a specific channel subunit for human hearing. Here, we demonstrate psychophysically that individuals with the mutant allele exhibit profound deficits in both ITD and ILD sensitivity, despite showing no obvious impairment in pure-tone sensitivity with either ear. Surprisingly, several individuals exhibited the auditory deficits even though they were pre-symptomatic for SCA13. We would expect that impairments of binaural processing as great as those observed in this family would result in prominent deficits in localization of sound sources and in loss of the "spatial release from masking" that aids in understanding speech in the presence of competing sounds.


Assuntos
Mutação , Canais de Potássio Shaw/genética , Transdução de Sinais/genética , Localização de Som , Degenerações Espinocerebelares/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Limiar Auditivo , Criança , Sinais (Psicologia) , Feminino , Genes Dominantes , Audição/genética , Audição/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Canais de Potássio Shaw/fisiologia , Transdução de Sinais/fisiologia , Ataxias Espinocerebelares/congênito , Degenerações Espinocerebelares/fisiopatologia , Adulto Jovem
18.
J Neurophysiol ; 110(5): 1097-106, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23741043

RESUMO

The neuropeptide vasoactive intestinal peptide (VIP) is expressed at high levels in the neurons of the suprachiasmatic nucleus (SCN). While VIP is known to be important to the input and output pathways from the SCN, the physiological effects of VIP on electrical activity of SCN neurons are not well known. Here the impact of VIP on firing rate of SCN neurons was investigated in mouse slice cultures recorded during the night. The application of VIP produced an increase in electrical activity in SCN slices that lasted several hours after treatment. This is a novel mechanism by which this peptide can produce long-term changes in central nervous system physiology. The increase in action potential frequency was blocked by a VIP receptor antagonist and lost in a VIP receptor knockout mouse. In addition, inhibitors of both the Epac family of cAMP binding proteins and cAMP-dependent protein kinase (PKA) blocked the induction by VIP. The persistent increase in spike rate following VIP application was not seen in SCN neurons from mice deficient in Kv3 channel proteins and was dependent on the clock protein PER1. These findings suggest that VIP regulates the long-term firing rate of SCN neurons through a VIPR2-mediated increase in the cAMP pathway and implicate the fast delayed rectifier (FDR) potassium currents as one of the targets of this regulation.


Assuntos
Neurônios/fisiologia , Núcleo Supraquiasmático/fisiologia , Peptídeo Intestinal Vasoativo/farmacologia , Animais , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Proteínas Circadianas Period/metabolismo , Canais de Potássio Shaw/genética , Canais de Potássio Shaw/fisiologia , Transdução de Sinais , Núcleo Supraquiasmático/efeitos dos fármacos
19.
J Physiol ; 591(10): 2579-91, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23478135

RESUMO

Adult neural stem/precursor cells (NPCs) play a pivotal role in neuronal plasticity throughout life. Among ion channels identified in adult NPCs, voltage-gated delayed rectifier K(+) (KDR) channels are dominantly expressed. However, the KDR channel subtype and its physiological role are still undefined. We used real-time quantitative RT-PCR and gene knockdown techniques to identify a major functional KDR channel subtype in adult NPCs. Dominant mRNA expression of Kv3.1, a high voltage-gated KDR channel, was quantitatively confirmed. Kv3.1 gene knockdown with specific small interfering RNAs (siRNA) for Kv3.1 significantly inhibited Kv3.1 mRNA expression by 63.9% (P < 0.001) and KDR channel currents by 52.2% (P < 0.001). This indicates that Kv3.1 is the subtype responsible for producing KDR channel outward currents. Resting membrane properties, such as resting membrane potential, of NPCs were not affected by Kv3.1 expression. Kv3.1 knockdown with 300 nm siRNA inhibited NPC growth (increase in cell numbers) by 52.9% (P < 0.01). This inhibition was attributed to decreased cell proliferation, not increased cell apoptosis. We also established a convenient in vitro imaging assay system to evaluate NPC differentiation using NPCs from doublecortin-green fluorescent protein transgenic mice. Kv3.1 knockdown also significantly reduced neuronal differentiation by 31.4% (P < 0.01). We have demonstrated that Kv3.1 is a dominant functional KDR channel subtype expressed in adult NPCs and plays key roles in NPC proliferation and neuronal lineage commitment during differentiation.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Neurais/citologia , Canais de Potássio Shaw/fisiologia , Células-Tronco Adultas/fisiologia , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/fisiologia , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética
20.
J Physiol ; 591(10): 2491-507, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23420657

RESUMO

Zinc, a divalent heavy metal ion and an essential mineral for life, regulates synaptic transmission and neuronal excitability via ion channels. However, its binding sites and regulatory mechanisms are poorly understood. Here, we report that Kv3 channel assembly, localization and activity are regulated by zinc through different binding sites. Local perfusion of zinc reversibly reduced spiking frequency of cultured neurons most likely by suppressing Kv3 channels. Indeed, zinc inhibited Kv3.1 channel activity and slowed activation kinetics, independent of its site in the N-terminal T1 domain. Biochemical assays surprisingly identified a novel zinc-binding site in the Kv3.1 C-terminus, critical for channel activity and axonal targeting, but not for the zinc inhibition. Finally, mutagenesis revealed an important role of the junction between the first transmembrane (TM) segment and the first extracellular loop in sensing zinc. Its mutant enabled fast spiking with relative resistance to the zinc inhibition. Therefore, our studies provide novel mechanistic insights into the multifaceted regulation of Kv3 channel activity and localization by divalent heavy metal ions.


Assuntos
Neurônios/fisiologia , Canais de Potássio Shaw/fisiologia , Zinco/farmacologia , Animais , Sítios de Ligação , Células Cultivadas , Cerebelo/citologia , Cerebelo/embriologia , Embrião de Mamíferos , Células HEK293 , Hipocampo/citologia , Hipocampo/embriologia , Humanos , Transporte Proteico , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...