Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurophysiol ; 125(5): 1954-1972, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33852808

RESUMO

Temporal coding precision of bushy cells in the ventral cochlear nucleus (VCN), critical for sound localization and communication, depends on the generation of rapid and temporally precise action potentials (APs). Voltage-gated potassium (Kv) channels are critically involved in this. The bushy cells in rat VCN express Kv1.1, 1.2, 1.3, 1.6, 3.1, 4.2, and 4.3 subunits. The Kv1.1 subunit contributes to the generation of a temporally precise single AP. However, the understanding of the functions of other Kv subunits expressed in the bushy cells is limited. Here, we investigated the functional diversity of Kv subunits concerning their contributions to temporal coding. We characterized the electrophysiological properties of the Kv channels with different subunits using whole cell patch-clamp recording and pharmacological methods. The neuronal firing pattern changed from single to multiple APs only when the Kv1.1 subunit was blocked. The Kv subunits, including the Kv1.1, 1.2, 1.6, or 3.1, were involved in enhancing temporal coding by lowering membrane excitability, shortening AP latencies, reducing jitter, and regulating AP kinetics. Meanwhile, all the Kv subunits contributed to rapid repolarization and sharpening peaks by narrowing half-width and accelerating fall rate, and the Kv1.1 subunit also affected the depolarization of AP. The Kv1.1, 1.2, and 1.6 subunits endowed bushy cells with a rapid time constant and a low input resistance of membrane for enhancing spike timing precision. The present results indicate that the Kv channels differentially affect intrinsic membrane properties to optimize the generation of rapid and reliable APs for temporal coding.NEW & NOTEWORTHY This study investigates the roles of Kv channels in effecting precision using electrophysiological and pharmacological methods in bushy cells. Different Kv channels have varying electrophysiological characteristics, which contribute to the interplay between changes in the membrane properties and regulation of neuronal excitability which then improve temporal coding. We conclude that the Kv channels are specialized to promote the precise and rapid coding of acoustic input by optimizing the generation of reliable APs.


Assuntos
Potenciais de Ação/fisiologia , Núcleo Coclear/fisiologia , Neurônios/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Feminino , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.1/fisiologia , Canal de Potássio Kv1.2/antagonistas & inibidores , Canal de Potássio Kv1.2/fisiologia , Canal de Potássio Kv1.6/antagonistas & inibidores , Canal de Potássio Kv1.6/fisiologia , Masculino , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley
2.
J Assoc Res Otolaryngol ; 20(6): 565-577, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31410614

RESUMO

The submillisecond acuity for detecting rapid spatial and temporal fluctuations in acoustic stimuli observed in humans and laboratory animals depends in part on select groups of auditory neurons that preserve synchrony from the ears to the binaural nuclei in the brainstem. These fibers have specialized synapses and axons that use a low-threshold voltage-activated outward current, IKL, conducted through Kv1 potassium ion channels. These are in turn coupled with HCN channels that express a mixed cation inward mixed current, IH, to support precise synchronized firing. The behavioral evidence is that their respective Kcna1 or HCN1 genes are absent in adult mice; the results are weak startle reflexes, slow responding to noise offsets, and poor sound localization. The present behavioral experiments were motivated by an in vitro study reporting increased IKL in an auditory nucleus in Kcna2-/- mice lacking the Kv1.2 subunit, suggesting that Kcna2-/- mice might perform better than Kcna2+/+ mice. Because Kcna2-/- mice have only a 17-18-day lifespan, we compared both preweanling Kcna2-/- vs. Kcna2+/+ mice and Kcna1-/- vs. Kcna1+/+ mice at P12-P17/18; then, the remaining mice were tested at P23/P25. Both null mutant strains had a stunted physique, but the Kcna1-/- mice had severe behavioral deficits while those in Kcna2-/- mice were relatively few and minor. The in vitro increase of IKL could have resulted from Kv1.1 subunits substituting for Kv1.2 units and the loss of the inhibitory "managerial" effect of Kv1.2 on Kv1.1. However, any increased neuronal synchronicity that accompanies increased IKL may not have been enough to affect behavior. All mice performed unusually well on the early spatial tests, but then, they fell towards adult levels. This unexpected effect may reflect a shift from summated independent monaural pathways to integrated binaural processing, as has been suggested for similar observations for human infants.


Assuntos
Canal de Potássio Kv1.1/fisiologia , Canal de Potássio Kv1.2/fisiologia , Localização de Som , Estimulação Acústica , Animais , Feminino , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.2/genética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Atividade Motora , Ruído , Reflexo de Sobressalto , Desmame
3.
Toxins (Basel) ; 11(6)2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-31212818

RESUMO

MiDCA1, a phospholipase A2 (PLA2) neurotoxin isolated from Micrurus dumerilii carinicauda coral snake venom, inhibited a major component of voltage-activated potassium (Kv) currents (41 ± 3% inhibition with 1 µM toxin) in mouse cultured dorsal root ganglion (DRG) neurons. In addition, the selective Kv2.1 channel blocker guangxitoxin (GxTx-1E) and MiDCA1 competitively inhibited the outward potassium current in DRG neurons. MiDCA1 (1 µM) reversibly inhibited the Kv2.1 current by 55 ± 8.9% in a Xenopus oocyte heterologous system. The toxin showed selectivity for Kv2.1 channels over all the other Kv channels tested in this study. We propose that Kv2.1 channel blockade by MiDCA1 underlies the toxin's action on acetylcholine release at mammalian neuromuscular junctions.


Assuntos
Cobras Corais , Venenos Elapídicos/toxicidade , Canal de Potássio Kv1.2/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/toxicidade , Animais , Células Cultivadas , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Canal de Potássio Kv1.2/genética , Canal de Potássio Kv1.2/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Oócitos/fisiologia , Fosfolipases A2 , Xenopus
4.
Physiol Rep ; 7(12): e14147, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31222975

RESUMO

Expression of Kv1.2 within Kv1.x potassium channel complexes is critical in maintaining appropriate neuronal excitability and determining the threshold for action potential firing. This is attributed to the interaction of Kv1.2 with a hitherto unidentified protein that confers bimodal channel activation gating, allowing neurons to adapt to repetitive trains of stimulation and protecting against hyperexcitability. One potential protein candidate is the sigma-1 receptor (Sig-1R), which regulates other members of the Kv1.x channel family; however, the biophysical nature of the interaction between Sig-1R and Kv1.2 has not been elucidated. We hypothesized that Sig-1R may regulate Kv1.2 and may further act as the unidentified modulator of Kv1.2 activation. In transiently transfected HEK293 cells, we found that ligand activation of the Sig-1R modulates Kv1.2 current amplitude. More importantly, Sig-1R interacts with Kv1.2 in baseline conditions to influence bimodal activation gating. These effects are abolished in the presence of the auxiliary subunit Kvß2 and when the Sig-1R mutation underlying ALS16 (Sig-1R-E102Q), is expressed. These data suggest that Kvß2 occludes the interaction of Sig-1R with Kv1.2, and that E102 may be a residue critical for Sig-1R modulation of Kv1.2. The results of this investigation describe an important new role for Sig-1R in the regulation of neuronal excitability and introduce a novel mechanism of pathophysiology in Sig-1R dysfunction.


Assuntos
Canal de Potássio Kv1.2/fisiologia , Receptores sigma/fisiologia , Células Cultivadas , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos/fisiologia , Células HEK293 , Humanos , Ativação do Canal Iônico/fisiologia , Canal de Potássio Kv1.2/efeitos dos fármacos , Canal de Potássio Kv1.2/metabolismo , Técnicas de Patch-Clamp/métodos , Fenazocina/análogos & derivados , Fenazocina/antagonistas & inibidores , Fenazocina/farmacologia , Receptores sigma/agonistas , Receptores sigma/metabolismo , Superfamília Shaker de Canais de Potássio/fisiologia , Receptor Sigma-1
5.
PLoS One ; 12(4): e0176078, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28426823

RESUMO

Current through the σ-pore was first detected in hKv1.3_V388C channels, where the V388C mutation in hKv1.3 channels opened a new pathway (σ-pore) behind the central α-pore. Typical for this mutant channel was inward current at potentials more negative than -100 mV when the central α-pore was closed. The α-pore blockers such as TEA+ and peptide toxins (CTX, MTX) could not reduce current through the σ-pore of hKv1.3_V388C channels. This new pathway would proceed in parallel to the α-pore in the S6-S6 interface gap. To see whether this phenomenon is restricted to hKv1.3 channels we mutated hKv1.2 at the homologue position (hKv1.2_V370C). By overexpression of hKv1.2_V370C mutant channels in COS-7 cells we could show typical σ-currents. The electrophysiological properties of the σ-pore in hKv1.3_V388C and hKv1.2_V370C mutant channels were similar. The σ-pore of hKv1.2_V370C channels was most permeable to Na+ and Li+ whereas Cl- and protons did not influence current through the σ-pore. Tetraethylammonium (TEA+), charybdotoxin (CTX) and maurotoxin (MTX), known α-pore blockers, could not reduce current through the σ-pore of hKv1.2_V370C channels. Taken together we conclude that the observation of σ-pore currents is not restricted to Kv1.3 potassium channels but can also be observed in a closely related potassium channel. This finding could have implications in the treatment of different ion channel diseases linked to mutations of the respective channels in regions close to homologue position investigated by us.


Assuntos
Canal de Potássio Kv1.2/fisiologia , Mutação , Substituição de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Cisteína/química , Canal de Potássio Kv1.2/química , Canal de Potássio Kv1.2/genética , Valina/química
6.
Physiol Res ; 66(3): 539-544, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28248535

RESUMO

Bis(3)-tacrine is a dimeric AChE inhibitor derived from tacrine with a potential to treat Alzheimer's disease. It was recently been reported to act as a fast off-rate antagonist of NMDA receptors with moderate affinity. In the present study, we aimed to explore whether bis(3)-tacrine could modulate the function of native sustained potassium current in cultured rat hippocampal neurons using whole-cell patch-clamp technique. We found that bis(3)-tacrine inhibited the amplitude of sustained potassium current in a reversible and concentration-dependent manner, with a potency two orders of magnitude higher than that of tacrine. The inhibition was voltage-independent between 0 to +60 mV. The IC(50) values for bis(3)-tacrine and tacrine inhibition of sustained potassium current were 0.45+/-0.07 and 50.5+/-4.8 microM, respectively. I-V curves showed a more potent inhibition of sustained potassium current by bis(3)-tacrine (1 microM) compared to tacrine at the same concentration. Bis(3)-tacrine hyperpolarized the activation curve of the current by 11.2 mV, albeit leaving the steady-state inactivation of the current unaffected.


Assuntos
Inibidores da Colinesterase/farmacologia , Hipocampo/fisiologia , Canal de Potássio Kv1.2/antagonistas & inibidores , Canal de Potássio Kv1.2/fisiologia , Neurônios/fisiologia , Tacrina/farmacologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Estimulação Elétrica/métodos , Hipocampo/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
7.
Behav Brain Res ; 326: 13-21, 2017 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-28223099

RESUMO

In the current study we set out to determine the effects of morpholino oligonucleotide (MO) knock-down of kcna2 on sleep-wake cycles in zebrafish. The results were compared to a non-overlapping MO injection, Dec2, who's mutant is also linked with a short sleep phenotype. Four groups of fish were used in the experiment: naïve fish, and fish injected with either control, kcna2, or Dec2 MO. All groups underwent 24-h behavioral monitoring of sleep-wake cycles at four and seven days-post-fertilization (dpf). First, we established an immobility dependent, sleep related, increase in arousal thresholds at both 4 and 7 dpf. Secondly, we show that kcna2 MO injected fish exhibit significantly less sleep behavior than controls and naïve fish, whereas Dec2 MO injections had similar but less severe effects. Finally, using kcna2 MO injected fish only, we turn to local field recordings at the level of the telencephalon and tectum opticum and rule out that the knock-down resulted in a non-specific increase in neural excitability that would mask sleep behavior.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Comportamento Animal/fisiologia , Encéfalo/fisiologia , Canal de Potássio Kv1.2/fisiologia , Larva/fisiologia , Sono/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/fisiologia , Fatores Etários , Animais , Animais Geneticamente Modificados , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fenômenos Eletrofisiológicos , Canal de Potássio Kv1.2/genética , Larva/genética , Morfolinos , Sono/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
8.
J Physiol ; 593(16): 3617-43, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26047212

RESUMO

KEY POINTS: We investigated the cellular mechanisms underlying mossy fibre-induced heterosynaptic long-term potentiation of perforant path (PP) inputs to CA3 pyramidal cells. Here we show that this heterosynaptic potentiation is mediated by downregulation of Kv1.2 channels. The downregulation of Kv1.2 preferentially enhanced PP-evoked EPSPs which occur at distal apical dendrites. Such enhancement of PP-EPSPs required activation of dendritic Na(+) channels, and its threshold was lowered by downregulation of Kv1.2. Our results may provide new insights into the long-standing question of how mossy fibre inputs constrain the CA3 network to sparsely represent direct cortical inputs. ABSTRACT: A short high frequency stimulation of mossy fibres (MFs) induces long-term potentiation (LTP) of direct cortical or perforant path (PP) synaptic inputs in hippocampal CA3 pyramidal cells (CA3-PCs). However, the cellular mechanism underlying this heterosynaptic modulation remains elusive. Previously, we reported that repetitive somatic firing at 10 Hz downregulates Kv1.2 in the CA3-PCs. Here, we show that MF inputs induce similar somatic firing and downregulation of Kv1.2 in the CA3-PCs. The effect of Kv1.2 downregulation was specific to PP synaptic inputs that arrive at distal apical dendrites. We found that the somatodendritic expression of Kv1.2 is polarized to distal apical dendrites. Compartmental simulations based on this finding suggested that passive normalization of synaptic inputs and polarized distributions of dendritic ionic channels may facilitate the activation of dendritic Na(+) channels preferentially at distal apical dendrites. Indeed, partial block of dendritic Na(+) channels using 10 nm tetrodotoxin brought back the enhanced PP-evoked excitatory postsynaptic potentials (PP-EPSPs) to the baseline level. These results indicate that activity-dependent downregulation of Kv1.2 in CA3-PCs mediates MF-induced heterosynaptic LTP of PP-EPSPs by facilitating activation of Na(+) channels at distal apical dendrites.


Assuntos
Região CA3 Hipocampal/fisiologia , Canal de Potássio Kv1.2/fisiologia , Células Piramidais/fisiologia , Animais , Células Cultivadas , Potenciais Pós-Sinápticos Excitadores , Feminino , Canal de Potássio Kv1.2/genética , Potenciação de Longa Duração , Masculino , Camundongos Knockout , Fibras Musgosas Hipocampais/fisiologia , Via Perfurante , Ratos Sprague-Dawley , Transmissão Sináptica
9.
Proc Natl Acad Sci U S A ; 111(19): E1950-9, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24782544

RESUMO

Voltage sensor domains (VSDs) regulate ion channels and enzymes by transporting electrically charged residues across a hydrophobic VSD constriction called the gating pore or hydrophobic plug. How the gating pore controls the gating charge movement presently remains debated. Here, using saturation mutagenesis and detailed analysis of gating currents from gating pore mutations in the Shaker Kv channel, we identified statistically highly significant correlations between VSD function and physicochemical properties of gating pore residues. A necessary small residue at position S240 in S1 creates a "steric gap" that enables an intracellular access pathway for the transport of the S4 Arg residues. In addition, the stabilization of the depolarized VSD conformation, a hallmark for most Kv channels, requires large side chains at positions F290 in S2 and F244 in S1 acting as "molecular clamps," and a hydrophobic side chain at position I237 in S1 acting as a local intracellular hydrophobic barrier. Finally, both size and hydrophobicity of I287 are important to control the main VSD energy barrier underlying transitions between resting and active states. Taken together, our study emphasizes the contribution of several gating pore residues to catalyze the gating charge transfer. This work paves the way toward understanding physicochemical principles underlying conformational dynamics in voltage sensors.


Assuntos
Ativação do Canal Iônico/fisiologia , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.1/fisiologia , Canal de Potássio Kv1.2/genética , Canal de Potássio Kv1.2/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos/fisiologia , Animais , Cristalografia por Raios X , Humanos , Interações Hidrofóbicas e Hidrofílicas , Cinética , Canal de Potássio Kv1.1/química , Canal de Potássio Kv1.2/química , Modelos Químicos , Dados de Sequência Molecular , Oócitos/fisiologia , Técnicas de Patch-Clamp , Estrutura Secundária de Proteína/fisiologia , Xenopus laevis
10.
Naunyn Schmiedebergs Arch Pharmacol ; 387(5): 469-76, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24682423

RESUMO

The ultrarapidly activating delayed rectifier current, I(Kur), is a main determinant of atrial repolarization in humans. I(Kur) and the underlying ion channel complex Kv1.5/Kvß1.2 are negatively regulated by protein kinase C. However, the exact mode of action is only incompletely understood. We therefore analyzed isoenzyme-specific regulation of the Kv1.5/Kvß1.2 ion channel complex by PKC. Cloned ion channel subunits were heterologously expressed in Xenopus oocytes, and measurements were performed using the double-electrode voltage-clamp technique. Activation of PKC with phorbol 12-myristate 13-acetate (PMA) resulted in a strong reduction of Kv1.5/Kvß1.2 current. This effect could be prevented using the PKC inhibitor staurosporine. Using the bisindolylmaleimide Ro-31-8220 as an inhibitor and ingenol as an activator of the conventional PKC isoforms, we were able to show that the Kv1.5/Kvß1.2 ion channel complex is mainly regulated by conventional isoforms. Whereas pharmacological inhibition of PKCα with HBDDE did not attenuate the PMA-induced effect, current reduction could be prevented using inhibitors of PKCß. Here, we show the isoform ßII plays a central role in the PKC-dependent regulation of Kv1.5/Kvß1.2 channels. These results add to the current understanding of isoenzyme-selective regulation of cardiac ion channels by protein kinases.


Assuntos
Canal de Potássio Kv1.2/fisiologia , Canal de Potássio Kv1.5/fisiologia , Miocárdio/metabolismo , Proteína Quinase C beta/fisiologia , Animais , Humanos , Isoenzimas/fisiologia , Ligação Proteica , Acetato de Tetradecanoilforbol/farmacologia , Xenopus
11.
Anesthesiology ; 121(2): 409-17, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24739997

RESUMO

Chronic pain, a common clinical symptom, is often treated inadequately or ineffectively in part due to the incomplete understanding of molecular mechanisms that initiate and maintain this disorder. Newly identified noncoding RNAs govern gene expression. Recent studies have shown that peripheral noxious stimuli drive expressional changes in noncoding RNAs and that these changes are associated with pain hypersensitivity under chronic pain conditions. This review first presents current evidence for the peripheral inflammation/nerve injury-induced change in the expression of two types of noncoding RNAs, microRNAs, and Kcna2 antisense RNA, in pain-related regions, particularly in the dorsal root ganglion. The authors then discuss how peripheral noxious stimuli induce such changes. The authors finally explore potential mechanisms of how expressional changes in dorsal root ganglion microRNAs and Kcna2 antisense RNA contribute to the development and maintenance of chronic pain. An understanding of these mechanisms may propose novel therapeutic strategies for preventing and/or treating chronic pain.


Assuntos
Dor Crônica/genética , Dor Crônica/fisiopatologia , RNA não Traduzido/genética , RNA não Traduzido/fisiologia , Humanos , Inflamação/genética , Inflamação/fisiopatologia , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Canal de Potássio Kv1.2/biossíntese , Canal de Potássio Kv1.2/genética , Canal de Potássio Kv1.2/fisiologia , MicroRNAs/biossíntese , MicroRNAs/genética , Neuralgia/genética
12.
Sci Rep ; 4: 4201, 2014 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-24569544

RESUMO

Members of the six-transmembrane segment family of ion channels share a common structural design. However, there are sequence differences between the members that confer distinct biophysical properties on individual channels. Currently, we do not have 3D structures for all members of the family to help explain the molecular basis for the differences in their biophysical properties and pharmacology. This is due to low-level expression of many members in native or heterologous systems. One exception is rat Kv1.2 which has been overexpressed in Pichia pastoris and crystallised. Here, we tested chimaeras of rat Kv1.2 with the hERG channel for function in Xenopus oocytes and for overexpression in Pichia. Chimaera containing the S1-S6 transmembrane region of HERG showed functional and pharmacological properties similar to hERG and could be overexpressed and purified from Pichia. Our results demonstrate that rat Kv1.2 could serve as a surrogate to express difficult-to-overexpress members of the six-transmembrane segment channel family.


Assuntos
Canal de Potássio Kv1.2/química , Canal de Potássio Kv1.2/ultraestrutura , Pichia/metabolismo , Sequência de Aminoácidos , Animais , Clonagem Molecular/métodos , Canal de Potássio Kv1.2/fisiologia , Conformação Molecular , Dados de Sequência Molecular , Pichia/genética , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade
13.
Proc Natl Acad Sci U S A ; 111(6): 2128-33, 2014 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-24464485

RESUMO

Quantitative structure-based modeling of voltage activation of ion channels is very challenging. For example, it is very hard to reach converging results, by microscopic simulations while macroscopic treatments involve major uncertainties regarding key features. The current work overcomes some of the above challenges by using our recently developed coarse-grained (CG) model in simulating the activation of the Kv1.2 channel. The CG model has allowed us to explore problems that cannot be fully addressed at present by microscopic simulations, while providing insights on some features that are not usually considered in continuum models, including the distribution of the electrolytes between the membrane and the electrodes during the activation process and thus the physical nature of the gating current. Here, we demonstrate that the CG model yields realistic gating charges and free energy landscapes that allow us to simulate the fluctuating gating current in the activation processes. Our ability to simulate the time dependence of the fast gating current allows us to reproduce the observed trend and provides a clear description of its relationship to the landscape involved in the activation process.


Assuntos
Ativação do Canal Iônico , Canal de Potássio Kv1.2/fisiologia , Modelos Biológicos , Eletrodos , Canal de Potássio Kv1.2/química , Modelos Moleculares
14.
Neurosci Lett ; 548: 143-8, 2013 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-23769686

RESUMO

Gintonin is a novel ginseng-derived G protein-coupled lysophosphatidic acid (LPA) receptor ligand. The primary action of gintonin is to elicit a transient increase in [Ca(2+)]i via activation of LPA receptor subtypes. Voltage-gated potassium (Kv) channels play important roles in synaptic transmission in nervous systems. The previous reports have shown that Kv channels can be regulated by Gαq/11 protein-coupled receptor ligands. In the present study, we examined the effects of gintonin on Kv1.2 channel activity expressed in Xenopus oocytes after injection of RNA encoding the human Kv1.2 α subunit. Gintonin treatment inhibited Kv1.2 channel activity in reversible and concentration-dependent manners. The inhibitory effect of gintonin on Kv1.2 channel activity was blocked by active phospholipase C inhibitor, inositol 1,4,5-triphosphate receptor antagonist, and intracellular Ca(2+) chelator. The co-expression of active receptor protein tyrosine phosphatase α (RPTPα) with Kv1.2 channel greatly attenuated gintonin-mediated inhibition of Kv1.2 channel activity, but attenuation was not observed with catalytically inactive RPTPα. Furthermore, neither genistein, a tyrosine kinase inhibitor, nor site-directed mutation of a tyrosine residue (Y132 to Y132F), which is phosphorylated by tyrosine kinase of the N-terminal of the Kv1.2 channel α subunit, significantly attenuated gintonin-mediated inhibition of Kv1.2 channel activity. These results indicate that the gintonin-mediated Kv1.2 channel regulation involves the dual coordination of both tyrosine kinase and RPTPα coupled to this receptor. Finally, gintonin-mediated regulation of Kv1.2 channel activity might explain one of the modulations of gintonin-mediated neuronal activities in nervous systems.


Assuntos
Canal de Potássio Kv1.2/antagonistas & inibidores , Canal de Potássio Kv1.2/fisiologia , Panax/química , Proteínas Tirosina Quinases/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Ácidos Lisofosfatídicos/agonistas , Receptores de Ácidos Lisofosfatídicos/metabolismo , Animais , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Complexos Multienzimáticos/metabolismo , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Extratos Vegetais/farmacologia , Xenopus laevis
15.
Nat Neurosci ; 16(8): 1024-31, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23792947

RESUMO

Neuropathic pain is a refractory disease characterized by maladaptive changes in gene transcription and translation in the sensory pathway. Long noncoding RNAs (lncRNAs) are emerging as new players in gene regulation, but how lncRNAs operate in the development of neuropathic pain is unclear. Here we identify a conserved lncRNA, named Kcna2 antisense RNA, for a voltage-dependent potassium channel mRNA, Kcna2, in first-order sensory neurons of rat dorsal root ganglion (DRG). Peripheral nerve injury increased Kcna2 antisense RNA expression in injured DRG through activation of myeloid zinc finger protein 1, a transcription factor that binds to the Kcna2 antisense RNA gene promoter. Mimicking this increase downregulated Kcna2, reduced total voltage-gated potassium current, increased excitability in DRG neurons and produced neuropathic pain symptoms. Blocking this increase reversed nerve injury-induced downregulation of DRG Kcna2 and attenuated development and maintenance of neuropathic pain. These findings suggest endogenous Kcna2 antisense RNA as a therapeutic target for the treatment of neuropathic pain.


Assuntos
Regulação da Expressão Gênica/genética , Inativação Gênica , Canal de Potássio Kv1.2/antagonistas & inibidores , Proteínas do Tecido Nervoso/antagonistas & inibidores , Neuralgia/genética , Neurônios Aferentes/fisiologia , RNA Longo não Codificante/fisiologia , Animais , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiopatologia , Vetores Genéticos , Células HEK293 , Humanos , Canal de Potássio Kv1.2/genética , Canal de Potássio Kv1.2/fisiologia , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Neuralgia/fisiopatologia , Neuralgia/prevenção & controle , Traumatismos dos Nervos Periféricos , Regiões Promotoras Genéticas , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/biossíntese , RNA Longo não Codificante/genética , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Nervos Espinhais/lesões , Transativadores/biossíntese , Transativadores/fisiologia
17.
J Gen Physiol ; 140(5): 481-93, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23071266

RESUMO

Voltage-dependent potassium (Kv) channels provide the repolarizing power that shapes the action potential duration and helps control the firing frequency of neurons. The K(+) permeation through the channel pore is controlled by an intracellularly located bundle-crossing (BC) gate that communicates with the voltage-sensing domains (VSDs). During prolonged membrane depolarizations, most Kv channels display C-type inactivation that halts K(+) conduction through constriction of the K(+) selectivity filter. Besides triggering C-type inactivation, we show that in Shaker and Kv1.2 channels (expressed in Xenopus laevis oocytes), prolonged membrane depolarizations also slow down the kinetics of VSD deactivation and BC gate closure during the subsequent membrane repolarization. Measurements of deactivating gating currents (reporting VSD movement) and ionic currents (BC gate status) showed that the kinetics of both slowed down in two distinct phases with increasing duration of the depolarizing prepulse. The biphasic slowing in VSD deactivation and BC gate closure was strongly correlated in time and magnitude. Simultaneous recordings of ionic currents and fluorescence from a probe tracking VSD movement in Shaker directly demonstrated that both processes were synchronized. Whereas the first slowing originates from a stabilization imposed by BC gate opening, the subsequent slowing reflects the rearrangement of the VSD toward its relaxed state (relaxation). The VSD relaxation was observed in the Ciona intestinalis voltage-sensitive phosphatase and in its isolated VSD. Collectively, our results show that the VSD relaxation is not kinetically related to C-type inactivation and is an intrinsic property of the VSD. We propose VSD relaxation as a general mechanism for depolarization-induced slowing of BC gate closure that may enable Kv1.2 channels to modulate the firing frequency of neurons based on the depolarization history.


Assuntos
Ativação do Canal Iônico , Canal de Potássio Kv1.2/fisiologia , Potenciais da Membrana , Animais , Genes Reporter , Humanos , Cinética , Canal de Potássio Kv1.2/química , Canal de Potássio Kv1.2/genética , Simulação de Dinâmica Molecular , Mutação de Sentido Incorreto , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Potássio/metabolismo , Estrutura Terciária de Proteína , Xenopus
18.
J Gen Physiol ; 140(5): 495-511, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23071269

RESUMO

The open state of voltage-gated potassium (Kv) channels is associated with an increased stability relative to the pre-open closed states and is reflected by a slowing of OFF gating currents after channel opening. The basis for this stabilization is usually assigned to intrinsic structural features of the open pore. We have studied the gating currents of Kv1.2 channels and found that the stabilization of the open state is instead conferred largely by the presence of cations occupying the inner cavity of the channel. Large impermeant intracellular cations such as N-methyl-d-glucamine (NMG(+)) and tetraethylammonium cause severe slowing of channel closure and gating currents, whereas the smaller cation, Cs(+), displays a more moderate effect on voltage sensor return. A nonconducting mutant also displays significant open state stabilization in the presence of intracellular K(+), suggesting that K(+) ions in the intracellular cavity also slow pore closure. A mutation in the S6 segment used previously to enlarge the inner cavity (Kv1.2-I402C) relieves the slowing of OFF gating currents in the presence of the large NMG(+) ion, suggesting that the interaction site for stabilizing ions resides within the inner cavity and creates an energetic barrier to pore closure. The physiological significance of ionic occupation of the inner cavity is underscored by the threefold slowing of ionic current deactivation in the wild-type channel compared with Kv1.2-I402C. The data suggest that internal ions, including physiological concentrations of K(+), allosterically regulate the deactivation kinetics of the Kv1.2 channel by impairing pore closure and limiting the return of voltage sensors. This may represent a primary mechanism by which Kv channel deactivation kinetics is linked to ion permeation and reveals a novel role for channel inner cavity residues to indirectly regulate voltage sensor dynamics.


Assuntos
Ativação do Canal Iônico , Canal de Potássio Kv1.2/fisiologia , Regulação Alostérica , Sequência de Aminoácidos , Animais , Cátions/metabolismo , Linhagem Celular , Césio/farmacologia , Humanos , Espaço Intracelular/química , Ativação do Canal Iônico/efeitos dos fármacos , Canal de Potássio Kv1.2/química , Canal de Potássio Kv1.2/metabolismo , Meglumina/farmacologia , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Potássio/metabolismo , Estrutura Terciária de Proteína , Tetraetilamônio/farmacologia
19.
Proc Natl Acad Sci U S A ; 108(15): 6109-14, 2011 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-21444776

RESUMO

The response of a membrane-bound Kv1.2 ion channel to an applied transmembrane potential has been studied using molecular dynamics simulations. Channel deactivation is shown to involve three intermediate states of the voltage sensor domain (VSD), and concomitant movement of helix S4 charges 10-15 Å along the bilayer normal; the latter being enabled by zipper-like sequential pairing of S4 basic residues with neighboring VSD acidic residues and membrane-lipid head groups. During the observed sequential transitions S4 basic residues pass through the recently discovered charge transfer center with its conserved phenylalanine residue, F(233). Analysis indicates that the local electric field within the VSD is focused near the F(233) residue and that it remains essentially unaltered during the entire process. Overall, the present computations provide an atomistic description of VSD response to hyperpolarization, add support to the sliding helix model, and capture essential features inferred from a variety of recent experiments.


Assuntos
Canal de Potássio Kv1.2/química , Canal de Potássio Kv1.2/fisiologia , Membrana Celular/química , Membrana Celular/fisiologia , Bicamadas Lipídicas/química , Simulação de Dinâmica Molecular , Estrutura Secundária de Proteína , Eletricidade Estática
20.
Biochimie ; 92(12): 1847-53, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20713119

RESUMO

Scorpion venom-derived peptidyl toxins are valuable pharmacological tools for investigating the structure-function relationship of ion channels. Here, we report the purification, sequencing and functional characterization of a new K(+) channel blocker (MeuKTX) from the venom of the scorpion Mesobuthus eupeus. Effects of MeuKTX on ten cloned potassium channels in Xenopus oocytes were evaluated using two-electrode voltage-clamp recordings. MeuKTX is the orthologue of BmKTX (α-KTx3.6), a known Kv1.3 blocker from the scorpion Mesobuthus martensii, and classified as α-KTx3.13. MeuKTX potently blocks rKv1.1, rKv1.2 and hKv1.3 channels with 50% inhibitory concentration (IC(50)) of 203.15 ± 4.06 pM, 8.92 ± 2.3 nM and 171 ± 8.56 pM, respectively, but does not affect rKv1.4, rKv1.5, hKv3.1, rKv4.3, and hERG channels even at 2 µM concentration. At this high concentration, MeuKTX is also active on rKv1.6 and Shaker IR. Our results also demonstrate that MeuKTX and BmKTX have the same channel spectrum and similar pharmacological potency. Analysis of the structure-function relationships of α-KTx3 subfamily toxins allows us to recognize several key sites which may be useful for designing toxins with improved activity on hKv1.3, an attractive target for T-cell mediated autoimmune diseases.


Assuntos
Bloqueadores dos Canais de Potássio/farmacologia , Venenos de Escorpião/farmacologia , Escorpiões/química , Sequência de Aminoácidos , Animais , Relação Dose-Resposta a Droga , Feminino , Humanos , Canal de Potássio Kv1.2/genética , Canal de Potássio Kv1.2/fisiologia , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Microinjeções , Dados de Sequência Molecular , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Oócitos/fisiologia , Técnicas de Patch-Clamp , Filogenia , Bloqueadores dos Canais de Potássio/classificação , Bloqueadores dos Canais de Potássio/isolamento & purificação , RNA Complementar/administração & dosagem , RNA Complementar/genética , Ratos , Venenos de Escorpião/genética , Venenos de Escorpião/isolamento & purificação , Homologia de Sequência de Aminoácidos , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...