Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 21(7)2020 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-32244431

RESUMO

Cilobradine (CIL, DK-AH269), an inhibitor of hyperpolarization-activated cation current (Ih), has been observed to possess pro-arrhythmic properties. Whether and how CIL is capable of perturbing different types of membrane ionic currents existing in electrically excitable cells, however, is incompletely understood. In this study, we intended to examine possible modifications by it or other structurally similar compounds of ionic currents in pituitary tumor (GH3) cells and in heart-derived H9c2 cells. The standard whole-cell voltage-clamp technique was performed to examine the effect of CIL on ionic currents. GH3-cell exposure to CIL suppressed the density of hyperpolarization-evoked Ih in a concentration-dependent manner with an effective IC50 of 3.38 µM. Apart from its increase in the activation time constant of Ih during long-lasting hyperpolarization, the presence of CIL (3 µM) distinctly shifted the steady-state activation curve of Ih triggered by a 2-s conditioning pulse to a hyperpolarizing direction by 10 mV. As the impedance-frequency relation of Ih was studied, its presence raised the impedance magnitude at the resonance frequency induced by chirp voltage. CIL also suppressed delayed-rectifier K+ current (IK(DR)) followed by the accelerated inactivation time course of this current, with effective IC50 (measured at late IK(DR)) or KD value of 3.54 or 3.77 µM, respectively. As the CIL concentration increased 1 to 3 µM, the inactivation curve of IK(DR) elicited by 1- or 10-s conditioning pulses was shifted to a hyperpolarizing potential by approximately 10 mV, and the recovery of IK(DR) inactivation during its presence was prolonged. The peak Na+ current (INa) during brief depolarization was resistant to being sensitive to the presence of CIL, yet to be either decreased by subsequent addition of A-803467 or enhanced by that of tefluthrin. In cardiac H9c2 cells, unlike the CIL effect, the addition of either ivabradine or zatebradine mildly led to a lowering in IK(DR) amplitude with no conceivable change in the inactivation time course of the current. Taken together, the compound like CIL, which was tailored to block hyperpolarization-activated cation (HCN) channels effectively, was also capable of altering the amplitude and gating of IK(DR), thereby influencing the functional activities of electrically excitable cells, such as GH3 cells.


Assuntos
Benzazepinas/farmacologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/efeitos dos fármacos , Canais de Potássio Shab/efeitos dos fármacos , Animais , Cátions , Linhagem Celular Tumoral , Transporte de Íons/efeitos dos fármacos , Ivabradina , Cinética , Técnicas de Patch-Clamp , Piperidinas , Neoplasias Hipofisárias , Potássio/farmacologia , Sódio
2.
Int J Neurosci ; 130(8): 781-787, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31847645

RESUMO

Purpose/Aim: Besides as a cholinesterase (ChE) inhibitor, tacrine is able to act on multiple targets such as nicotinic receptors (nAChRs) and voltage-gated K+ (Kv) channels. Kv2.1, a Kv channel subunit underlying delayed rectifier currents with slow kinetics of inactivation, is highly expressed in the mammalian brain, especially in the hippocampus. Nevertheless, limited data are available concerning the relationship between tacrine and Kv2.1 channels. In the present study, we explore the possible effects of tacrine on Kv2.1 channels in heterologous expression systems and N2A cells.Materials and methods: The change of expression and currents of Kv2.1 after treatment with tacrine was detected by PCR and whole-cell recordings, respectively. WST-8 experiments were performed to reveal the effects of tacrine on cell proliferation.Results: Incubation with tacrine induced a significant reduction of the mRNA level of Kv2.1 channels in HEK293 cells. The decline of corresponding currents carried by Kv2.1 was also observed. Moreover, the proliferation rates of HEK293 cells with Kv2.1 channel were substantially enhanced after treatment with this chemical for 24 h. Similar results were also detected after exposure to tacrine in N2A cells with native expression of Kv2.1 channels.Conclusion: These lines of evidence indicate that application of tacrine downregulates the expression of Kv2.1 channels and increase cell proliferation. The effect of tacrine on Kv2.1 channels may provide an alternative explanation for its neuroprotective action.


Assuntos
Proliferação de Células/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Canais de Potássio Shab/efeitos dos fármacos , Tacrina/farmacologia , Animais , Linhagem Celular Tumoral , Regulação para Baixo , Células HEK293 , Humanos , Camundongos , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase , RNA Mensageiro , Sais de Tetrazólio
3.
ACS Chem Neurosci ; 9(12): 2886-2891, 2018 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-30001098

RESUMO

Photoswitchable blockers of potassium channels can be used to optically control neuronal excitability and hold great promise for vision restoration. Here, we report a series of improved photoswitchable blockers that are furnished with a new pharmacophore based on the local anesthetic bupivacaine. These azobupivacaines (ABs) enable optical control over the delayed rectifier channel Kv2.1. and target the two-pore domain potassium channel TREK-1. For the first time, we have identified a compound that blocks conductance in the dark and potentiates it upon illumination. Using light as a trigger, ABs efficiently and reversibly silence action potential firing of hippocampal neurons in acute mouse brain slices.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Compostos Azo/farmacologia , Bupivacaína/análogos & derivados , Luz , Neurônios/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Domínios Poros em Tandem/efeitos dos fármacos , Canais de Potássio Shab/efeitos dos fármacos , Animais , Compostos Azo/síntese química , Células HEK293 , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Camundongos , Neurônios/metabolismo , Fenômenos Ópticos , Bloqueadores dos Canais de Potássio/síntese química , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Canais de Potássio Shab/antagonistas & inibidores , Canais de Potássio Shab/metabolismo
4.
J Appl Toxicol ; 38(5): 696-704, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29297590

RESUMO

Methamphetamine (Meth) is an illicit psychostimulant with high abuse potential and severe neurotoxicity. Recent studies have shown that dysfunctions in learning and memory induced by Meth may partially reveal the mechanisms of neuronal channelopathies. Kv2.1, the primary delayed rectifying potassium channel in neurons, is responsible for mediating apoptotic current surge. However, whether Kv2.1 is involved in Meth-mediated neural injury remains unknown. In the present study, the treatment of primary cultured hippocampal neurons with Meth indicated that Meth induced a time- and dose-dependent augmentation of Kv2.1 protein expression, accompanied by elevated cleaved-caspase 3 and declined bcl-2/bax ratio. The blockage of Kv2.1 with the inhibitor GxTx-1E or the knockdown of the channel noticeably abrogated the pro-apoptotic effects mediated by Meth, demonstrating the specific roles of Kv2.1 in Meth-mediated neural damage. Additionally, the p38 mitogen-activated protein kinase (MAPK) signaling was demonstrated to be involved in Meth-mediated Kv2.1 upregulation and in the subsequent pro-apoptotic effects, as treatment with a p38 MAPK inhibitor significantly attenuated Meth-mediated Kv2.1 upregulation and cell apoptosis. Of note, PRE-084, a sigma-1 receptor agonist, obviously attenuated Meth-induced upregulation of Kv2.1 expression, neural apoptosis and p38 MAPK activation. Taken together, these results reveal a novel mechanism involved in Meth-induced neural death with implications for therapeutic interventions for Meth users.


Assuntos
Apoptose/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Metanfetamina/toxicidade , Neurônios/efeitos dos fármacos , Canais de Potássio Shab/efeitos dos fármacos , Animais , Western Blotting , Relação Dose-Resposta a Droga , Feminino , Técnicas de Silenciamento de Genes , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
5.
Toxicon ; 124: 8-14, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27810559

RESUMO

Jingzhaotoxin-XI (JZTX-XI) is a 34-residue peptide from the Chinese tarantula Chilobrachys jingzhao venom that potently inhibits both voltage-gated sodium channel Nav1.5 and voltage-gated potassium channel Kv2.1. In the present study, we further showed that JZTX-XI blocked Kv2.1 currents with the IC50 value of 0.39 ± 0.06 µM. JZTX-XI significantly shifted the current-voltage (I-V) curves and normalized conductance-voltage (G-V) curves of Kv2.1 channel to more depolarized voltages. Ala-scanning mutagenesis analyses demonstrated that mutants I273A, F274A, and E277A reduced toxin binding affinity by 10-, 16-, and 18-fold, respectively, suggesting that three common residues (I273, F274, E277) in the Kv2.1 S3b segment contribute to the formation of JZTX-XI receptor site, and the acidic residue Glu at the position 277 in Kv2.1 is the most important residue for JZTX-XI sensitivity. A single replacement of E277 with Asp(D) increased toxin inhibitory activity. These results establish that JZTX-XI inhibits Kv2.1 activation by trapping the voltage sensor in the rested state through a similar mechanism to that of HaTx1, but these two toxins have small differences in the most crucial molecular determinant. Furthermore, the in-depth investigation of the subtle differences in molecular determinants may be useful for increasing our understanding of the molecular details regarding toxin-channel interactions.


Assuntos
Peptídeos/toxicidade , Canais de Potássio Shab/efeitos dos fármacos , Venenos de Aranha/toxicidade , Sequência de Aminoácidos , Animais , Sítios de Ligação , Mutagênese Sítio-Dirigida , Homologia de Sequência de Aminoácidos , Canais de Potássio Shab/química , Canais de Potássio Shab/genética
6.
J Neurosci ; 35(50): 16404-17, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26674866

RESUMO

Little is known about the voltage-dependent potassium currents underlying spike repolarization in midbrain dopaminergic neurons. Studying mouse substantia nigra pars compacta dopaminergic neurons both in brain slice and after acute dissociation, we found that BK calcium-activated potassium channels and Kv2 channels both make major contributions to the depolarization-activated potassium current. Inhibiting Kv2 or BK channels had very different effects on spike shape and evoked firing. Inhibiting Kv2 channels increased spike width and decreased the afterhyperpolarization, as expected for loss of an action potential-activated potassium conductance. BK inhibition also increased spike width but paradoxically increased the afterhyperpolarization. Kv2 channel inhibition steeply increased the slope of the frequency-current (f-I) relationship, whereas BK channel inhibition had little effect on the f-I slope or decreased it, sometimes resulting in slowed firing. Action potential clamp experiments showed that both BK and Kv2 current flow during spike repolarization but with very different kinetics, with Kv2 current activating later and deactivating more slowly. Further experiments revealed that inhibiting either BK or Kv2 alone leads to recruitment of additional current through the other channel type during the action potential as a consequence of changes in spike shape. Enhancement of slowly deactivating Kv2 current can account for the increased afterhyperpolarization produced by BK inhibition and likely underlies the very different effects on the f-I relationship. The cross-regulation of BK and Kv2 activation illustrates that the functional role of a channel cannot be defined in isolation but depends critically on the context of the other conductances in the cell. SIGNIFICANCE STATEMENT: This work shows that BK calcium-activated potassium channels and Kv2 voltage-activated potassium channels both regulate action potentials in dopamine neurons of the substantia nigra pars compacta. Although both channel types participate in action potential repolarization about equally, they have contrasting and partially opposite effects in regulating neuronal firing at frequencies typical of bursting. Our analysis shows that this results from their different kinetic properties, with fast-activating BK channels serving to short-circuit activation of Kv2 channels, which tend to slow firing by producing a deep afterhyperpolarization. The cross-regulation of BK and Kv2 activation illustrates that the functional role of a channel cannot be defined in isolation but depends critically on the context of the other conductances in the cell.


Assuntos
Potenciais de Ação/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Neurônios/fisiologia , Canais de Potássio Shab/fisiologia , Substância Negra/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Neurônios Dopaminérgicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos/fisiologia , Potenciais Evocados/efeitos dos fármacos , Feminino , Cinética , Canais de Potássio Ativados por Cálcio de Condutância Alta/efeitos dos fármacos , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Recrutamento Neurofisiológico , Canais de Potássio Shab/efeitos dos fármacos , Substância Negra/citologia , Substância Negra/efeitos dos fármacos
7.
J Pept Sci ; 20(3): 159-64, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24464516

RESUMO

Scolopendra subspinipes mutilans, also known as Chinese red-headed centipede, is a venomous centipede from East Asia and Australasia. Venom from this animal has not been researched as thoroughly as venom from snakes, snails, scorpions, and spiders. In this study, we isolated and characterized SsmTx-I, a novel neurotoxin from the venom of S. subspinipes mutilans. SsmTx-I contains 36 residues with four cysteines forming two disulfide bonds. It had low sequence similarity (<10%) with other identified peptide toxins. By whole-cell recording, SsmTx-I significantly blocked voltage-gated K⁺ channels in dorsal root ganglion neurons with an IC50 value of 200 nM, but it had no effect on voltage-gated Na⁺ channels. Among the nine K⁺ channel subtypes expressed in human embryonic kidney 293 cells, SsmTx-I selectively blocked the Kv2.1 current with an IC50 value of 41.7 nM, but it had little effect on currents mediated by other K⁺ channel subtypes. Blockage of Kv2.1 by SsmTx-I was not associated with significant alteration of steady-state activation, suggesting that SsmTx-I might act as a simple inhibitor or channel blocker rather than a gating modifier. Our study reported a specific Kv2.1-blocker from centipede venom and provided a basis for future investigations of SsmTx-I, for example on structure-function relationships, mechanism of action, and pharmacological potential.


Assuntos
Medicamentos de Ervas Chinesas/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Canais de Potássio Shab/efeitos dos fármacos , Peçonhas/farmacologia , Animais , Linhagem Celular , Alcaloides Diterpenos , Humanos , Técnicas de Patch-Clamp , Ratos
8.
Yao Xue Xue Bao ; 48(1): 38-44, 2013 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-23600139

RESUMO

To investigate the electrophysiology mechanisms of new anxiolytic and antidepressant drug: 4-butyl-alpha-agarofuran (AF-5), patch clamp-recording was used to test the effects of AF-5 on voltage-dependent sodium currents, voltage-dependent potassium currents, L-type voltage-dependent calcium currents and GABA dependent Cl(-) currents in primary cultured rat cortical neurons. Effects of AF-5 on Kv2.1 currents, expressed stably in HEK293 cells, were also tested. Our results showed that, delayed rectifier potassium currents (I(K(DR, L-type voltage-dependent calcium currents (I(LC-ca)) in primary cultured rat cortical neurons and Kv2.1 currents in HEK293 cells were significantly inhibited by AF-5, with IC50 as 6.17, 4.4 and 5.29 micromol x L(-1) respectively. However, voltage-dependent sodium currents (I(Na)), GABA dependent Cl(-) currents and transient outward potassium currents (I(K(A)) in primary cultured rat cortical neurons were not significantly blocked by AF-5. Our results concluded that, blocked I(K(DR)) and I(L-Ca) currents may be one of the mechanisms of anxiolytic and antidepression actions of AF-5.


Assuntos
Antidepressivos/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Córtex Cerebral/citologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Sesquiterpenos/farmacologia , Animais , Células Cultivadas , Canais de Cloreto/efeitos dos fármacos , Canais de Potássio de Retificação Tardia/efeitos dos fármacos , Células HEK293 , Humanos , Neurônios/citologia , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Canais de Potássio Shab/efeitos dos fármacos , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos
9.
Toxicon ; 59(2): 265-71, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22155305

RESUMO

Jingzhaotoxin-XIII (JZTX-XIII), a 35 residue polypeptide, with the ability to inhibit voltage-dependent potassium channels in the shab (Kv2) and shal (Kv4) subfamilies, was purified from the venom of the Chinese tarantula Chilobrachys jingzhao. Electrophysiological recordings carried out in Xenopus laevis oocytes showed that JZTX-XIII acted as gating modifier of voltage-dependent K+ channels which inhibited the Kv2.1 channel and Kv4.1 channel, with the IC50 value of 0.47 µM and 1.17 µM, respectively. JZTX-XIII shares high sequence similarity with gating modifier toxins inhibiting a wide variety of ion channels including Nav1.5 subtype, but it showed no Nav1.5 channel activity. Structure-function analysis indicates that the acidic residues of Glu10 and Glu17 in JZTX-XIII might be responsible for the loss of the Nav1.5 channel inhibitory potency for JZTX-XIII.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Peptídeos/farmacologia , Venenos de Aranha/farmacologia , Aranhas/química , Sequência de Aminoácidos , Animais , Dados de Sequência Molecular , Neurotoxinas/química , Neurotoxinas/farmacologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Técnicas de Patch-Clamp , Peptídeos/química , Canais de Potássio Shab/efeitos dos fármacos , Canais de Sódio/efeitos dos fármacos , Venenos de Aranha/química , Xenopus laevis/metabolismo
10.
Exp Physiol ; 96(9): 836-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21551266

RESUMO

Our understanding of carbon monoxide (CO) as an endogenous signalling molecule has expanded enormously in recent years, to the point where it can now be safely exploited therapeutically. In more recent years, the ability of CO to modulate ion channel activity has added further diversity to its activity. Here, I summarize briefly the established ion channel targets of CO and describe in more detail its actions on two example ion channels (the cardiac L-type Ca(2+) channel and the neuronal delayed rectifier, Kv2.1). The physiological consequences of such modulation are also considered. It is clear that our knowledge of the cellular effects of this gas is incomplete, and further study is required to further understand and so more fully exploit its therapeutic potential.


Assuntos
Monóxido de Carbono/farmacologia , Canais Iônicos/efeitos dos fármacos , Canais de Cálcio Tipo L/efeitos dos fármacos , Humanos , Canais de Potássio Shab/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
11.
Channels (Austin) ; 5(1): 56-64, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21084865

RESUMO

Celecoxib is a drug designed to selectively inhibit COX-2, an inflammation-inducible cyclooxygenase isoform, over the constitutively expressed COX-1 isoform. In addition to this selective inhibition it is now known that celecoxib exerts a variety of effects on several types of ion channels, thus producing secondary physiological effects. In this work we demonstrate that at therapeutically relevant concentrations celecoxib interacts with Shab K(+) channels specifically promoting a fast inactivation gating (without blocking the pore or significantly affecting other gating processes). At least two celecoxib molecules bind to each channel promoting a fast inactivation that develops from both open and closed states. Channel inactivation in turn causes a reduction of the size of I(K). Taken together, our observations show that in addition to its intended therapeutic target celecoxib is a useful tool to further study the mechanism of Shab channel inactivation.


Assuntos
Inibidores de Ciclo-Oxigenase 2/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Potássio/metabolismo , Pirazóis/farmacologia , Canais de Potássio Shab/efeitos dos fármacos , Sulfonamidas/farmacologia , Animais , Baculoviridae/genética , Celecoxib , Linhagem Celular , Inibidores de Ciclo-Oxigenase 2/metabolismo , Cinética , Potenciais da Membrana , Ligação Proteica , Pirazóis/metabolismo , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Canais de Potássio Shab/genética , Canais de Potássio Shab/metabolismo , Sulfonamidas/metabolismo , Transfecção
12.
Brain Res ; 1359: 67-74, 2010 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-20801107

RESUMO

Recent research suggests that some clinical effects of coxibs, selective inhibitors of cyclooxygenase-2 (COX-2), can be mediated via modulation of ion channels. It has been shown that clinically relevant concentrations of celecoxib can cause inhibition or augmentation of various ionic currents and alter functioning of neurons and myocytes. Independence of these effects from inhibition of cyclooxygenases raises an important question if other structurally related COX-2 inhibitors can affect ion channels in similar fashion. Here we studied effects of SC-791, a highly selective COX-2 inhibitor, on K(v)2.1 channels expressed in HEK-293 cells. SC-791 reversibly inhibited K(v)2.1 in voltage-dependent manner with stronger inhibition at negative potentials. The values of IC50 were 1.8 µM and 7.2 µM for suppression of peak current at -20 and +40 mV, respectively. The current was reduced via acceleration of inactivation, hyperpolarizing shift in the half-inactivation potential and a large depolarizing shift in the half-activation potential. In addition, SC-791 accelerated all other aspects of K(v)2.1 kinetics: activation, deactivation and recovery from inactivation. Our results show that SC-791 modified K(v)2.1 gating, but, unlike celecoxib, did not induce channel block. These findings help to understand the mechanisms of unanticipated action of COX-2 inhibitors on voltage-activated potassium channels and their physiological implications.


Assuntos
Inibidores de Ciclo-Oxigenase 2/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Isoxazóis/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Canais de Potássio Shab/efeitos dos fármacos , Sulfonamidas/farmacologia , Células HEK293 , Humanos , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Canais de Potássio Shab/metabolismo
13.
J Physiol ; 588(Pt 15): 2691-706, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20547671

RESUMO

Quinidine is a commonly used antiarrhythmic agent and a tool to study ion channels. Here it is reported that quinidine equilibrates within seconds across the Sf9 plasma membrane, blocking the open pore of Shab channels from the intracellular side of the membrane in a voltage-dependent manner with 1:1 stoichiometry. On binding to the channels, quinidine interacts with pore K(+) ions in a mutually destabilizing manner. As a result, when the channels are blocked by quinidine with the cell bathed in an external medium lacking K(+), the Shab conductance G(K) collapses irreversibly, despite the presence of a physiological [K(+)] in the intracellular solution. The quinidine-promoted collapse of Shab G(K) resembles the collapse of Shaker G(K) observed with 0 K(+) solutions on both sides of the membrane: thus the extent of G(K) drop depends on the number of activating pulses applied in the presence of quinidine, but is independent of the pulse duration. Taken together the observations indicate that, as in Shaker, the quinidine-promoted collapse of Shab G(K) occurs during deactivation of the channels, at the end of each activating pulse, with a probability of 0.1 per pulse at 80 mV. It appears that when Shab channels are open, the pore conformation able to conduct is stable in the absence of K(+), but on deactivation this conformation collapses irreversibly.


Assuntos
Permeabilidade da Membrana Celular/fisiologia , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/fisiologia , Potássio/metabolismo , Quinidina/administração & dosagem , Quinidina/farmacocinética , Canais de Potássio Shab/fisiologia , Animais , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Condutividade Elétrica , Potenciais da Membrana/efeitos dos fármacos , Canais de Potássio Shab/efeitos dos fármacos , Spodoptera
14.
Life Sci ; 86(21-22): 775-80, 2010 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-20385147

RESUMO

A manifestation in neurodegeneration is apoptosis of neurons. Neurons undergoing apoptosis may lose a substantial amount of cytosolic K+ through a number of pathways including K+ efflux via voltage-gated K+ (Kv) channels. The consequent drop in cytosolic [K+] relieves inhibition of an array of pro-apoptotic enzymes such as caspases and nucleases. Blocking Kv channels has been known to prevent neuronal apoptosis by preventing K+ efflux. Some neural diseases such as epilepsy are caused by neuronal hyperexcitability, which eventually may lead to neuronal apoptosis. Reduction in activities of A-type Kv channels and Kv7 subfamily members is amongst the etiological causes of neuronal hyperexcitation; enhancing the opening of these channels may offer opportunities of remedy. This review discusses the potential uses of Kv channel modulators as neuroprotective drugs.


Assuntos
Fármacos Neuroprotetores/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Humanos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Bloqueadores dos Canais de Potássio/uso terapêutico , Canais de Potássio/fisiologia , Canais de Potássio Shab/efeitos dos fármacos , Canais de Potássio Shab/fisiologia
15.
J Pharmacol Exp Ther ; 332(2): 437-45, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19915071

RESUMO

Ion channels are known to be modulated by antidepressant drugs, but the molecular mechanisms are not known. We have shown that the antidepressant drug amoxapine suppresses rectifier outward K(+) (I(K)) currents in mouse cortical neurons. At a concentration of 10 to 500 muM, amoxapine reversibly inhibited I(K) in a dose-dependent manner and modulated both steady-state activation and inactivation properties. The application of forskolin or dibutyryl cAMP mimicked the inhibitory effect of amoxapine on I(K) and abolished further inhibition by amoxapine. N-[2-(p-Bromocinnamylamino)ethyl]-5-iso-quinolinesulphonamide (H-89), a protein kinase A (PKA) inhibitor, augmented I(K) amplitudes and completely eliminated amoxapine inhibition of I(K). Amoxapine was also found to significantly increase intracellular cAMP levels. The effects of amoxapine on I(K) were abolished by preincubation with 5-hydroxytryptamine (5-HT) and the antagonists of 5-HT(2) receptor. Moreover, intracellular application of guanosine 5'-[gammathio]-triphosphate increased I(K) amplitudes and prevented amoxapine-induced inhibition. The selective Kv2.1 subunit blocker Jingzhaotoxin-III reduced I(K) amplitudes by 30% and also significantly abolished the inhibitory effect of amoxapine. Together these results suggest that amoxapine inhibits I(K) in mouse cortical neurons by cAMP/PKA-dependent pathway associated with the 5-HT receptor, and suggest that the Kv2.1 alpha-subunit may be the target for this inhibition.


Assuntos
Amoxapina/farmacologia , Antidepressivos de Segunda Geração/farmacologia , Córtex Cerebral/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/efeitos dos fármacos , Canais de Potássio de Retificação Tardia/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Técnicas de Cultura de Células , AMP Cíclico/metabolismo , Canais de Potássio de Retificação Tardia/fisiologia , Interações Medicamentosas , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Neurônios/metabolismo , Neurônios/fisiologia , Antagonistas do Receptor 5-HT2 de Serotonina , Canais de Potássio Shab/efeitos dos fármacos
16.
Proc Natl Acad Sci U S A ; 106(37): 15903-8, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19717445

RESUMO

An estimated 3% of the global population are infected with hepatitis C virus (HCV), and the majority of these individuals will develop chronic liver disease. As with other chronic viruses, establishment of persistent infection requires that HCV-infected cells must be refractory to a range of pro-apoptotic stimuli. In response to oxidative stress, amplification of an outward K(+) current mediated by the Kv2.1 channel, precedes the onset of apoptosis. We show here that in human hepatoma cells either infected with HCV or harboring an HCV subgenomic replicon, oxidative stress failed to initiate apoptosis via Kv2.1. The HCV NS5A protein mediated this effect by inhibiting oxidative stress-induced p38 MAPK phosphorylation of Kv2.1. The inhibition of a host cell K(+) channel by a viral protein is a hitherto undescribed viral anti-apoptotic mechanism and represents a potential target for antiviral therapy.


Assuntos
Apoptose/fisiologia , Hepacivirus/fisiologia , Hepacivirus/patogenicidade , Canais de Potássio Shab/antagonistas & inibidores , Proteínas não Estruturais Virais/fisiologia , 2,2'-Dipiridil/análogos & derivados , 2,2'-Dipiridil/farmacologia , Linhagem Celular , Dissulfetos/farmacologia , Hepacivirus/genética , Hepatite C Crônica/metabolismo , Hepatite C Crônica/virologia , Humanos , Estresse Oxidativo , Canais de Potássio Shab/efeitos dos fármacos , Canais de Potássio Shab/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Alcohol ; 43(1): 45-50, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19185209

RESUMO

Delayed-rectifier Kv2.1 channels are the principal component of voltage-sensitive K+ currents (I(K)) in hippocampal neurons and are critical regulators of somatodendritic excitability. In a recent study, we demonstrated that surface trafficking and phosphorylation of Kv2.1 channels is modulated by NMDA-type glutamate receptors and that astroglial excitatory amino acid transporters 2 (EAAT2) regulate the coupling of NMDA receptors and Kv2.1 channels. Because ethanol is known to acutely inhibit NMDA receptors, we sought to determine if NMDA receptor and astroglial EAAT2 modulation of Kv2.1 channels is impaired by ethanol in the rodent hippocampus. As expected, bath application of NMDA to hippocampal cultures reduced the size of Kv2.1 clusters and produced a hyperpolarizing shift in the voltage-dependent activation of I(K) that was associated with dephosphorylated Kv2.1 channels. Ethanol, applied acutely, prevented the hyperpolarizing shift in activation of I(K) induced by NMDA and restored Kv2.1 clustering and phosphorylation to near control levels. Ethanol also attenuated the dephosphorylation of Kv2.1 channels produced by the EAAT2 selective inhibitor dihydrokainic acid. These data demonstrate that acute ethanol disrupts changes in Kv2.1 channels that follow NMDA receptor activation and impairs astroglial regulation of the functional coupling between NMDA receptors and Kv2.1 channels.


Assuntos
Astrócitos/metabolismo , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Transportador 2 de Aminoácido Excitatório/fisiologia , Hipocampo/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Canais de Potássio Shab/efeitos dos fármacos , Animais , Astrócitos/efeitos dos fármacos , Células Cultivadas , Eletrofisiologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Transportador 2 de Aminoácido Excitatório/antagonistas & inibidores , Hipocampo/efeitos dos fármacos , Ácido Caínico/análogos & derivados , Ácido Caínico/farmacologia , Técnicas de Patch-Clamp , Fosforilação , Ratos , Ratos Sprague-Dawley
18.
J Neurochem ; 108(4): 909-19, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19077057

RESUMO

Chronic loss of intracellular K(+) can induce neuronal apoptosis in pathological conditions. However, the mechanism by which the K(+) channels are regulated in this process remains largely unknown. Here, we report that the increased membrane expression of Kv2.1 proteins in cortical neurons deprived of serum, a condition known to induce K(+) loss, promotes neuronal apoptosis. The increase in I(K) current density and apoptosis in the neurons deprived of serum were inhibited by a dominant negative form of Kv2.1 and MK801, an antagonist to NMDA receptors. The membrane level of Kv2.1 and its interaction with SNAP25 were increased, whereas the Kv2.1 phosphorylation was inhibited in the neurons deprived of serum. Botulinum neurotoxin, an agent known to prevent formation of soluble N-ethylmaleimide-sensitive factor attachment protein receptor complex, suppressed the increase in I(K) current density. Together, these results suggest that NMDA receptor-dependent Kv2.1 membrane translocation is regulated by a soluble N-ethylmaleimide-sensitive factor attachment protein receptor-dependent vesicular trafficking mechanism and is responsible for neuronal cell death induced by chronic loss of K(+).


Assuntos
Apoptose/fisiologia , Córtex Cerebral/metabolismo , Neurônios/metabolismo , Neurotoxinas/metabolismo , Deficiência de Potássio/metabolismo , Canais de Potássio Shab/metabolismo , Animais , Apoptose/efeitos dos fármacos , Toxinas Botulínicas/farmacologia , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Meios de Cultura Livres de Soro/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteínas Sensíveis a N-Etilmaleimida/metabolismo , Fármacos Neuromusculares/farmacologia , Neurônios/efeitos dos fármacos , Neurotoxinas/farmacologia , Técnicas de Patch-Clamp , Potássio/metabolismo , Deficiência de Potássio/fisiopatologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ratos , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Canais de Potássio Shab/efeitos dos fármacos , Canais de Potássio Shab/genética , Proteína 25 Associada a Sinaptossoma/metabolismo , Vesículas Transportadoras/metabolismo
19.
Channels (Austin) ; 3(6): 437-47, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21150283

RESUMO

Gating modifier peptides alter gating of voltage-gated potassium (KV) channels by binding to the voltage sensor paddle and changing the energetics of channel opening. Since the voltage sensor paddle is a modular motif with low sequence similarity across families, targeting of this region should yield highly specific channel modifiers. To test this idea, we developed a binding assay with the KV2.1 gating modifier, GxTX-1E. Monoiodotyrosine-GxTX-1E (125I-GxTX-1E) binds with high affinity (IC50 = 4 nM) to CHO cells stably expressing hKV2.1 channels, but not to CHO cells expressing Maxi-K channels. Binding of 125I-GxTX-1E to KV2.1 channels is inhibited by another KV2.1 gating modifier, stromatoxin (IC50 = 30 nM), but is not affected by iberiotoxin or charybdotoxin, pore blocking peptides of other types of potassium channels, or by ProTx-II, a selective gating modifier peptide of the voltage-gated sodium channel NaV1.7. Specific 125I-GxTX-1E binding is not detectable when CHO-KV2.1 cells are placed in high external potassium, suggesting that depolarization favors dissociation of the peptide. The binding assay was adapted to a 384-well format, allowing high throughput screening of large compound libraries. Interestingly, we discovered that compounds related to PAC, a di-substituted cyclohexyl KV channel blocker, displayed inhibitory binding activity. These data establish the feasibility of screening large libraries of compounds in an assay that monitors the displacement of a gating modifier from the channel's voltage sensor. Future screens using this approach will ultimately test whether the voltage sensor of KV channels can be selectively targeted by small molecules to modify channel function.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Ativação do Canal Iônico/efeitos dos fármacos , Canais de Potássio Shab/metabolismo , Animais , Proteínas de Artrópodes , Células CHO , Cricetinae , Cricetulus , Humanos , Concentração Inibidora 50 , Peptídeos/farmacologia , Ligação Proteica , Canais de Potássio Shab/efeitos dos fármacos , Venenos de Aranha/farmacologia
20.
BMC Physiol ; 8: 2, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18261223

RESUMO

BACKGROUND: K+ channels are diverse; both in terms of their function and their molecular composition. Shal subunits were first described in Drosophila. There are three mammalian orthologs, which are members of the Kv4 subfamily. They are involved in neuronal firing patterns as well as control of the cardiac action potential duration. RESULTS: Here, we report the biophysical and pharmacological characterization of zShal3, which is the ortholog of the mammalian Kv4.3 subunit, which in mammals is involved in action potential repolarization and gives rise to neuronal A-type K+ currents involved in somatodendretic signal integration. CONCLUSION: We demonstrate that zShal has similar functional and pharmacological characteristics compared to Kv4.3 and it is similarly regulated by pharmacological agents and by the Kv4 accessory subunit, NCS-1.


Assuntos
Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/fisiologia , Canais de Potássio Shab/química , Canais de Potássio Shab/fisiologia , Peixe-Zebra/fisiologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Ativação do Canal Iônico/efeitos dos fármacos , Dados de Sequência Molecular , Subunidades Proteicas , Canais de Potássio Shab/efeitos dos fármacos , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...