Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 293
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Molecules ; 27(13)2022 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-35807390

RESUMO

Voltage-gated Na+ (NaV) channels are significant therapeutic targets for the treatment of cardiac and neurological disorders, thus promoting the search for novel NaV channel ligands. With the objective of discovering new blockers of NaV channel ligands, we screened an In-House vegetal alkaloid library using fluorescence cell-based assays. We screened 62 isoquinoline alkaloids (IA) for their ability to decrease the FRET signal of voltage sensor probes (VSP), which were induced by the activation of NaV channels with batrachotoxin (BTX) in GH3b6 cells. This led to the selection of five IA: liriodenine, oxostephanine, thalmiculine, protopine, and bebeerine, inhibiting the BTX-induced VSP signal with micromolar IC50. These five alkaloids were then assayed using the Na+ fluorescent probe ANG-2 and the patch-clamp technique. Only oxostephanine and liriodenine were able to inhibit the BTX-induced ANG-2 signal in HEK293-hNaV1.3 cells. Indeed, liriodenine and oxostephanine decreased the effects of BTX on Na+ currents elicited by the hNaV1.3 channel, suggesting that conformation change induced by BTX binding could induce a bias in fluorescent assays. However, among the five IA selected in the VSP assay, only bebeerine exhibited strong inhibitory effects against Na+ currents elicited by the hNav1.2 and hNav1.6 channels, with IC50 values below 10 µM. So far, bebeerine is the first BBIQ to have been reported to block NaV channels, with promising therapeutical applications.


Assuntos
Alcaloides , Corantes Fluorescentes , Alcaloides/farmacologia , Batraquiotoxinas/metabolismo , Batraquiotoxinas/farmacologia , Viés , Células HEK293 , Humanos , Isoquinolinas/farmacologia , Ligantes , Sódio/metabolismo
2.
Cell Chem Biol ; 29(4): 615-624.e5, 2022 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-34963066

RESUMO

Voltage-gated sodium channels (NaVs) are targets for a number of acute poisons. Many of these agents act as allosteric modulators of channel activity and serve as powerful chemical tools for understanding channel function. Herein, we detail studies with batrachotoxin (BTX), a potent steroidal amine, and three ester derivatives prepared through de novo synthesis against recombinant NaV subtypes (rNaV1.4 and hNaV1.5). Two of these compounds, BTX-B and BTX-cHx, are functionally equivalent to BTX, hyperpolarizing channel activation and blocking both fast and slow inactivation. BTX-yne-a C20-n-heptynoate ester-is a conspicuous outlier, eliminating fast but not slow inactivation. This property differentiates BTX-yne among other NaV modulators as a unique reagent that separates inactivation processes. These findings are supported by functional studies with bacterial NaVs (BacNaVs) that lack a fast inactivation gate. The availability of BTX-yne should advance future efforts aimed at understanding NaV gating mechanisms and designing allosteric regulators of NaV activity.


Assuntos
Batraquiotoxinas , Canais de Sódio Disparados por Voltagem , Batraquiotoxinas/farmacologia , Ésteres , Sódio/metabolismo
3.
J Gen Physiol ; 151(2): 186-199, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30587506

RESUMO

Batrachotoxin (BTX), an alkaloid from skin secretions of dendrobatid frogs, causes paralysis and death by facilitating activation and inhibiting deactivation of eukaryotic voltage-gated sodium (Nav) channels, which underlie action potentials in nerve, muscle, and heart. A full understanding of the mechanism by which BTX modifies eukaryotic Nav gating awaits determination of high-resolution structures of functional toxin-channel complexes. Here, we investigate the action of BTX on the homotetrameric prokaryotic Nav channels NaChBac and NavSp1. By combining mutational analysis and whole-cell patch clamp with molecular and kinetic modeling, we show that BTX hinders deactivation and facilitates activation in a use-dependent fashion. Our molecular model shows the horseshoe-shaped BTX molecule bound within the open pore, forming hydrophobic H-bonds and cation-π contacts with the pore-lining helices, leaving space for partially dehydrated sodium ions to permeate through the hydrophilic inner surface of the horseshoe. We infer that bulky BTX, bound at the level of the gating-hinge residues, prevents the S6 rearrangements that are necessary for closure of the activation gate. Our results reveal general similarities to, and differences from, BTX actions on eukaryotic Nav channels, whose major subunit is a single polypeptide formed by four concatenated, homologous, nonidentical domains that form a pseudosymmetric pore. Our determination of the mechanism by which BTX activates homotetrameric voltage-gated channels reveals further similarities between eukaryotic and prokaryotic Nav channels and emphasizes the tractability of bacterial Nav channels as models of voltage-dependent ion channel gating. The results contribute toward a deeper, atomic-level understanding of use-dependent natural and synthetic Nav channel agonists and antagonists, despite their overlapping binding motifs on the channel proteins.


Assuntos
Proteínas de Bactérias/metabolismo , Batraquiotoxinas/farmacologia , Agonistas de Canais de Sódio/farmacologia , Canais de Sódio/metabolismo , Bacillus , Proteínas de Bactérias/agonistas , Proteínas de Bactérias/química , Linhagem Celular , Humanos , Ativação do Canal Iônico , Rhodobacteraceae , Canais de Sódio/química
4.
Science ; 354(6314): 865-869, 2016 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-27856903

RESUMO

The steroidal neurotoxin (-)-batrachotoxin functions as a potent agonist of voltage-gated sodium ion channels (NaVs). Here we report concise asymmetric syntheses of the natural (-) and non-natural (+) antipodes of batrachotoxin, as well both enantiomers of a C-20 benzoate-modified derivative. Electrophysiological characterization of these molecules against NaV subtypes establishes the non-natural toxin enantiomer as a reversible antagonist of channel function, markedly different in activity from (-)-batrachotoxin. Protein mutagenesis experiments implicate a shared binding side for the enantiomers in the inner pore cavity of NaV These findings motivate and enable subsequent studies aimed at revealing how small molecules that target the channel inner pore modulate NaV dynamics.


Assuntos
Batraquiotoxinas/síntese química , Batraquiotoxinas/farmacologia , Proteínas Musculares/antagonistas & inibidores , Bloqueadores do Canal de Sódio Disparado por Voltagem/síntese química , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Animais , Sítios de Ligação , Proteínas Musculares/química , Proteínas Musculares/genética , Mutação Puntual , Estrutura Secundária de Proteína , Ratos , Canais de Sódio/química , Canais de Sódio/genética , Bloqueadores do Canal de Sódio Disparado por Voltagem/química
5.
Curr Top Membr ; 78: 117-44, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27586283

RESUMO

Voltage-gated sodium channels are targets for many toxins and medically important drugs. Despite decades of intensive studies in industry and academia, atomic mechanisms of action are still not completely understood. The major cause is a lack of high-resolution structures of eukaryotic channels and their complexes with ligands. In these circumstances a useful approach is homology modeling that employs as templates X-ray structures of potassium channels and prokaryotic sodium channels. On one hand, due to inherent limitations of this approach, results should be treated with caution. In particular, models should be tested against relevant experimental data. On the other hand, docking of drugs and toxins in homology models provides a unique possibility to integrate diverse experimental data provided by mutational analysis, electrophysiology, and studies of structure-activity relations. Here we describe how homology modeling advanced our understanding of mechanisms of several classes of ligands. These include tetrodotoxins and mu-conotoxins that block the outer pore, local anesthetics that block of the inner pore, batrachotoxin that binds in the inner pore but, paradoxically, activates the channel, pyrethroid insecticides that activate the channel by binding at lipid-exposed repeat interfaces, and scorpion alpha and beta-toxins, which bind between the pore and voltage-sensing domains and modify the channel gating. We emphasize importance of experimental data for elaborating the models.


Assuntos
Canais de Sódio Disparados por Voltagem/metabolismo , Animais , Batraquiotoxinas/química , Batraquiotoxinas/metabolismo , Batraquiotoxinas/farmacologia , Sítios de Ligação , Conotoxinas/química , Conotoxinas/metabolismo , Conotoxinas/toxicidade , Inseticidas/química , Inseticidas/metabolismo , Inseticidas/toxicidade , Ativação do Canal Iônico/efeitos dos fármacos , Ligantes , Simulação de Dinâmica Molecular , Método de Monte Carlo , Estrutura Terciária de Proteína , Piretrinas/química , Piretrinas/metabolismo , Piretrinas/toxicidade , Esteroides/química , Esteroides/metabolismo , Tetrodotoxina/química , Tetrodotoxina/metabolismo , Tetrodotoxina/toxicidade , Agonistas do Canal de Sódio Disparado por Voltagem/química , Agonistas do Canal de Sódio Disparado por Voltagem/metabolismo , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/metabolismo , Canais de Sódio Disparados por Voltagem/química
6.
ACS Chem Neurosci ; 7(10): 1463-1468, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27501251

RESUMO

A novel family of small molecule inhibitors of voltage-gated sodium channels (NaVs) based on the structure of batrachotoxin (BTX), a well-known channel agonist, is described. Protein mutagenesis and electrophysiology experiments reveal the binding site as the inner pore region of the channel, analogous to BTX, alkaloid toxins, and local anesthetics. Homology modeling of the eukaryotic channel based on recent crystallographic analyses of bacterial NaVs suggests a mechanism of action for ion conduction block.


Assuntos
Batraquiotoxinas/análise , Batraquiotoxinas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Animais , Batraquiotoxinas/síntese química , Células CHO , Cricetulus , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Modelos Moleculares , Estrutura Molecular , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutação , Técnicas de Patch-Clamp , Ratos , Bloqueadores dos Canais de Sódio/síntese química , Canais de Sódio/genética , Canais de Sódio/metabolismo , Relação Estrutura-Atividade
7.
Channels (Austin) ; 7(4): 263-74, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23695971

RESUMO

Miniature persistent late Na(+) currents in cardiomyocytes have been linked to arrhythmias and sudden death. The goals of this study are to establish a stable cell line expressing robust persistent cardiac Na(+) currents and to test Class 1 antiarrhythmic drugs for selective action against resting and open states. After transient transfection of an inactivation-deficient human cardiac Na(+) channel clone (hNav1.5-CW with L409C/A410W double mutations), transfected mammalian HEK293 cells were treated with 1 mg/ml G-418. Individual G-418-resistant colonies were isolated using glass cylinders. One colony with high expression of persistent Na(+) currents was subjected to a second colony selection. Cells from this colony remained stable in expressing robust peak Na(+) currents of 996 ± 173 pA/pF at +50 mV (n = 20). Persistent late Na(+) currents in these cells were clearly visible during a 4-second depolarizing pulse albeit decayed slowly. This slow decay is likely due to slow inactivation of Na(+) channels and could be largely eliminated by 5 µM batrachotoxin. Peak cardiac hNav1.5-CW Na(+) currents were blocked by tetrodotoxin with an IC(50) value of 2.27 ± 0.08 µM (n = 6). At clinic relevant concentrations, Class 1 antiarrhythmics are much more selective in blocking persistent late Na(+) currents than their peak counterparts, with a selectivity ratio ranging from 80.6 (flecainide) to 3 (disopyramide). We conclude that (1) Class 1 antiarrhythmics differ widely in their resting- vs. open-channel selectivity, and (2) stably transfected HEK293 cells expressing large persistent hNav1.5-CW Na(+) currents are suitable for studying as well as screening potent open-channel blockers.


Assuntos
Antiarrítmicos/farmacologia , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Transfecção , Batraquiotoxinas/farmacologia , Células HEK293 , Humanos , Mutação , Miócitos Cardíacos/efeitos dos fármacos , Fatores de Tempo
8.
Insect Biochem Mol Biol ; 41(7): 446-50, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21426938

RESUMO

Deltamethrin, a pyrethroid insecticide, and BTG 502, an alkylamide insecticide, target voltage-gated sodium channels. Deltamethrin binds to a unique receptor site and causes prolonged opening of sodium channels by inhibiting deactivation and inactivation. Previous (22)Na(+) influx and receptor binding assays using mouse brain synaptoneurosomes showed that BTG 502 antagonized the binding and action of batrachotoxin (BTX), a site 2 sodium channel neurotoxin. However, the effect of BTG 502 has not been examined directly on sodium channels expressed in Xenopus oocytes. In this study, we examined the effect of BTG 502 on wild-type and mutant cockroach sodium channels expressed in Xenopus oocytes. Toxin competition experiments confirmed that BTG 502 antagonizes the action of BTX and possibly shares a common receptor site with BTX. However, unlike BTX which causes persistent activation of sodium channels, BTG 502 reduces the amplitude of peak sodium current. A previous study showed that BTG 502 was more toxic to pyrethroid-resistant house flies possessing a super-kdr (knockdown resistance) mechanism than to pyrethroid-susceptible house flies. However, we found that the cockroach sodium channels carrying the equivalent super-kdr mutations (M918T and L1014F) were not more sensitive to BTG 502 than the wild-type channel. Instead, a kdr mutation, F1519I, which reduces pyrethroid binding, abolished the action of BTG 502. These results provide evidence the actions of alkylamide and pyrethroid insecticides require a common sodium channel residue.


Assuntos
Baratas , Inseticidas/farmacologia , Naftalenos/farmacologia , Nitrilas/farmacologia , Oócitos/metabolismo , Fenilalanina/metabolismo , Alcamidas Poli-Insaturadas/farmacologia , Piretrinas/farmacologia , Proteínas Recombinantes/metabolismo , Canais de Sódio , Animais , Batraquiotoxinas/farmacologia , Sítios de Ligação , Ligação Competitiva , Baratas/genética , Baratas/metabolismo , Feminino , Controle de Insetos/métodos , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Potenciais da Membrana , Mutação , Oócitos/citologia , Técnicas de Patch-Clamp , Fenilalanina/genética , Ligação Proteica , Ratos , Proteínas Recombinantes/genética , Sódio/metabolismo , Agonistas de Canais de Sódio , Canais de Sódio/genética , Canais de Sódio/metabolismo , Transfecção , Xenopus laevis
9.
Chem Biol ; 16(8): 893-906, 2009 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-19716479

RESUMO

Hoiamide A, a novel bioactive cyclic depsipeptide, was isolated from an environmental assemblage of the marine cyanobacteria Lyngbya majuscula and Phormidium gracile collected in Papua New Guinea. This stereochemically complex metabolite possesses a highly unusual structure, which likely derives from a mixed peptide-polyketide biogenetic origin, and includes a peptidic section featuring an acetate extended and S-adenosyl methionine modified isoleucine moiety, a triheterocyclic fragment bearing two alpha-methylated thiazolines and one thiazole, and a highly oxygenated and methylated C15-polyketide substructure. Pure hoiamide A potently inhibited [(3)H]batrachotoxin binding to voltage-gated sodium channels (IC(50) = 92.8 nM), activated sodium influx (EC(50) = 2.31 microM) in mouse neocortical neurons, and exhibited modest cytotoxicity to cancer cells. Further investigation revealed that hoiamide A is a partial agonist of site 2 on the voltage-gated sodium channel.


Assuntos
Cianobactérias/química , Depsipeptídeos/química , Agonistas de Canais de Sódio , Animais , Batraquiotoxinas/farmacologia , Sítios de Ligação , Depsipeptídeos/isolamento & purificação , Depsipeptídeos/toxicidade , Camundongos , Neurônios/metabolismo , Papua Nova Guiné , Receptores de N-Metil-D-Aspartato/metabolismo , Sódio/metabolismo , Canais de Sódio/metabolismo , Estereoisomerismo
10.
Brain Res ; 1203: 61-7, 2008 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-18321475

RESUMO

Recently, we showed that the 20(S)-ginsenoside Rg3 (Rg3), an active ingredient of Panax ginseng, inhibits rat brain NaV1.2 channel peak currents (INa). Batrachotoxin (BTX) is a steroidal alkaloid neurotoxin and activates NaV channels through interacting with transmembrane domain-I-segment 6 (IS6) of channels. Recent report shows that ginsenoside inhibits BTX binding in rat brain membrane fractions. However, it needs to be confirmed whether biochemical mechanism is relevant physiologically and which residues of the BTX binding sites are important for ginsenoside regulations. Here, we demonstrate that mutations of BTX binding sites such as N418K and L421K of rat brain NaV1.2 and L437K of mouse skeletal muscle NaV1.4 channel reduce or abolish Rg3 inhibition of I(Na) and attenuate Rg3-mediated depolarizing shift of the activation voltage and use-dependent inhibition. These results indicate that BTX binding sites play an important role in modifying Rg3-mediated Na+ channel properties.


Assuntos
Batraquiotoxinas/farmacologia , Ginsenosídeos/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Proteínas Musculares/fisiologia , Neurotoxinas/farmacologia , Canais de Sódio/fisiologia , Animais , Batraquiotoxinas/química , Sítios de Ligação/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ginsenosídeos/química , Ativação do Canal Iônico/fisiologia , Leucina/genética , Lisina/genética , Camundongos , Microinjeções , Proteínas Musculares/genética , Músculo Esquelético , Canal de Sódio Disparado por Voltagem NAV1.2 , Proteínas do Tecido Nervoso , Oócitos , Técnicas de Patch-Clamp , Mutação Puntual/fisiologia , Estrutura Terciária de Proteína/fisiologia , Ratos , Canais de Sódio/genética , Xenopus laevis
11.
Ross Fiziol Zh Im I M Sechenova ; 93(5): 531-43, 2007 May.
Artigo em Russo | MEDLINE | ID: mdl-17650621

RESUMO

The voltage-gated sodium channels play a key role in the generation of action potential in excitable cells. Sodium channels are targeted by a number of modulating ligands. Despite numerous studies, the mechanisms of action of many ligands are still unknown. The main cause of the problem is the absence of the channel structure. Sodium channels belong to the superfamily of P-loop channels that also the data abowt includes potassium and calcium channels and the channels of ionotropic glutamate receptors. Crystallization of several potassium channels has opened a possibility to analyze the structure of other members of the superfamily using the homology modeling approach. The present study summarizes the results of several recent modelling studies of such sodium channel ligands as tetrodotoxin, batrachotoxin and local anesthetics. Comparison of available experimental data with X-ray structures of potassium channels has provided a new level of understanding of the mechanisms of action of sodium channel ligands and has allowed proposing several testable hypotheses.


Assuntos
Anestésicos/química , Batraquiotoxinas/química , Modelos Moleculares , Bloqueadores dos Canais de Sódio/química , Canais de Sódio/química , Tetrodotoxina/química , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Sequência de Aminoácidos , Anestésicos/farmacologia , Batraquiotoxinas/farmacologia , Sítios de Ligação , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Ligantes , Dados de Sequência Molecular , Estrutura Molecular , Ligação Proteica , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/fisiologia , Relação Estrutura-Atividade , Tetrodotoxina/farmacologia
12.
Sleep Med Rev ; 11(5): 361-75, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17646118

RESUMO

Current treatment approaches to the problem of obstructive sleep apnoea (OSA) have limitations. Specifically, invasive anatomical-based surgery and dental appliances typically do not alleviate obstruction at an acceptable rate, and compliance to continuous positive airway pressure (CPAP) devices is frequently suboptimal. Neurotoxinological treatment approaches are widespread in the field of medicine, but as yet have not been evaluated as a treatment for sleep-disordered breathing. In this review, it is argued that despite widespread recognition of the loss of upper airway (UA) muscular tone and/or reflexes in the expression of OSA, most treatment interventions to date have focused on anatomical principles alone. Several hypothesised neurotoxinological interventions aimed at either enhancing UA neuromuscular tone and/or reflexes are proposed, and some preliminary data is presented. Although in its early infancy, with considerable toxicity studies in animals yet to be done, a neurotoxinological approach to the problem of OSA holds promise as a future treatment, with the potential for both high effectiveness and patient compliance.


Assuntos
Músculo Esquelético/efeitos dos fármacos , Neurotoxinas/farmacologia , Neurotoxinas/uso terapêutico , Apneia Obstrutiva do Sono/tratamento farmacológico , Sono/efeitos dos fármacos , Batraquiotoxinas/farmacologia , Batraquiotoxinas/uso terapêutico , Sistemas de Liberação de Medicamentos , Avaliação de Medicamentos , Humanos , Toxinas Marinhas/farmacologia , Toxinas Marinhas/uso terapêutico , Músculo Esquelético/metabolismo , Projetos de Pesquisa , Venenos de Escorpião/farmacologia , Venenos de Escorpião/uso terapêutico , Apneia Obstrutiva do Sono/fisiopatologia , Venenos de Serpentes/farmacologia , Venenos de Serpentes/uso terapêutico , Veratridina/farmacologia , Veratridina/uso terapêutico
13.
Pflugers Arch ; 454(2): 277-87, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17205354

RESUMO

Sequence alignment of four S6 segments in the human cardiac Na+ channel suggests that serine-401 (hNav1.5-S401) at D1S6 along with asparagine-927 (N927) at D2S6, serine-1458 (S1458) at D3S6, and phenylalanine-1760 (F1760) at D4S6 may jointly form a pore-facing S(401)N(927)S(1458)F(1760) ring. Importantly, this pore-facing structure is adjacent to the putative gating-hinge (G(400)G(926)G(1457)S(1759)) and close to the selectivity filter. Within this SNSF ring, only S401 has not yet been identified as a batrachotoxin (BTX) sensing residue. We therefore created S401 mutants with 12 substitutions (S401C,W,P,A,K,F,R,E,L,N,D,G) and assayed their BTX sensitivity. All S401 mutants expressed Na+ currents but often with altered gating characteristics. Ten mutants were found sensitive to 5 muM BTX, which eliminated Na+ channel fast inactivation after repetitive pulses. However, S401K and S401R became BTX resistant. In addition, the block of open and inactivated hNav1.5-S401K Na+ channels by local anesthetic bupivacaine was reduced by approximately 8-10-fold, but not the block of resting Na+ channels. Qualitatively, these ligand-sensing phenotypes of hNav1.5-S401K channels resemble those of S1458K and F1760K channels reported earlier. Together, our results support that residue hNav1.5-S401 at D1S6 is facing the inner cavity and is in close proximity to the receptor sites for BTX and for local anesthetics.


Assuntos
Anestésicos Locais/metabolismo , Batraquiotoxinas/metabolismo , Proteínas Musculares/fisiologia , Serina/metabolismo , Canais de Sódio/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Anestésicos Locais/farmacologia , Batraquiotoxinas/farmacologia , Sítios de Ligação/genética , Bupivacaína/farmacologia , Linhagem Celular , Simulação por Computador , Estimulação Elétrica , Eletrofisiologia , Expressão Gênica/genética , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutagênese Sítio-Dirigida , Canal de Sódio Disparado por Voltagem NAV1.5 , Ligação Proteica , Proteínas Recombinantes/metabolismo , Serina/genética , Sódio/metabolismo , Canais de Sódio/genética , Canais de Sódio/metabolismo , Transfecção
14.
Channels (Austin) ; 1(3): 179-88, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18690024

RESUMO

Batrachotoxin (BTX) not only keeps the voltage-gated Na(+) channel open persistently but also reduces its single-channel conductance. Although a BTX receptor has been delimited within the inner cavity of Na(+) channels, how Na(+) ions flow through the BTX-bound permeation pathway remains unclear. In this report we tested a hypothesis that Na(+) ions traverse a narrow gap between bound BTX and residue N927 at D2S6 of cardiac hNa(v)1.5 Na(+) channels. We found that BTX at 5 microM indeed elicited a strong block of hNa(v)1.5-N927K currents (approximately 70%) after 1000 repetitive pulses (+50 mV/20 ms at 2 Hz) without any effects on Na(+) channel gating. Once occurred, this unique use-dependent block of hNa(v)1.5-N927K Na(+) channels recovered little at holding potential (-140 mV), demonstrating that BTX block is irreversible under our experimental conditions. Such an irreversible effect likewise developed in fast inactivation-deficient hNa(v)1.5-N927K Na(+) channels albeit with a faster on-rate; approximately 90% of peak Na(+) currents were abolished by BTX after 200 repetitive pulses (+50 mV/20 ms). This use-dependent block of fast inactivation-deficient hNa(v)1.5-N927K Na(+) channels by BTX was duration dependent. The longer the pulse duration the larger the block developed. Among N927K/W/R/H/D/S/Q/G/E substitutions in fast inactivation-deficient hNa(v)1.5 Na(+) channels, only N927K/R Na(+) currents were highly sensitive to BTX block. We conclude that (a) BTX binds within the inner cavity and partly occludes the permeation pathway and (b) residue hNa(v)1.5-N927 is critical for ion permeation between bound BTX and D2S6, probably because the side-chain of N927 helps coordinate permeating Na(+) ions.


Assuntos
Batraquiotoxinas/farmacologia , Proteínas Musculares/antagonistas & inibidores , Mutação , Miócitos Cardíacos/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/farmacologia , Sódio/metabolismo , Animais , Asparagina , Batraquiotoxinas/química , Batraquiotoxinas/metabolismo , Sítios de Ligação , Linhagem Celular , Permeabilidade da Membrana Celular , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana , Modelos Moleculares , Estrutura Molecular , Proteínas Musculares/química , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5 , Ligação Proteica , Conformação Proteica , Ratos , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/metabolismo , Canais de Sódio/química , Canais de Sódio/genética , Canais de Sódio/metabolismo , Fatores de Tempo , Transfecção
15.
Mol Pharmacol ; 69(3): 788-95, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16354762

RESUMO

A structural model of the rNav1.4 Na+ channel with batrachotoxin (BTX) bound within the inner cavity suggested that the BTX pyrrole moiety is located between a lysine residue at the DEKA selectivity filter (Lys1237) and an adjacent phenylalanine residue (Phe1236). We tested this pyrrole-binding model by site-directed mutagenesis of Phe1236 at D3/P-loop with 11 amino acids. Mutants F1236D and F1236E expressed poorly, whereas nine other mutants either expressed robust Na+ currents, like the wild-type (F1236Y/Q/K), or somewhat reduced current (F1236G/A/C/N/W/R). Gating properties were altered modestly in most mutant channels, with F1236G displaying the greatest shift in activation and steady-state fast inactivation (-10.1 and -7.5 mV, respectively). Mutants F1236K and F1236R were severely resistant to BTX after 1000 repetitive pulses (+50 mV/20 ms at 2 Hz), whereas seven other mutants were sensitive but with reduced magnitudes compared with the wild type. It is noteworthy that rNav1.4-F1236K mutant Na+ channels remained highly sensitive to block by the local anesthetic bupivacaine, unlike several other BTX-resistant mutant channels. Our data thus support a model in which BTX, when bound within the inner cavity, interacts with the D3/P-loop directly. Such a direct interaction provides clues on how BTX alters the Na+ channel selectivity and conductance.


Assuntos
Batraquiotoxinas/farmacologia , Simulação por Computador , Proteínas Musculares/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Canais de Sódio/efeitos dos fármacos , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Bupivacaína/farmacologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Musculares/química , Proteínas Musculares/genética , Mutação , Fenilalanina/química , Fenilalanina/genética , Conformação Proteica , Ratos , Canais de Sódio/química , Canais de Sódio/genética
16.
FEBS Lett ; 579(20): 4207-12, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16083886

RESUMO

Sodium channel activators, batrachotoxin and veratridine, cause sodium channels to activate easier and stay open longer than normal channels. Traditionally, this was explained by an allosteric mechanism. However, increasing evidence suggests that activators can bind inside the pore. Here, we model the open sodium channel with activators and propose a novel mechanism of their action. The activator-bound channel retains a hydrophilic pathway for ions between the ligand and conserved asparagine in segment S6 of repeat II. One end of the activator approaches the selectivity filter, decreasing the channel conductance and selectivity. The opposite end reaches the gate stabilizing it in the open state.


Assuntos
Batraquiotoxinas/farmacologia , Modelos Moleculares , Agonistas de Canais de Sódio , Canais de Sódio/química , Veratridina/farmacologia , Batraquiotoxinas/química , Batraquiotoxinas/metabolismo , Sítios de Ligação , Transporte Biológico , Cátions Monovalentes/metabolismo , Ligantes , Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Veratridina/química , Veratridina/metabolismo
17.
Anal Biochem ; 342(2): 260-70, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15950910

RESUMO

A 96-well-microplate-based ion flux method utilizing readily available autoradiographic phosphorimaging detection is described. Nicotinic acetylcholine receptor-mediated (22)Na influx in four cultured cell lines provided satisfactory concentration-response data for epibatidine and several other nicotinic agonists. The data were consistent with data obtained using standard 6-well assays. Assays for nicotinic-receptor-mediated (86)Rb efflux produced data similar to data obtained with the (22)Na influx assay. However, assays for (45)Ca influx were not successful, although (45)Ca was readily detected and quantified. Voltage-gated sodium channel-mediated (22)Na influx in a neuroblastoma cell line allowed assay of the effects of such sodium channel activators as batrachotoxin and a pumiliotoxin B/scorpion venom combination. Phosphorimaging detection allows for reliable beta counting of up to 1,200 simultaneous samples with excellent sensitivity and is amenable for application to high-throughput screening.


Assuntos
Autorradiografia/instrumentação , Autorradiografia/métodos , Receptores Nicotínicos/fisiologia , Canais de Sódio/fisiologia , Alcaloides/farmacologia , Batraquiotoxinas/farmacologia , Radioisótopos de Cálcio , Linhagem Celular , Humanos , Indolizinas/farmacologia , Piperidinas/farmacologia , Receptores Nicotínicos/efeitos dos fármacos , Radioisótopos de Rubídio , Venenos de Escorpião/farmacologia , Sensibilidade e Especificidade , Radioisótopos de Sódio
18.
Am J Physiol Heart Circ Physiol ; 289(1): H137-45, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15961372

RESUMO

The presence and properties of voltage-gated Na+ channels in mesenteric artery smooth muscle cells (SMCs) were studied using whole cell patch-clamp recording. SMCs from mouse and rat mesenteric arteries were enzymatically dissociated using two dissociation protocols with different enzyme combinations. Na+ and Ca2+ channel currents were present in myocytes isolated with collagenase and elastase. In contrast, Na+ currents were not detected, but Ca2+ currents were present in cells isolated with papain and collagenase. Ca2+ currents were blocked by nifedipine. The Na+ current was insensitive to nifedipine, sensitive to changes in the extracellular Na+ concentration, and blocked by tetrodotoxin with an IC50 at 4.3 nM. The Na+ conductance was half maximally activated at -16 mV, and steady-state inactivation was half-maximal at -53 mV. These values are similar to those reported in various SMC types. In the presence of 1 microM batrachotoxin, the Na+ conductance-voltage relationship was shifted by 27 mV in the hyperpolarizing direction, inactivation was almost completely eliminated, and the deactivation rate was decreased. The present study indicates that TTX-sensitive, voltage-gated Na+ channels are present in SMCs from the rat and mouse mesenteric artery. The presence of these channels in freshly isolated SMC depends critically on the enzymatic dissociation conditions. This could resolve controversy about the presence of Na+ channels in arterial smooth muscle.


Assuntos
Ativação do Canal Iônico , Artérias Mesentéricas/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo , Tetrodotoxina/farmacologia , Animais , Batraquiotoxinas/farmacologia , Condutividade Elétrica , Eletrofisiologia , Homeostase , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Sódio/metabolismo , Sódio/farmacologia
19.
FEBS Lett ; 577(1-2): 245-8, 2004 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-15527793

RESUMO

Batrachotoxin is a potent modulator of voltage-gated sodium channels, leading to irreversible depolarisation of nerves and muscles, fibrillation, arrhythmias and eventually cardiac failure. Since its discovery, field researchers also reported numbness after their skin came into contact with this toxin. Intrigued by this phenomenon, we determined the effect of batrachotoxin on the voltage-gated sodium channel Nav1.8, which is considered to be a key player in nociception. As a result, we discovered that batrachotoxin profoundly modulates this channel: the inactivation process is severely altered, the voltage-dependence of activation is shifted towards more hyperpolarised potentials resulting in the opening of Nav1.8 at more negative membrane potentials and the ion selectivity is modified.


Assuntos
Batraquiotoxinas/farmacologia , Canais de Sódio/efeitos dos fármacos , Animais , Anuros , Potenciais da Membrana , Técnicas de Patch-Clamp , Xenopus laevis
20.
J Anesth ; 18(2): 100-6, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15127257

RESUMO

PURPOSE: To investigate the response to general anesthetics of different sodium-channel subtypes, we examined the effects of pentobarbital, a close thiopental analogue, on single sodium channels from human skeletal muscle and compared them to existing data from human brain and human ventricular muscle channels. METHODS: Sodium channels from a preparation of human skeletal muscle were incorporated into planar lipid bilayers, and the steady-state behavior of single sodium channels and their response to pentobarbital was examined in the presence of batrachotoxin, a sodium-channel activator. Single-channel currents were recorded before and after the addition of pentobarbital (0.34-1.34 mM). RESULTS: In symmetrical 500 mM NaCl, human skeletal muscle sodium channels had an averaged single-channel conductance of 21.0 +/- 0.6 pS, and the channel fractional open time was 0.96 +/- 0.04. The activation midpoint potential was -96.2 +/- 1.6 mV. Extracellular tetrodotoxin blocked the channel with a half-maximal concentration (k1/2) of 60 nM at 0 mV. Pentobarbital reduced the time-averaged conductance of single skeletal muscle sodium channels in a concentration-dependent manner (inhibitory concentration 50% [IC50] = 0.66 mM). The steady-state activation was shifted to more hyperpolarized potentials (-16.7 mV at 0.67 mM pentobarbital). CONCLUSION: In the planar lipid bilayer system, skeletal muscle sodium channels have some electrophysiological properties that are significantly different compared with those of sodium channels from cardiac or from central nervous tissue. In contrast to the control data, these different human sodium channel subtypes showed the same qualitative and quantitative response to the general anesthetic pentobarbital. The implication of these effects for overall anesthesia will depend on the role the individual channels play within their neuronal networks, but suppression of both central nervous system and peripheral sodium channels may add to general anesthetic effects.


Assuntos
Anestésicos Gerais/farmacologia , Músculo Esquelético/fisiologia , Pentobarbital/farmacologia , Canais de Sódio/efeitos dos fármacos , Animais , Batraquiotoxinas/farmacologia , Encéfalo/metabolismo , Bovinos , Eletrofisiologia , Ventrículos do Coração/metabolismo , Humanos , Técnicas In Vitro , Perna (Membro) , Bicamadas Lipídicas , Potenciais da Membrana/efeitos dos fármacos , Músculo Esquelético/inervação , Condução Nervosa/efeitos dos fármacos , Canais de Sódio/metabolismo , Canais de Sódio/fisiologia , Tetrodotoxina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...