Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 293
Filter
1.
Molecules ; 27(13)2022 Jun 28.
Article in English | MEDLINE | ID: mdl-35807390

ABSTRACT

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.


Subject(s)
Alkaloids , Fluorescent Dyes , Alkaloids/pharmacology , Batrachotoxins/metabolism , Batrachotoxins/pharmacology , Bias , HEK293 Cells , Humans , Isoquinolines/pharmacology , Ligands , Sodium/metabolism
2.
Curr Top Membr ; 78: 117-44, 2016.
Article in English | MEDLINE | ID: mdl-27586283

ABSTRACT

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.


Subject(s)
Voltage-Gated Sodium Channels/metabolism , Animals , Batrachotoxins/chemistry , Batrachotoxins/metabolism , Batrachotoxins/pharmacology , Binding Sites , Conotoxins/chemistry , Conotoxins/metabolism , Conotoxins/toxicity , Insecticides/chemistry , Insecticides/metabolism , Insecticides/toxicity , Ion Channel Gating/drug effects , Ligands , Molecular Dynamics Simulation , Monte Carlo Method , Protein Structure, Tertiary , Pyrethrins/chemistry , Pyrethrins/metabolism , Pyrethrins/toxicity , Steroids/chemistry , Steroids/metabolism , Tetrodotoxin/chemistry , Tetrodotoxin/metabolism , Tetrodotoxin/toxicity , Voltage-Gated Sodium Channel Agonists/chemistry , Voltage-Gated Sodium Channel Agonists/metabolism , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/metabolism , Voltage-Gated Sodium Channels/chemistry
3.
Cell Chem Biol ; 29(4): 615-624.e5, 2022 04 21.
Article in English | MEDLINE | ID: mdl-34963066

ABSTRACT

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.


Subject(s)
Batrachotoxins , Voltage-Gated Sodium Channels , Batrachotoxins/pharmacology , Esters , Sodium/metabolism
4.
Science ; 258(5080): 310-3, 1992 Oct 09.
Article in English | MEDLINE | ID: mdl-1357749

ABSTRACT

Presynaptic calcium channels are crucial elements of neuronal excitation-secretion coupling. In mammalian brain, they have been difficult to characterize because most presynaptic terminals are too small to probe with electrodes, and available pharmacological tools such as dihydropyridines and omega-conotoxin are largely ineffective. Subsecond measurements of synaptosomal glutamate release have now been used to assess presynaptic calcium channel activity in order to study the action of peptide toxins from the venom of the funnel web spider Agelenopsis aperta, which is known to inhibit dihydropyridine and omega-conotoxin-resistant neuronal calcium currents. A presynaptic calcium channel important in glutamate release is shown to be omega-Aga-IVA sensitive and omega-conotoxin resistant.


Subject(s)
Calcium Channels/physiology , Glutamates/metabolism , Spider Venoms/pharmacology , Agatoxins , Animals , Batrachotoxins/pharmacology , Brain/physiology , Brain/ultrastructure , Calcium/pharmacology , Egtazic Acid/pharmacology , Frontal Lobe/ultrastructure , Glutamic Acid , Kinetics , Mollusk Venoms/pharmacology , Potassium Chloride/pharmacology , Rats , Synaptosomes/physiology , omega-Agatoxin IVA , omega-Conotoxin GVIA
5.
Science ; 187(4181): 1087-9, 1975 Mar 21.
Article in English | MEDLINE | ID: mdl-46619

ABSTRACT

Batrachotoxin (BTX) irreversibly blocks fast axoplasmic transport in nerve in concentrations as low as 0.2 micromolar. The action of BTX was studied in cat sciatic nerves in vitro by measuring the rate of the crest outflow after injection of the L7 dorsal root ganglion with [3-H]leucine. Tetrodotoxin, which in itself does not affect fast axoplasmic transport, inhibited the blocking action of BTX. Unlike the BTX block of nerve and muscle membrane excitability brought about through increased permeability to sodium ion, the BTX block of fast axoplasmic transport occurs with or without sodium ion in the medium. High concentrations of calcium ion protected against the blocking action of BTX, while magnesium ion did not. An action of BTX on the transport mechanism inside the fibers was indicated by the small reduction of adenosine triphosphate plus creatine phosphate, which in itself did not account for the block of axoplasmic transport.


Subject(s)
Axonal Transport/drug effects , Batrachotoxins/pharmacology , Neurons/drug effects , Animals , Calcium/pharmacology , Cats , Depression, Chemical , Leucine/metabolism , Magnesium/pharmacology , Sciatic Nerve/drug effects , Tetrodotoxin/pharmacology , Tritium
6.
Science ; 208(4450): 1383-5, 1980 Jun 20.
Article in English | MEDLINE | ID: mdl-6246586

ABSTRACT

Batrachotoxin is present in remarkably high amounts in the skin of Phyllobates terribilis. Levels of batrachotoxin tend to be reduced when P. terribilis is maintained in captivity, but even after being confined for up to 6 years, these frogs were still at least five times more toxic than other Phyllobates species used by natives for poisoning blowgun darts. Batrachotoxin was not detectable in F1 progeny reared to maturity in captivity. Nerve and muscle preparations from wild-caught frogs and from the nontoxic F1 frogs were both insensitive to batrachotoxin. The regulatory site controlling sodium-channel activation and permeability appears to have been minimally altered to prevent interaction with batrachotoxin, but is still sensitive to other sodium conductance activators (veratridine, grayanotoxin) to which the frogs arenot exposed naturally.


Subject(s)
Anura/physiology , Batrachotoxins/pharmacology , Diterpenes/pharmacology , Ion Channels/drug effects , Veratridine/pharmacology , Veratrine/analogs & derivatives , Age Factors , Animals , Batrachotoxins/metabolism , Membrane Potentials/drug effects , Motor Endplate/drug effects , Synaptic Transmission/drug effects
7.
J Gen Physiol ; 151(2): 186-199, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30587506

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Batrachotoxins/pharmacology , Sodium Channel Agonists/pharmacology , Sodium Channels/metabolism , Bacillus , Bacterial Proteins/agonists , Bacterial Proteins/chemistry , Cell Line , Humans , Ion Channel Gating , Rhodobacteraceae , Sodium Channels/chemistry
8.
Neuron ; 16(2): 407-13, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8789955

ABSTRACT

Few experimental data illuminate the relationship between the molecular structures that mediate ion conduction through voltage-dependent ion channels and the structures responsible for sensing transmembrane voltage and controlling gating. To fill this void, we have used a strongly cationic, mutated mu-conotoxin peptide, which only partially blocks current through voltage-dependent sodium channels, to study voltage-dependent activation gating in both bound and unbound channels. When the peptide binds to the ion-conducting pore, it inhibit channel opening, necessitating stronger depolarization for channel activation. We show that this activation shift could result entirely from electrostatic inhibition of the movement of the voltage-sensing S4 charges and estimate the approximate physical distance through which the S4 charges move.


Subject(s)
Peptides, Cyclic/pharmacology , Sodium Channels/drug effects , Sodium Channels/physiology , Animals , Batrachotoxins/pharmacology , Calcium/pharmacology , Charybdotoxin/pharmacology , Diethylamines/pharmacology , Electric Conductivity , Electrophysiology , Mathematics , Peptides, Cyclic/metabolism , Rats
9.
Brain Res ; 1203: 61-7, 2008 Apr 08.
Article in English | MEDLINE | ID: mdl-18321475

ABSTRACT

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.


Subject(s)
Batrachotoxins/pharmacology , Ginsenosides/pharmacology , Ion Channel Gating/drug effects , Muscle Proteins/physiology , Neurotoxins/pharmacology , Sodium Channels/physiology , Animals , Batrachotoxins/chemistry , Binding Sites/drug effects , Dose-Response Relationship, Drug , Ginsenosides/chemistry , Ion Channel Gating/physiology , Leucine/genetics , Lysine/genetics , Mice , Microinjections , Muscle Proteins/genetics , Muscle, Skeletal , NAV1.2 Voltage-Gated Sodium Channel , Nerve Tissue Proteins , Oocytes , Patch-Clamp Techniques , Point Mutation/physiology , Protein Structure, Tertiary/physiology , Rats , Sodium Channels/genetics , Xenopus laevis
10.
Sleep Med Rev ; 11(5): 361-75, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17646118

ABSTRACT

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.


Subject(s)
Muscle, Skeletal/drug effects , Neurotoxins/pharmacology , Neurotoxins/therapeutic use , Sleep Apnea, Obstructive/drug therapy , Sleep/drug effects , Batrachotoxins/pharmacology , Batrachotoxins/therapeutic use , Drug Delivery Systems , Drug Evaluation , Humans , Marine Toxins/pharmacology , Marine Toxins/therapeutic use , Muscle, Skeletal/metabolism , Research Design , Scorpion Venoms/pharmacology , Scorpion Venoms/therapeutic use , Sleep Apnea, Obstructive/physiopathology , Snake Venoms/pharmacology , Snake Venoms/therapeutic use , Veratridine/pharmacology , Veratridine/therapeutic use
11.
Science ; 354(6314): 865-869, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27856903

ABSTRACT

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.


Subject(s)
Batrachotoxins/chemical synthesis , Batrachotoxins/pharmacology , Muscle Proteins/antagonists & inhibitors , Voltage-Gated Sodium Channel Blockers/chemical synthesis , Voltage-Gated Sodium Channel Blockers/pharmacology , Animals , Binding Sites , Muscle Proteins/chemistry , Muscle Proteins/genetics , Point Mutation , Protein Structure, Secondary , Rats , Sodium Channels/chemistry , Sodium Channels/genetics , Voltage-Gated Sodium Channel Blockers/chemistry
12.
ACS Chem Neurosci ; 7(10): 1463-1468, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27501251

ABSTRACT

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.


Subject(s)
Batrachotoxins/analysis , Batrachotoxins/pharmacology , Sodium Channel Blockers/pharmacology , Animals , Batrachotoxins/chemical synthesis , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Models, Molecular , Molecular Structure , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation , Patch-Clamp Techniques , Rats , Sodium Channel Blockers/chemical synthesis , Sodium Channels/genetics , Sodium Channels/metabolism , Structure-Activity Relationship
13.
Biochim Biophys Acta ; 648(1): 93-9, 1981 Oct 20.
Article in English | MEDLINE | ID: mdl-6271214

ABSTRACT

(1) Single myelinated nerve fibers of Rana esculenta were treated with the steroidal alkaloid batrachotoxin, and Na+ currents and Na+-current fluctuations were measured near the resting potential under voltage-clamp conditions. Between test pulses the fibres were held at hyperpolarizing membrane potentials. (2) The spectral density of Na+-current fluctuations was fitted by the sum of a 1/f component and a Lorentzian function. The time constant tau c = 1/(2 pi fc) obtained from the corner frequency fc of the Lorentzian function approximately agreed with the activation time constant tau m of the macroscopic currents. (3) The conductance gamma of a single Na+ channel modified by batrachotoxin was calculated from the integral of the Lorentzian function and the steady-state Na+ current. At the resting potential V = 0 we obtained gamma - 1.6 pS, higher gamma-values of 3.2 and 3.45 pS were found at V = --8 and --16 mV, respectively. (4) The conductance of a modified Na+ channel is significantly lower than the values 6.4 to 8.85 pS reported in the literature for normal Na+ channels. Hence, our experiments are in agreement with the view that batrachotoxin acts in an 'all-or-none' manner on Na+ channels and creates a distinct population of modified channels.


Subject(s)
Batrachotoxins/pharmacology , Ion Channels/drug effects , Nerve Fibers, Myelinated/metabolism , Sodium/metabolism , Animals , Electric Conductivity , Membrane Potentials , Rana esculenta
14.
Biochim Biophys Acta ; 1194(2): 215-22, 1994 Sep 14.
Article in English | MEDLINE | ID: mdl-7918535

ABSTRACT

The voltage-dependent action of the intravenous anesthetic pentobarbital on human brain sodium channels activated by batrachotoxin was examined using planar lipid bilayer methods. Fractional open time-data were fitted by Boltzmann functions to yield simple parameters characterizing the voltage-dependence of the fractional open time. Pentobarbital caused a dose-dependent reduction of the maximum fractional open time of the sodium channel and a shift of the potential of half-maximal open time towards hyperpolarized potentials, whereas the slope parameter of the Boltzmann-fits was unaffected. A statistically significant increase of the variability of these parameters was found only in the case of the maximum fractional open time, indicating a random fluctuation of pentobarbital-induced suppression of the sodium channels over time. The voltage-dependent action of pentobarbital probably results from either a pentobarbital-modification of channel activation gating and/or a modification of the pentobarbital action by the gating process itself.


Subject(s)
Batrachotoxins/pharmacology , Brain/drug effects , Pentobarbital/pharmacology , Sodium Channels/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Humans , Lipid Bilayers/metabolism , Membrane Potentials
15.
Biochim Biophys Acta ; 858(1): 31-7, 1986 Jun 13.
Article in English | MEDLINE | ID: mdl-2423130

ABSTRACT

We studied the dose-response relationship between gamma radiation and batrachotoxin-stimulated sodium influx in neuroblastoma cells in tissue culture. We also tested the hypothesis that changes in sodium channel conformation may alter the radiosensitivity of the channel. We found that gamma radiation inhibited toxin-stimulated 22Na uptake at doses beyond a threshold of 200-300 Gy. No effects were seen following doses below 100 Gy. This inhibition of sodium permeability was seen when the cells were irradiated with sodium channels in the closed or inactivated, nonconducting states. However, when the channels were in the toxin-opened, conducting state, gamma radiation had no effect at doses up to 2000 Gy. Our results support earlier electrophysiological studies that showed that high doses of ionizing radiation are required to produce a measurable decrease in sodium permeability. In addition, our data suggest that by changing the sodium channel conformation, batrachotoxin appears to alter radiosensitive chemical bonds in the gating or ion-conducting portion of the channel.


Subject(s)
Ion Channels/radiation effects , Sodium/physiology , Batrachotoxins/pharmacology , Cell Line , Dose-Response Relationship, Radiation , Electric Conductivity , Gamma Rays , Ion Channels/drug effects , Membrane Potentials/drug effects , Membrane Potentials/radiation effects , Neuroblastoma , Protein Conformation
16.
Biochim Biophys Acta ; 883(1): 15-25, 1986 Aug 06.
Article in English | MEDLINE | ID: mdl-2425852

ABSTRACT

Activation of alpha 1-adrenergic receptors by norepinephrine in guinea pig cortical synaptoneurosomes augments accumulations of cyclic AMP elicited by 2-chloroadenosine and concomitantly increases formation of inositol phosphates. Various agents that affect calcium channels or sites of action of calcium have little or no effect on cyclic AMP accumulation elicited either with 2-chloroadenosine, or with a 2-chloroadenosine/norepinephrine combination, nor did they markedly affect formation of inositol phosphates elicited by norepinephrine. However, EGTA reduces both cyclic AMP accumulation and inositol phosphate formation. Agents such as batrachotoxin, scorpion (Leiurus) venom and pumiliotoxin B that are active at voltage-dependent sodium channels enhance accumulations of cyclic AMP and inositol phosphates. These effects are blocked by tetrodotoxin. It is proposed that enhanced influx of sodium ions increases phosphatidylinositol metabolism, resulting in formation of diacylglycerols and inositol phosphates, and that the former, through activation of protein kinase, causes an enhancement of cyclic AMP accumulations in brain tissue.


Subject(s)
Cerebral Cortex/metabolism , Cyclic AMP/metabolism , Indolizines , Inositol Phosphates/metabolism , Ion Channels/physiology , Piperidines , Sodium/metabolism , Sugar Phosphates/metabolism , Synaptosomes/metabolism , 2-Chloroadenosine , Adenosine/analogs & derivatives , Adenosine/pharmacology , Alkaloids/pharmacology , Animals , Batrachotoxins/pharmacology , Calcium/physiology , Egtazic Acid/pharmacology , Guinea Pigs , Ion Channels/drug effects , Male , Norepinephrine/pharmacology , Scorpion Venoms/pharmacology , Tetrodotoxin/pharmacology
17.
FEBS Lett ; 579(20): 4207-12, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16083886

ABSTRACT

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.


Subject(s)
Batrachotoxins/pharmacology , Models, Molecular , Sodium Channel Agonists , Sodium Channels/chemistry , Veratridine/pharmacology , Batrachotoxins/chemistry , Batrachotoxins/metabolism , Binding Sites , Biological Transport , Cations, Monovalent/metabolism , Ligands , Sodium/metabolism , Sodium Channel Blockers/pharmacology , Veratridine/chemistry , Veratridine/metabolism
18.
J Gen Physiol ; 82(3): 365-84, 1983 Sep.
Article in English | MEDLINE | ID: mdl-6313850

ABSTRACT

The expression of Na+ channels during differentiation of cultured embryonic chick skeletal muscle cells was investigated using saxitoxin (STX) and batrachotoxin (BTX), which previously have been shown to interact with distinct, separate receptor sites of the voltage-sensitive Na+ channel of excitable cells. In the present study, parallel measurements of binding of [3H]-STX (STX) and of BTX-activated 22Na+ uptake (Na influx) were made in order to establish the temporal relationship of the appearance of these two Na+ channel activities during myogenesis. Na influx was clearly measurable in 2-d cells; from day 3 to day 7 the maximum Na influx approximately doubled when measured with saturating BTX concentrations potentiated by Leiurus scorpion toxin, while the apparent affinity of BTX, measured without scorpion toxin, also increased. Saturable STX binding did not appear consistently until day 3; from then until day 7 the STX binding capacity increased about threefold, whereas the equilibrium dissociation constant (KD) decreased about fourfold. Although Na influx in cells of all ages was totally inhibited by STX or tetrodotoxin (TTX) at 10 microM, lower concentrations (2-50 nM) blocked the influx in 7-d cells much more effectively than that in 3-d cells, where half the flux was resistant to STX at 20-50 nM. Similar but smaller differences characterized the block by TTX. In addition, when protein synthesis is inhibited by cycloheximide, both Na influx and STX binding activities disappear more rapidly in 3-d than in 7-d cells, which shows that these functions are less stable metabolically in the younger cells.


Subject(s)
Ion Channels/metabolism , Muscles/embryology , Sodium/metabolism , Animals , Batrachotoxins/pharmacology , Catalysis , Cells, Cultured , Chick Embryo , Muscles/cytology , Saxitoxin/metabolism , Time Factors
19.
J Gen Physiol ; 97(3): 605-25, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1645396

ABSTRACT

Na+ permeation through normal and batrachotoxin (BTX)-modified squid axon Na+ channels was characterized. Unmodified and toxin-modified Na+ channels were studied simultaneously in outside-out membrane patches using the cut-open axon technique. Current-voltage relations for both normal and BTX-modified channels were measured over a wide range of Na+ concentrations and voltages. Channel conductance as a function of Na+ concentration curves showed that within the range 0.015-1 M Na+ the normal channel conductance is 1.7-2.5-fold larger than the BTX-modified conductance. These relations cannot be fitted by a simple Langmuir isotherm. Channel conductance at low concentrations was larger than expected from a Michaelis-Menten behavior. The deviations from the simple case were accounted for by fixed negative charges located in the vicinity of the channel entrances. Fixed negative charges near the pore mouths would have the effect of increasing the local Na+ concentration. The results are discussed in terms of energy profiles with three barriers and two sites, taking into consideration the effect of the fixed negative charges. Either single- or multi-ion pore models can account for all the permeation data obtained in both symmetric and asymmetric conditions. In a temperature range of 5-15 degrees C, the estimated Q10 for the conductance of the BTX-modified Na+ channel was 1.53. BTX appears not to change the Na+ channel ion selectively (for the conditions used) or the surface charge located near the channel entrances.


Subject(s)
Axons/metabolism , Sodium Channels/metabolism , Sodium/metabolism , Animals , Axons/physiology , Batrachotoxins/pharmacology , Biological Transport, Active , Decapodiformes , Electrophysiology , Models, Biological , Sodium/physiology , Sodium Channels/physiology , Temperature
20.
J Gen Physiol ; 105(2): 267-87, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7760019

ABSTRACT

We have synthesized a model local anesthetic (LA), N-(2-di-N-butyl-aminoethyl)-4-azidobenzamide (DNB-AB), containing the photoactivatable aryl azido moiety, which is known to form a covalent bond to adjacent molecules when exposed to UV light (Fleet, G.W., J.R. Knowles, and R.R. Porter. 1972. Biochemical Journal. 128:499-508. Ji, T.H. 1979. Biochimica et Biophysica Acta. 559:39-69). We studied the effects of DNB-AB on the sodium current (INa) under whole-cell voltage clamp in clonal mammalian GH3 cells and on 3[H]-BTX-B binding to sheep brain synaptoneurosomes. In the absence of UV illumination, DNB-AB behaved similarly to known LAs, producing both reversible block of peak INa (IC50 = 26 microM, 20 degrees C) and reversible inhibition of 3[H]-BTX-B (50 nM in the presence of 0.12 microgram/liter Leiurus quinquestriatus scorpion venom) binding (IC50 = 3.3 microM, 37 degrees C), implying a noncovalent association between DNB-AB and its receptor(s). After exposure to UV light, both block of INa and inhibition of 3[H]-BTX-B binding were only partially reversible (INa = 42% of control; 3[H]-BTX-B binding = 23% of control) showing evidence of a light-dependent, covalent association between DNB-AB and its receptor(s). In the absence of drug, UV light had less effect on INa (post exposure INa = 96% of control) or on 3[H]-BTX-B binding (post exposure binding = 70% of control). The irreversible block of INa was partially protected by coincubation of DNB-AB with 1 mM bupivacaine (IC50 = 45 microM, for INa inhibition at 20 degrees C, Wang, G.K., and S.Y. Wang. 1992. Journal of General Physiology. 100:1003-1020), (post exposure INa = 73% of control). The irreversible inhibition of 3[H]-BTX-B binding also was partially protected by coincubation with bupivacaine (500 microM, 37 degrees C) (post exposure binding = 51% of control), suggesting that the site of irreversible inhibition of both INa and 3[H]-BTX-B binding is shared with the clinical LA bupivacaine.


Subject(s)
Affinity Labels/chemical synthesis , Anesthetics, Local/chemistry , Anesthetics, Local/pharmacology , Azides/pharmacology , Batrachotoxins/pharmacology , Sodium Channel Blockers , Animals , Azides/chemical synthesis , Azides/chemistry , Binding Sites/physiology , Bupivacaine/pharmacology , Photochemistry , Rabbits , Synaptosomes/physiology , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL