Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.613
Filter
Add more filters

Publication year range
1.
Am J Physiol Cell Physiol ; 323(3): C749-C762, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35876287

ABSTRACT

Isolated smooth muscle cells (SMCs) from mouse bronchus were studied using the whole cell patch-clamp technique at ∼21°C. Stepping from -100 mV to -20 mV evoked inward currents of mean amplitude -275 pA. These inactivated (tau = 1.1 ms) and were abolished when external Na+ was substituted with N-Methyl-d-glucamine. In current-voltage protocols, current peaked at -10 mV and reversed between +20 and +30 mV. The V1/2s of activation and inactivation were -25 and -86 mV, respectively. The current was highly sensitive to tetrodotoxin (IC50 = 1.5 nM) and the NaV1.7 subtype-selective blocker, PF-05089771 (IC50 = 8.6 nM), consistent with NaV1.7 as the underlying pore-forming α subunit. Two NaV1.7-selective antibodies caused membrane-delineated staining of isolated SMC, as did a nonselective pan-NaV antibody. RT-PCR, performed on groups of ∼15 isolated SMCs, revealed transcripts for NaV1.7 in 7/8 samples. Veratridine (30 µM), a nonselective NaV channel activator, reduced peak current evoked by depolarization but induced a sustained current of 40 pA. Both effects were reversed by tetrodotoxin (100 nM). In tension experiments, veratridine (10 µM) induced contractions that were entirely blocked by atropine (1 µM). However, in the presence of atropine, veratridine was able to modulate the pattern of activity induced by a combination of U-46619 (a thromboxane A2 mimetic) and PGE2 (prostaglandin E2), by eliminating bursts in favor of sustained phasic contractions. These effects were readily reversed to control-like activity by tetrodotoxin (100 nM). In conclusion, mouse bronchial SMCs functionally express NaV1.7 channels that are capable of modulating contractile activity, at least under experimental conditions.


Subject(s)
Bronchi , Myocytes, Smooth Muscle , Animals , Atropine Derivatives/metabolism , Atropine Derivatives/pharmacology , Bronchi/metabolism , Mice , Myocytes, Smooth Muscle/metabolism , Sodium/metabolism , Tetrodotoxin/metabolism , Tetrodotoxin/pharmacology , Veratridine/metabolism , Veratridine/pharmacology
2.
Int J Mol Sci ; 23(4)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35216338

ABSTRACT

The cardiac sodium ion channel (NaV1.5) is a protein with four domains (DI-DIV), each with six transmembrane segments. Its opening and subsequent inactivation results in the brief rapid influx of Na+ ions resulting in the depolarization of cardiomyocytes. The neurotoxin veratridine (VTD) inhibits NaV1.5 inactivation resulting in longer channel opening times, and potentially fatal action potential prolongation. VTD is predicted to bind at the channel pore, but alternative binding sites have not been ruled out. To determine the binding site of VTD on NaV1.5, we perform docking calculations and high-throughput electrophysiology experiments in the present study. The docking calculations identified two distinct binding regions. The first site was in the pore, close to the binding site of NaV1.4 and NaV1.5 blocking drugs in experimental structures. The second site was at the "mouth" of the pore at the cytosolic side, partly solvent-exposed. Mutations at this site (L409, E417, and I1466) had large effects on VTD binding, while residues deeper in the pore had no effect, consistent with VTD binding at the mouth site. Overall, our results suggest a VTD binding site close to the cytoplasmic mouth of the channel pore. Binding at this alternative site might indicate an allosteric inactivation mechanism for VTD at NaV1.5.


Subject(s)
Mouth/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Sodium/metabolism , Veratridine/pharmacology , Binding Sites/physiology , Cell Line , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Neurotoxins/pharmacology
3.
Mol Pharm ; 16(9): 3948-3956, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31361498

ABSTRACT

Many oral mucosal conditions cause considerable and prolonged pain that to date has been difficult to alleviate via topical delivery, and the use of injection causes many patients dental anxiety and needle-prick pain. Therefore, developing a noninjectable drug delivery system as an alternative administration procedure may vastly improve the health and wellbeing of these patients. Recent advances in the development of mucoadhesive electrospun patches for the direct delivery of therapeutics to the oral mucosa offer a potential solution, but as yet, the release of local anesthetics from this system and their uptake by oral tissue have not been demonstrated. Here, we demonstrate the fabrication of lidocaine-loaded electrospun fiber patches, drug release, and subsequent uptake and permeation through the porcine buccal mucosa. Lidocaine HCl and lidocaine base were incorporated into the electrospun patches to evaluate the difference in drug permeation for the two drug compositions. Lidocaine released from the lidocaine HCl-containing electrospun patches was significantly quicker than from the lidocaine base patches, with double the amount of drug released from the lidocaine HCl patches in the first 15 min (0.16 ± 0.04 mg) compared to that from the lidocaine base patches (0.07 ± 0.01 mg). The permeation of lidocaine from the lidocaine HCl electrospun patches through ex vivo porcine buccal mucosa was also detected in 15 min, whereas permeation of lidocaine from the lidocaine base patch was not detected. Matrix-assisted laser desorption ionization-mass spectrometry imaging was used to investigate localization of lidocaine within the oral tissue. Lidocaine in the solution as well as from the mucoadhesive patch penetrated into the buccal mucosal tissue in a time-dependent manner and was detectable in the lamina propria after only 15 min. Moreover, the lidocaine released from lidocaine HCl electrospun patches retained biological activity, inhibiting veratridine-mediated opening of voltage-gated sodium channels in SH-SY5Y neuroblastoma cells. These data suggest that a mucoadhesive electrospun patch may be used as a vehicle for rapid uptake and sustained anesthetic drug delivery to treat or prevent oral pain.


Subject(s)
Anesthetics/pharmacokinetics , Drug Delivery Systems/methods , Lidocaine/pharmacokinetics , Mouth Mucosa/drug effects , Oral Mucosal Absorption/physiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Administration, Buccal , Anesthetics/administration & dosage , Animals , Cell Line, Tumor , Drug Liberation , Facial Pain/drug therapy , Humans , Lidocaine/administration & dosage , Mouth Mucosa/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Swine , Tissue Distribution , Veratridine/pharmacology , Voltage-Gated Sodium Channel Agonists/pharmacology , Voltage-Gated Sodium Channel Blockers/administration & dosage
4.
Acta Pharmacol Sin ; 39(11): 1716-1724, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29950616

ABSTRACT

Veratridine is a lipid-soluble neurotoxin derived from plants in the family Liliaceae. It has been broadly investigated for its action as a sodium channel agonist. However, the effects of veratridine on subtypes of sodium channels, especially Nav1.7, remain to be studied. Here, we investigated the effects of veratridine on human Nav1.7 ectopically expressed in HEK293A cells and recorded Nav1.7 currents from the cells using whole-cell patch clamp technique. We found that veratridine exerted a dose-dependent inhibitory effect on the peak current of Nav1.7, with the half-maximal inhibition concentration (IC50) of 18.39 µM. Meanwhile, veratridine also elicited tail current (linearly) and sustained current [half-maximal concentration (EC50): 9.53 µM], also in a dose-dependent manner. Veratridine (75 µM) shifted the half-maximal activation voltage of the Nav1.7 activation curve in the hyperpolarized direction, from -21.64 ± 0.75 mV to -28.14 ± 0.66 mV, and shifted the half-inactivation voltage of the steady-state inactivation curve from -59.39 ± 0.39 mV to -73.78 ± 0.5 mV. An increased frequency of stimulation decreased the peak and tail currents of Nav1.7 for each pulse along with pulse number, and increased the accumulated tail current at the end of train stimulation. These findings reveal the different modulatory effects of veratridine on the Nav1.7 peak current and tail current.


Subject(s)
Ion Channel Gating/drug effects , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Veratridine/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Humans
5.
Bioorg Med Chem Lett ; 27(5): 1247-1251, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28143690

ABSTRACT

Crambescin B carboxylic acid, a synthetic analog of crambescin B, was recently found to inhibit the voltage-sensitive sodium channels (VSSC) in a cell-based assay using neuroblastoma Neuro 2A cells. In the present study, whole-cell patch-clamp recordings were conducted with three heterologously expressed VSSC subtypes, Nav1.2, Nav1.6 and Nav1.7, in a human embryonic kidney cell line HEK293T to further characterize the inhibition of VSSC by crambescin B carboxylic acid. Contrary to the previous observation, crambescin B carboxylic acid did not inhibit peak current evoked by depolarization from the holding potential of -100mV to the test potential of -10mV in the absence or presence of veratridine (VTD). In the presence of VTD, however, crambescin B carboxylic acid diminished VTD-induced sustained and tail currents through the three VSSC subtypes in a dose-dependent manner, whereas TTX inhibited both the peak current and the VTD-induced sustained and tail currents through all subtypes of VSSC tested. We thus concluded that crambescin B carboxylic acid does not block VSSC in a similar manner to TTX but modulate the action of VTD, thereby causing an apparent block of VSSC in the cell-based assay.


Subject(s)
Pyrimidines/pharmacology , Spiro Compounds/pharmacology , Veratridine/chemistry , Voltage-Gated Sodium Channels/drug effects , Animals , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Inhibitory Concentration 50 , Molecular Structure , Pyrimidines/chemistry , Spiro Compounds/chemistry , Veratridine/pharmacology , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacology
6.
J Neurochem ; 137(2): 177-89, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26821584

ABSTRACT

In epilepsy, the GABA and glutamate balance may be disrupted and a transient decrease in extracellular calcium occurs before and during a seizure. Flow Cytometry based fluorescence activated particle sorting experiments quantified synaptosomes from human neocortical tissue, from both epileptic and non-epileptic patients (27.7% vs. 36.9% GABAergic synaptosomes, respectively). Transporter-mediated release of GABA in human and rat neocortical synaptosomes was measured using the superfusion technique for the measurement of endogenous GABA. GABA release was evoked by either a sodium channel activator or a sodium/potassium-ATPase inhibitor when exocytosis was possible or prevented, and when the sodium/calcium exchanger was active or inhibited. The transporter-mediated release of GABA is because of elevated intracellular sodium. A reduction in the extracellular calcium increased this release (in both non-epileptic and epileptic, except Rasmussen encephalitis, synaptosomes). The inverse was seen during calcium doubling. In humans, GABA release was not affected by exocytosis inhibition, that is, it was solely transporter-mediated. However, in rat synaptosomes, an increase in GABA release at zero calcium was only exhibited when the exocytosis was prevented. The absence of calcium amplified the sodium/calcium exchanger activity, leading to elevated intracellular sodium, which, together with the stimulation-evoked intracellular sodium increment, enhanced GABA transporter reversal. Sodium/calcium exchange inhibitors diminished GABA release. Thus, an important seizure-induced extracellular calcium reduction might trigger a transporter- and sodium/calcium exchanger-related anti-seizure mechanism by augmenting transporter-mediated GABA release, a mechanism absent in rats. Uniquely, the additional increase in GABA release because of calcium-withdrawal dwindled during the course of illness in Rasmussen encephalitis. Seizures cause high Na(+) influx through action potentials. A transient decrease in [Ca(2+)]e (seizure condition) increases GABA transporter (GAT)-mediated GABA release because of elevated [Na(+)]i. This amplifies the Sodium-Calcium-Exchanger (NCX) activity, further increasing [Na(+)]i and GABA release. The reduction in [Ca(2+)]e triggers a GAT-NCX related anti-seizure mechanism by augmenting GAT-mediated GABA release. This mechanism, obvious in humans, is absent in rats.


Subject(s)
Calcium/metabolism , Neocortex/metabolism , Neocortex/pathology , Seizures/pathology , Sodium/metabolism , Synaptosomes/metabolism , Adolescent , Adult , Aged , Aniline Compounds/pharmacology , Animals , Child , Child, Preschool , Enzyme Inhibitors/pharmacology , Female , GABA Plasma Membrane Transport Proteins/metabolism , Humans , Infant , Male , Middle Aged , Neurotoxins/pharmacology , Ouabain/pharmacology , Phenyl Ethers/pharmacology , Rats , Rats, Wistar , Synaptosomes/drug effects , Tetanus Toxin/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Tritium/metabolism , Veratridine/pharmacology , Young Adult
7.
Cell Physiol Biochem ; 38(3): 1182-98, 2016.
Article in English | MEDLINE | ID: mdl-26983082

ABSTRACT

BACKGROUND/AIMS: Common systems for the quantification of cellular contraction rely on animal-based models, complex experimental setups or indirect approaches. The herein presented CellDrum technology for testing mechanical tension of cellular monolayers and thin tissue constructs has the potential to scale-up mechanical testing towards medium-throughput analyses. Using hiPS-Cardiac Myocytes (hiPS-CMs) it represents a new perspective of drug testing and brings us closer to personalized drug medication. METHODS: In the present study, monolayers of self-beating hiPS-CMs were grown on ultra-thin circular silicone membranes and deflect under the weight of the culture medium. Rhythmic contractions of the hiPS-CMs induced variations of the membrane deflection. The recorded contraction-relaxation-cycles were analyzed with respect to their amplitudes, durations, time integrals and frequencies. Besides unstimulated force and tensile stress, we investigated the effects of agonists and antagonists acting on Ca2+ channels (S-Bay K8644/verapamil) and Na+ channels (veratridine/lidocaine). RESULTS: The measured data and simulations for pharmacologically unstimulated contraction resembled findings in native human heart tissue, while the pharmacological dose-response curves were highly accurate and consistent with reference data. CONCLUSION: We conclude that the combination of the CellDrum with hiPS-CMs offers a fast, facile and precise system for pharmacological, toxicological studies and offers new preclinical basic research potential.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Ion Channels/agonists , Ion Channels/antagonists & inhibitors , Myocytes, Cardiac/cytology , Stress, Mechanical , Cell Culture Techniques/methods , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/drug effects , Lidocaine/pharmacology , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Verapamil/pharmacology , Veratridine/pharmacology
8.
Acta Pharmacol Sin ; 37(11): 1432-1441, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27569391

ABSTRACT

AIM: The augmentation of late sodium current (INa.L) not only causes intracellular Na+ accumulation, which results in intracellular Ca2+ overload via the reverse mode of the Na+/Ca2+ exchange current (reverse-INCX), but also prolongs APD and induces early afterdepolarizations (EAD), which can lead to arrhythmia and cardiac dysfunction. Thus, the inhibition of INa.L is considered to be a potential way for therapeutic intervention in ischemia and heart failure. In this study we investigated the effects of tolterodine (Tol), a competitive muscarinic receptor antagonist, on normal and veratridine (Ver)-augmented INa.L, reverse-INCX and APD in isolated rabbit ventricular myocytes, which might contribute to its cardioprotective activity. METHODS: Rabbit ventricular myocytes were prepared. The INa.L and reverse-INCX were recorded in voltage clamp mode, whereas action potentials and Ver-induced early afterdepolarizations (EADs) were recorded in current clamp mode. Drugs were applied via superfusion. RESULTS: Tol (3-120 nmol/L) concentration-dependently inhibited the normal and Ver-augmented INa.L with IC50 values of 32.08 nmol/L and 42.47 nmol/L, respectively. Atropine (100 µmol/L) did not affect the inhibitory effects of Tol (30 nmol/L) on Ver-augmented INa.L. In contrast, much high concentrations of Tol was needed to inhibit the transient sodium current (INa.T) with an IC50 value of 183.03 µmol/L. In addition, Tol (30 nmol/L) significantly shifted the inactivation curve of INa.T toward a more depolarizing membrane potential without affecting its activation characteristics. Moreover, Tol (30 nmol/L) significantly decreased Ver-augmented reverse-INCX. Tol (30 nmol/L) increased the action potential duration (APD) by 16% under the basal conditions. Ver (20 µmol/L) considerably extended the APD and evoked EADs in 18/24 cells (75%). In the presence of Ver, Tol (30 nmol/L) markedly decreased the APD and eliminated EADs (0/24 cells). CONCLUSION: Tol inhibits normal and Ver-augmented INaL and decreases Ver-augmented reverse-INCX. In addition, Tol reverses the prolongation of the APD and eliminates the EADs induced by Ver, thus prevents Ver-induced arrhythmia.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Muscarinic Antagonists/pharmacology , Myocytes, Cardiac/drug effects , Sodium Channel Blockers/pharmacology , Sodium Channels/physiology , Sodium-Calcium Exchanger/metabolism , Tolterodine Tartrate/pharmacology , Veratridine/pharmacology , Action Potentials , Animals , Female , Heart Ventricles/cytology , In Vitro Techniques , Male , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Rabbits
9.
Pflugers Arch ; 467(7): 1431-1443, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25081243

ABSTRACT

Although beat-to-beat variability (short-term variability, SV) of action potential duration (APD) is considered as a predictor of imminent cardiac arrhythmias, the underlying mechanisms are still not clear. In the present study, therefore, we aimed to determine the role of the major cardiac ion currents, APD, stimulation frequency, and changes in the intracellular Ca(2+) concentration ([Ca(2+)]i) on the magnitude of SV. Action potentials were recorded from isolated canine ventricular cardiomyocytes using conventional microelectrode techniques. SV was an exponential function of APD, when APD was modified by current injections. Drug effects were characterized as relative SV changes by comparing the drug-induced changes in SV to those in APD according to the exponential function obtained with current pulses. Relative SV was increased by dofetilide, HMR 1556, nisoldipine, and veratridine, while it was reduced by BAY K8644, tetrodotoxin, lidocaine, and isoproterenol. Relative SV was also increased by increasing the stimulation frequency and [Ca(2+)]i. In summary, relative SV is decreased by ion currents involved in the negative feedback regulation of APD (I Ca, I Ks, and I Kr), while it is increased by I Na and I to. We conclude that drug-induced effects on SV should be evaluated in relation with the concomitant changes in APD. Since relative SV was decreased by ion currents playing critical role in the negative feedback regulation of APD, blockade of these currents, or the beta-adrenergic pathway, may carry also some additional proarrhythmic risk in addition to their well-known antiarrhythmic action.


Subject(s)
Action Potentials , Heart Ventricles/cytology , Ion Channels/metabolism , Myocytes, Cardiac/physiology , Potassium Channel Blockers/pharmacology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Calcium/metabolism , Cardiotonic Agents/pharmacology , Cells, Cultured , Chromans/pharmacology , Dogs , Feedback, Physiological , Female , Ion Channels/antagonists & inhibitors , Ion Transport , Isoproterenol/pharmacology , Lidocaine/pharmacology , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nisoldipine/pharmacology , Phenethylamines/pharmacology , Sulfonamides/pharmacology , Tetrodotoxin/pharmacology , Veratridine/pharmacology
10.
Arch Insect Biochem Physiol ; 90(3): 131-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26183110

ABSTRACT

A neuronal morphological phenotype can be induced in cultured Spodoptera frugiperda insect cells (Sf21) by supplementing serum-containing media with 20-hydroxyecdysone (20-HE) and/or insulin. In this study, the primary objectives were to determine any role of ion channels in mediating the morphological change in cells treated with 20-HE and insulin, and whether serum was required to observe this effect. Results showed serum-free media also induced growth of processes in Sf21 cells, but at a lower percentage than that found previously in cells bathed in serum-containing media. Veratridine, a sodium channel activator, increased cell survival when applied in combination with 20-HE to Sf21 cells, and the effect was blocked by tetrodotoxin (1 µM) a known sodium channel blocker. Cobalt, a calcium channel blocker, showed significant inhibition of cell process growth when applied in combination with both 20-HE and 20-HE plus veratridine. Cobalt also showed significant inhibition of cell process growth when applied in combination with insulin. Thus, some type of sodium channel, as well as a mechanism for transmembrane calcium ion movement, are apparently expressed in Sf21 cells and are involved in the differentiation process. These cell lines may be used in a wide variety of endeavors, including the screening of insecticides, as well as foster basic studies of neurodevelopment and ecdysone action.


Subject(s)
Ecdysterone/pharmacology , Ion Channels/antagonists & inhibitors , Neurons/drug effects , Animals , Calcium Channel Blockers/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cobalt/pharmacology , Culture Media, Serum-Free , Insulin/pharmacology , Neurons/cytology , Serum , Sf9 Cells , Sodium Channel Blockers/pharmacology , Spodoptera , Tetrodotoxin/pharmacology , Veratridine/pharmacology
11.
Can J Physiol Pharmacol ; 93(7): 527-34, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25928391

ABSTRACT

This study was designed to investigate the influence of cytosolic Ca(2+) levels ([Ca(2+)]i) on action potential duration (APD) and on the incidence of early afterdepolarizations (EADs) in canine ventricular cardiomyocytes. Action potentials (AP) of isolated cells were recorded using conventional sharp microelectrodes, and the concomitant [Ca(2+)]i was monitored with the fluorescent dye Fura-2. EADs were evoked at a 0.2 Hz pacing rate by inhibiting the rapid delayed rectifier K(+) current with dofetilide, by activating the late sodium current with veratridine, or by activating the L-type calcium current with BAY K8644. These interventions progressively prolonged the AP and resulted in initiation of EADs. Reducing [Ca(2+)]i by application of the cell-permeant Ca(2+) chelator BAPTA-AM lengthened the AP at 1.0 Hz if it was applied alone, in the presence of veratridine, or in the presence of BAY K8644. However, BAPTA-AM shortened the AP if the cells were pretreated with dofetilide. The incidence of the evoked EADs was strongly reduced by BAPTA-AM in dofetilide, moderately reduced in veratridine, whereas EAD incidence was increased by BAPTA-AM in the presence of BAY K8644. Based on these experimental data, changes in [Ca(2+)]i have marked effects on APD as well as on the incidence of EADs; however, the underlying mechanisms may be different, depending on the mechanism of EAD generation. As a consequence, reduction of [Ca(2+)]i may eliminate EADs under some, but not all, experimental conditions.


Subject(s)
Action Potentials/physiology , Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Cytosol/metabolism , Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/physiopathology , Calcium Channel Agonists/pharmacology , Calcium Chelating Agents/pharmacology , Cells, Cultured , Cytosol/drug effects , Dogs , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Female , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Male , Myocytes, Cardiac/drug effects , Phenethylamines/pharmacology , Sulfonamides/pharmacology , Time Factors , Veratridine/pharmacology
12.
Bioelectromagnetics ; 36(5): 386-97, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25884360

ABSTRACT

This paper investigates the influence of pulsed magnetic fields (PMFs) on amplitude of evoked, compound action potential (CAP) recorded from the segments of sciatic nerve in vitro. PMFs were applied for 30 min at frequency of 0.16 Hz and intensity of 15 mT. In confirmation of our previous reports, PMF exposure enhanced amplitude of CAPs. The effect persisted beyond PMF activation period. As expected, CAP amplitude was attenuated by antagonists of sodium channel, lidocaine, and tetrodotoxin. Depression of the potential by sodium channels antagonists was reversed by subsequent exposure to PMFs. The effect of elevated potassium concentration and veratridine on the action potential was modified by exposure to PMFs as well. Neither inhibitors of protein kinase C and protein kinase A, nor known free radicals scavengers had any effects on PMF action. Possible mechanisms of PMF action are discussed.


Subject(s)
Action Potentials/physiology , Magnetic Fields , Sodium Channels/metabolism , Action Potentials/drug effects , Animals , Ascorbic Acid/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Free Radical Scavengers/pharmacology , Lidocaine/pharmacology , Male , Mice , Microelectrodes , Potassium/metabolism , Protein Kinase C/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/physiology , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Veratridine/pharmacology , Vitamin E/metabolism
13.
J Neurosci ; 33(35): 13957-71, 13971a, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23986233

ABSTRACT

Neuronal respiration is controlled by ATP demand and Ca2+ but the roles played by each are unknown, as any Ca2+ signal also impacts on ATP demand. Ca2+ can control mitochondrial function through Ca2+-regulated mitochondrial carriers, the aspartate-glutamate and ATP-Mg/Pi carriers, ARALAR/AGC1 and SCaMC-3, respectively, or in the matrix after Ca2+ transport through the Ca2+ uniporter. We have studied the role of Ca2+ signaling in the regulation of mitochondrial respiration in intact mouse cortical neurons in basal conditions and in response to increased workload caused by increases in [Na+]cyt (veratridine, high-K+ depolarization) and/or [Ca2+]cyt (carbachol). Respiration in nonstimulated neurons on 2.5-5 mm glucose depends on ARALAR-malate aspartate shuttle (MAS), with a 46% drop in aralar KO neurons. All stimulation conditions induced increased OCR (oxygen consumption rate) in the presence of Ca2+, which was prevented by BAPTA-AM loading (to preserve the workload), or in Ca2+-free medium (which also lowers cell workload). SCaMC-3 limits respiration only in response to high workloads and robust Ca2+ signals. In every condition tested Ca2+ activation of ARALAR-MAS was required to fully stimulate coupled respiration by promoting pyruvate entry into mitochondria. In aralar KO neurons, respiration was stimulated by veratridine, but not by KCl or carbachol, indicating that the Ca2+ uniporter pathway played a role in the first, but not in the second condition, even though KCl caused an increase in [Ca2+]mit. The results suggest a requirement for ARALAR-MAS in priming pyruvate entry in mitochondria as a step needed to activate respiration by Ca2+ in response to moderate workloads.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium/metabolism , Cell Respiration/genetics , Homeostasis , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Neurons/metabolism , Animals , Calcium Signaling , Carbachol/pharmacology , Cell Respiration/drug effects , Cerebral Cortex/cytology , Glucose/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Oxygen/metabolism , Potassium Chloride/pharmacology , Pyruvates/metabolism , Sodium/metabolism , Veratridine/pharmacology
14.
J Neurosci ; 33(20): 8716-28, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23678115

ABSTRACT

How rhythms are generated by neuronal networks is fundamental to understand rhythmic behaviors such as respiration, locomotion, and mastication. Respiratory rhythm is generated by the preBötzinger complex (preBötC), an anatomically and functionally discrete population of brainstem neurons, central and necessary for respiratory rhythm. In specific in vitro conditions, preBötC neurons depend on voltage-dependent inward currents to generate respiratory rhythm. In the mature and intact organism, where preBötC neurons are deeply embedded in the respiratory network, the contribution of ionic currents to respiratory rhythm is unclear. We propose that a set of ionic currents plays a key role in generating respiratory rhythm in the mature organism in vivo. By microperfusing ionic current blockers into the preBötC of adult rats, we identify the hyperpolarization-activated cation current as a critical component of the mechanism promoting respiratory rhythm, and that this current, in combination with the persistent sodium current, is essential to respiratory rhythm in vivo. Importantly, both currents contribute to rhythmic activity in states of anesthesia, quiet wakefulness, and sleep, but not when the organism is engaged in active behaviors. These data show that a set of ionic currents at the preBötC imparts the network with rhythmicity in reduced states of arousal, although the network can override their contribution to adjust its activity for nonrhythmic behaviors in active wakefulness.


Subject(s)
Periodicity , Respiratory Center/physiology , Respiratory Mechanics/physiology , Sodium Channels/physiology , Sodium-Potassium-Chloride Symporters/physiology , Analysis of Variance , Animals , Cardiovascular Agents/pharmacology , Electroencephalography , Excitatory Amino Acid Antagonists/pharmacology , Functional Laterality/drug effects , Functional Laterality/physiology , In Vitro Techniques , Male , Membrane Potentials/drug effects , Microdialysis , Motor Activity/drug effects , Muscles/drug effects , Muscles/physiology , Neurons , Patch-Clamp Techniques , Pyrimidines/pharmacology , Rats , Rats, Wistar , Receptors, Neurokinin-1/metabolism , Respiratory Center/drug effects , Riluzole/pharmacology , Sleep , Veratridine/pharmacology , Wakefulness
15.
Biochim Biophys Acta ; 1830(3): 2591-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23201194

ABSTRACT

BACKGROUND: Herein we report the discovery of a cystine-crosslinked peptide from Porifera along with high-quality spatial details accompanied by the description of its unique effect on neuronal calcium influx. METHODS: Asteropsin A (ASPA) was isolated from the marine sponge Asteropus sp., and its structure was independently determined using X-ray crystallography (0.87 angstroms) and solution NMR spectroscopy. RESULTS: An N-terminal pyroglutamate modification, uncommon cis proline conformations, and absence of basic residues helped distinguish ASPA from other cystine-crosslinked knot peptides. ASPA enhanced Ca2+ influx in murine cerebrocortical neuron cells following the addition of the Na+ channel activator veratridine but did not modify the oscillation frequency or amplitude of neuronal Ca2+ currents alone. Allosterism at neurotoxin site 2 was not observed, suggesting an alternative to the known Na+ channel interaction. CONCLUSIONS: Together with a distinct biological activity, the origin of ASPA suggests a new subclass of cystine-rich knot peptides associated with Porifera. GENERAL SIGNIFICANCE: The discovery of ASPA represents a distinctive addition to an emerging subclass of cystine-crosslinked knot peptides from Porifera.


Subject(s)
Calcium/metabolism , Cystine/chemistry , Neurons/drug effects , Peptides/chemistry , Porifera/chemistry , Action Potentials/drug effects , Amino Acid Sequence , Animals , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Crystallography, X-Ray , Ion Transport/drug effects , Kinetics , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Sequence Data , Neurons/cytology , Neurons/metabolism , Peptides/isolation & purification , Peptides/pharmacology , Primary Cell Culture , Protein Conformation , Protein Folding , Sodium Channels/metabolism , Veratridine/pharmacology
16.
J Neural Transm (Vienna) ; 121(11): 1321-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25008583

ABSTRACT

Glutamate is thought to be the most important excitatory neurotransmitter in the CNS, while glutamine predominantly serves as a precursor and metabolite in the glutamate-glutamine cycle. To verify the interaction between intrinsic extracellular glutamate, y-aminobutyric acid (GABA) levels and glial glutamine outflow in human tissue, fresh brain slices from human frontal cortex were incubated in superfusion chambers in vitro. Human neocortical tissue was obtained during surgical treatment of subcortical brain tumors. For superfusion experiments, the white matter was separated and discarded from the gray matter, which finally contained all six neocortical layers. Outflows of endogenous glutamate, GABA and glutamine were established after a 40-min washout period and amounts were simultaneously quantified after two-phase derivatization by high-performance liquid chromatography with electrochemical detection. Under basal conditions, amounts of glutamate could be found 20-fold in comparison to the inhibitory neurotransmitter GABA, whereas this excitatory predominance markedly declined after veratridine-induced activation. The basal glutamate:glutamine ratio of extracellular levels was approximately 1:2. Blockade or activation of the voltage-gated sodium channel by tetrodotoxin or veratridine significantly modulated glutamate levels, but the glutamate:glutamine ratio was nearly constant with 1:2. When the EAAT blocker TBOA was employed, glutamine remained nearly unchanged whereas glutamate significantly enhanced. These results led us to suggest that glutamine release through glial SN1 is related to EAAT activity that can be modulated by intrinsic extracellular glutamate in human cortical slices.


Subject(s)
Cerebral Cortex/cytology , Extracellular Fluid/metabolism , Glutamic Acid/metabolism , Neurons/physiology , Adult , Aged , Aspartic Acid/pharmacology , Chromatography, High Pressure Liquid , Extracellular Fluid/drug effects , Female , Glutamine/metabolism , Humans , In Vitro Techniques , Male , Methionine/analogs & derivatives , Methionine/pharmacology , Middle Aged , Neurons/drug effects , Tetrodotoxin/pharmacology , Time Factors , Veratridine/pharmacology , gamma-Aminobutyric Acid/metabolism
17.
J Nat Prod ; 77(11): 2553-60, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25343669

ABSTRACT

A small library of synthetic (-)-palmyrolide A diastereomers, analogues, and acyclic precursors have been examined with respect to their interaction with voltage-gated sodium channels (VGSCs). Toward this goal, the ability of (-)-palmyrolide A and analogues to antagonize veratridine-stimulated Na(+) influx in primary cultures of mouse cerebrocortical neurons was assessed. We found that synthetic (-)-palmyrolide A and its enantiomer functioned as VGSC antagonists to block veratridine-induced sodium influx. A detailed NMR and computational analysis of four diastereomers revealed that none had the same combination of shape and electrostatic potential as exhibited by natural (-)-palmyrolide A. These data indicate that the relative configuration about the tert-butyl and methyl substituents appears to be a prerequisite for biological function. Additional testing revealed that the enamide double bond was not necessary for blocking veratridine-induced sodium influx, whereas the acyclic analogues and other macrolide diastereomers tested were inactive as inhibitors of VGSCs, suggesting that the intact macrolide was required.


Subject(s)
Macrolides/chemistry , Macrolides/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Animals , Mice , Molecular Structure , Neurons/drug effects , Stereoisomerism , Veratridine/pharmacology , Voltage-Gated Sodium Channel Blockers/chemistry
18.
Mar Drugs ; 12(9): 4868-82, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25251033

ABSTRACT

Brevetoxins are a family of ladder-framed polyether toxins produced during blooms of the marine dinoflagellate, Karenia brevis. Consumption of shellfish or finfish exposed to brevetoxins can lead to the development of neurotoxic shellfish poisoning. The toxic effects of brevetoxins are believed to be due to the activation of voltage-sensitive sodium channels in cell membranes. The traditional cytotoxicity assay for detection of brevetoxins uses the Neuro-2A cell line, which must first be treated with the neurotoxins, ouabain and veratridine, in order to become sensitive to brevetoxins. In this study, we demonstrate several drawbacks of the Neuro-2A assay, which include variability for the EC50 values for brevetoxin and non-linear triphasic dose response curves. Ouabain/ veratridine-treated Neuro-2A cells do not show a typical sigmoidal dose response curve in response to brevetoxin, but rather, have a polynomial shaped curve, which makes calculating EC50 values highly variable. We describe a new fluorescence live cell imaging model, which allows for accurate calculation of cytotoxicity via nuclear staining and additional measurement of other viability parameters depending on which aspect of the cell is stained. In addition, the SJCRH30 cell line shows promise as an alternative to Neuro-2A cells for testing brevetoxins without the need for ouabain and veratridine.


Subject(s)
Antineoplastic Agents/toxicity , Dinoflagellida/chemistry , Dinoflagellida/ultrastructure , Marine Toxins/toxicity , Oxocins/toxicity , Animals , Apoptosis/drug effects , Cardiotonic Agents/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Microscopy, Fluorescence , Ouabain/pharmacology , Rats , Veratridine/pharmacology
19.
Article in English | MEDLINE | ID: mdl-24964655

ABSTRACT

Nonstructural protein 1 (NS1) of the highly pathogenic avian influenza virus (H5N1) contains a conserved RNA binding domain (RBD) that inhibits antiviral functions of host-innate immune response. Dimerization of NS1 forms a central groove and binds to double stranded (ds) RNA. This region might serve as a potential drug target. In this study, three dimensional structure model of NS1 RBD protein was constructed and virtual screening was performed to identify lead compounds that bound within and around the central groove. The virtual screening showed that 5 compounds bound within the central groove with binding energy ranging between -16.05 and -17.36 Kcal/mol. Two commercially available compounds, estradiol and veratridine, were selected for using in an in vitro screening assay. The results showed that neither of the compounds could inhibit the association between dsRNA and NS1 RBD protein. In addition, 34 herbal extracts were examined for their inhibitory effects. Five of them were able to inhibit association between NS1 RBD and dsRNA in electrophoresis mobility shift assay. Four herbs, Terminalia belirica, Salacia chinensis, Zingiber montanum and Peltophorum pterocarpum, could reduce > 50% of infectivity of H5N1 in a cell-based assay, and it is worth further studying their potential use as source of antiviral drugs.


Subject(s)
Antiviral Agents/pharmacology , Estradiol/pharmacology , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/metabolism , Lead/pharmacology , Plant Extracts/pharmacology , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , Veratridine/pharmacology , Viral Nonstructural Proteins/metabolism , Polymerase Chain Reaction
20.
J Neurosci ; 32(8): 2714-21, 2012 Feb 22.
Article in English | MEDLINE | ID: mdl-22357855

ABSTRACT

We report a novel coupled system of sodium-activated potassium currents (I(KNa)) and persistent sodium currents (I(NaP)), the components of which are widely distributed throughout the brain. Its existence and importance has not been previously recognized. Although I(KNa) was known to exist in many cell types, the source of Na(+) which activates I(KNa) remained a mystery. We now show in single membrane patches generated from the somas of rat neurons that sodium influx through I(NaP) is sufficient for activation of K(Na) channels, without substantial contribution from the transient sodium current or bulk [Na(+)](i). I(NaP) was found to be active at cell membrane resting potentials, a finding that may explain why I(KNa) can be evoked from negative holding potentials. These results show an unanticipated role for I(NaP) in activating a negative feedback system countering the excitable effects I(NaP); the interrelatedness of I(NaP) and I(KNa) suggests new ways neurons can tune their excitability.


Subject(s)
Ion Channel Gating/physiology , Membrane Potentials/physiology , Neurons/physiology , Potassium Channels/physiology , Sodium Channels/metabolism , Sodium/metabolism , Aminopyridines/pharmacology , Animals , Animals, Newborn , Biophysics , Cells, Cultured , Cesium/pharmacology , Chlorides/pharmacology , Electric Stimulation , Female , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Ions/metabolism , Male , Membrane Potentials/drug effects , Neurons/drug effects , Olfactory Bulb/cytology , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Rats , Sodium/pharmacology , Sodium Channel Blockers/pharmacology , Tetraethylammonium/pharmacology , Tetrodotoxin/pharmacology , Veratridine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL