Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 263
1.
FEBS Lett ; 598(8): 889-901, 2024 Apr.
Article En | MEDLINE | ID: mdl-38563123

BeKm-1 is a peptide toxin from scorpion venom that blocks the pore of the potassium channel hERG (Kv11.1) in the human heart. Although individual protein structures have been resolved, the structure of the complex between hERG and BeKm-1 is unknown. Here, we used molecular dynamics and ensemble docking, guided by previous double-mutant cycle analysis data, to obtain an in silico model of the hERG-BeKm-1 complex. Adding to the previous mutagenesis study of BeKm-1, our model uncovers the key role of residue Arg20, which forms three interactions (a salt bridge and hydrogen bonds) with the channel vestibule simultaneously. Replacement of this residue even by lysine weakens the interactions significantly. In accordance, the recombinantly produced BeKm-1R20K mutant exhibited dramatically decreased activity on hERG. Our model may be useful for future drug design attempts.


Arginine , ERG1 Potassium Channel , Molecular Dynamics Simulation , Scorpion Venoms , Animals , Humans , Arginine/chemistry , Arginine/metabolism , ERG1 Potassium Channel/chemistry , ERG1 Potassium Channel/metabolism , HEK293 Cells , Molecular Docking Simulation , Mutation , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Scorpion Venoms/chemistry , Scorpion Venoms/genetics , Scorpion Venoms/metabolism
2.
J Phys Chem B ; 128(2): 429-439, 2024 Jan 18.
Article En | MEDLINE | ID: mdl-38179652

The KCNA5 gene provides the code for the α-subunit of the potassium channel Kv1.5. The genetic variant H463R in the Kv1.5 channel has been reported to cause a functional loss in atrial fibrillation (AF) patients. Understanding the mutations at a molecular level is key to developing improved therapeutics concerning cardiac hKv1.5 and hKv1.4 channels. Molecular dynamics and umbrella sampling free energy simulations are an effective tool to understand the mutation's effect on ion conduction, which we have employed and found that the hKv1.5[H463R] mutation imposes an energy barrier on the ion conduction pathway compared to the wild-type channel's ion free energy and pore structure. These results imply that the arginine mutation associated with the AF disease in particular modulates the inactivation process of hKv1.5. Kv1.4, encoded by the KCNA4 gene, is also present in the heart. Therefore, we considered simulation studies of the equivalent H507R mutation in the hKv1.4 channel and found that the mutation slightly reduces the ion conduction barrier in the ion conduction pathway, making it insignificant.


Molecular Dynamics Simulation , Potassium Channels , Humans , Mutation , Potassium Channels/metabolism , Heart , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism
3.
Prostaglandins Other Lipid Mediat ; 169: 106782, 2023 12.
Article En | MEDLINE | ID: mdl-37741358

OBJECTIVE: This study aimed to investigate vasoactive effect mechanisms of cilostazol in rat thoracic aorta. MATERIALS AND METHODS: The vessel rings prepared from the thoracic aortas of the male rats were placed in the chambers of the isolated tissue bath system. The resting tone was adjusted to 1 g. Following the equilibration phase, potassium chloride or phenylephrine was used to contract the vessel rings. When achieving a steady contraction, cilostazol was applied cumulatively (10-8-10-4 M). In the presence of potassium channel blockers or signaling pathway inhibitors, the same experimental procedure was performed. RESULTS: Cilostazol exhibited a significant vasorelaxant effect in a concentration-dependent manner (pD2: 5.94 ± 0.94) (p < .001). The vasorelaxant effect level of cilostazol was significantly reduced by the endothelial nitric oxide synthase inhibitor L-NAME (10-4 M), soluble guanylate cyclase inhibitor methylene blue (10 µM), cyclooxygenase 1/2 inhibitor indomethacin (5 µM), adenosine monophosphate-activated protein kinase inhibitor compound C (10 µM), non-selective potassium channel blocker tetraethylammonium chloride (10 mM), large-conductance calcium-activated potassium channel blocker iberiotoxin (20 nM), voltage-gated potassium channel blocker 4-Aminopyridine (1 mM), and inward-rectifier potassium channel blocker BaCl2 (30 µM) (p < .001). Moreover, incubation of cilostazol (10-4 M) significantly reduced caffeine (10 mM), cyclopiazonic acid (10 µM), and phorbol 12-myristate 13-acetate-induced (100 µM) vascular contractions (p < .001). CONCLUSIONS: In the rat thoracic aorta, the vasodilator action level of cilostazol is quite noticeable. The vasorelaxant effects of cilostazol are mediated by the eNOS/NO/cGMP pathway, prostanoids, AMPK pathway, PKC, potassium channels, and calcium channels.


Calcium Channels , Vasodilation , Rats , Male , Animals , Cilostazol/pharmacology , Cilostazol/metabolism , Calcium Channels/metabolism , Calcium Channels/pharmacology , Potassium Channels/metabolism , Potassium Channels/pharmacology , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/metabolism , Nitric Oxide Synthase Type III/metabolism , Prostaglandins/metabolism , Vasodilator Agents/pharmacology , Endothelium, Vascular , Calcium/metabolism , Calcium/pharmacology
4.
Int J Mol Sci ; 23(3)2022 Feb 02.
Article En | MEDLINE | ID: mdl-35163644

Peptide pore blockers and their fluorescent derivatives are useful molecular probes to study the structure and functions of the voltage-gated potassium Kv1.3 channel, which is considered as a pharmacological target in the treatment of autoimmune and neurological disorders. We present Kv1.3 fluorescent ligand, GFP-MgTx, constructed on the basis of green fluorescent protein (GFP) and margatoxin (MgTx), the peptide, which is widely used in physiological studies of Kv1.3. Expression of the fluorescent ligand in E. coli cells resulted in correctly folded and functionally active GFP-MgTx with a yield of 30 mg per 1 L of culture. Complex of GFP-MgTx with the Kv1.3 binding site is reported to have the dissociation constant of 11 ± 2 nM. GFP-MgTx as a component of an analytical system based on the hybrid KcsA-Kv1.3 channel is shown to be applicable to recognize Kv1.3 pore blockers of peptide origin and to evaluate their affinities to Kv1.3. GFP-MgTx can be used in screening and pre-selection of Kv1.3 channel blockers as potential drug candidates.


Green Fluorescent Proteins/metabolism , Kv1.3 Potassium Channel , Peptides/metabolism , Potassium Channel Blockers/metabolism , Binding Sites , Humans , Kv1.3 Potassium Channel/analysis , Kv1.3 Potassium Channel/metabolism , Ligands , Protein Binding
5.
Biochem Genet ; 60(2): 504-526, 2022 Apr.
Article En | MEDLINE | ID: mdl-34286408

Different toxins acting on Kv1.3 channel have been isolated from animal venom. MeuKTX toxin from Mesobuthus eupeus phillipsi scorpion and shtx-k toxin from Stichodactyla haddoni sea anemone have been identified as two effective Kv1.3 channel blockers. In this work, we characterized the genomic organization of both toxins. MeuKTX gene contains one intron and two exons, similar to the most scorpion toxins. There are a few reports of genomic structure of sea anemone toxins acting on Kv channels. The sequence encoding mature peptide of shtx-k was located in an exon separated by an intron from the coding exon of the propeptide and signal region. In order to make a peptide with more affinity for Kv1.3 channel and greater stability, the shtx-k/ MeuKTX chimeric peptide was designed and constructed using splicing by overlap extension-PCR (SOE-PCR) method. MeuKTX, shtx-k, and shtx-k/MeuKTX were cloned and the expression of the soluble proteins in E. coli was determined. Molecular docking studies indicated more inhibitory effect of shtx-k/MeuKTX on Kv1.3 channel compared to shtx-k and MeuKTX toxins. Key amino acids binding channel from both toxins, also involved in interaction of chimeric peptide with channel. Our results showed that the fusion peptide, shtx-k/MeuKTX could be an effective agent to target Kv1.3 channel.


Scorpion Venoms , Sea Anemones , Amino Acid Sequence , Animals , Escherichia coli , Genomics , Molecular Docking Simulation , Peptides/chemistry , Peptides/genetics , Peptides/pharmacology , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Scorpion Venoms/chemistry , Scorpion Venoms/genetics , Scorpions/chemistry , Scorpions/genetics , Scorpions/metabolism , Sea Anemones/chemistry , Sea Anemones/genetics , Sea Anemones/metabolism
6.
Toxicology ; 464: 153018, 2021 12.
Article En | MEDLINE | ID: mdl-34757159

The human ether-à-go-go-related gene (hERG) encodes the Kv11.1 voltage-gated potassium ion (K+) channel that conducts the rapidly activating delayed rectifier current (IKr) in cardiomyocytes to regulate the repolarization process. Some drugs, as blockers of hERG potassium channels, cannot be marketed due to prolonged QT intervals, as well known as cardiotoxicity. Predetermining the binding affinity values between drugs and hERG through in silico methods can greatly reduce the time and cost required for experimental verification. In this study, we collected 9,215 compounds with AutoDock Vina's docking structures as training set, and collected compounds from four references as test sets. A series of models for predicting the binding affinities of hERG blockers were built based on five machine learning algorithms and combinations of interaction features and ligand features. The model built by support vector regression (SVR) using the combination of all features achieved the best performance on both tenfold cross-validation and external verification, which was selected and named as TSSF-hERG (target-specific scoring function for hERG). TSSF-hERG is more accurate than the classic scoring function of AutoDock Vina and the machine-learning-based generic scoring function RF-Score, with a Pearson's correlation coefficient (Rp) of 0.765, a Spearman's rank correlation coefficient (Rs) of 0.757, a root-mean-square error (RMSE) of 0.585 in a tenfold cross-validation study. All results demonstrated that TSSF-hERG would be useful for improving the power of binding affinity prediction between hERG and compounds, which can be further used for prediction or virtual screening of the hERG-related cardiotoxicity of drug candidates.


Cardiotoxicity/etiology , ERG1 Potassium Channel/antagonists & inhibitors , Machine Learning , Potassium Channel Blockers/toxicity , Algorithms , Cardiotoxicity/physiopathology , ERG1 Potassium Channel/metabolism , Humans , Molecular Docking Simulation , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Protein Binding
7.
J Mol Cell Cardiol ; 158: 163-177, 2021 09.
Article En | MEDLINE | ID: mdl-34062207

Drug isomers may differ in their proarrhythmia risk. An interesting example is the drug sotalol, an antiarrhythmic drug comprising d- and l- enantiomers that both block the hERG cardiac potassium channel and confer differing degrees of proarrhythmic risk. We developed a multi-scale in silico pipeline focusing on hERG channel - drug interactions and used it to probe and predict the mechanisms of pro-arrhythmia risks of the two enantiomers of sotalol. Molecular dynamics (MD) simulations predicted comparable hERG channel binding affinities for d- and l-sotalol, which were validated with electrophysiology experiments. MD derived thermodynamic and kinetic parameters were used to build multi-scale functional computational models of cardiac electrophysiology at the cell and tissue scales. Functional models were used to predict inactivated state binding affinities to recapitulate electrocardiogram (ECG) QT interval prolongation observed in clinical data. Our study demonstrates how modeling and simulation can be applied to predict drug effects from the atom to the rhythm for dl-sotalol and also increased proarrhythmia proclivity of d- vs. l-sotalol when accounting for stereospecific beta-adrenergic receptor blocking.


Adrenergic beta-Antagonists/chemistry , Adrenergic beta-Antagonists/metabolism , Anti-Arrhythmia Agents/chemistry , Anti-Arrhythmia Agents/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Long QT Syndrome/metabolism , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Signal Transduction/drug effects , Sotalol/chemistry , Sotalol/metabolism , Adrenergic beta-Antagonists/pharmacology , Anti-Arrhythmia Agents/pharmacology , Cryoelectron Microscopy/methods , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/chemistry , HEK293 Cells , Humans , Molecular Dynamics Simulation , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Potassium Channel Blockers/pharmacology , Protein Binding/drug effects , Sotalol/pharmacology , Stereoisomerism
8.
J Med Chem ; 64(11): 7691-7701, 2021 06 10.
Article En | MEDLINE | ID: mdl-34038119

A renal outer medullary potassium channel (ROMK, Kir1.1) is a putative drug target for a novel class of diuretics with potential for treating hypertension and heart failure. Our first disclosed clinical ROMK compound, 2 (MK-7145), demonstrated robust diuresis, natriuresis, and blood pressure lowering in preclinical models, with reduced urinary potassium excretion compared to the standard of care diuretics. However, 2 projected to a short human half-life (∼5 h) that could necessitate more frequent than once a day dosing. In addition, a short half-life would confer a high peak-to-trough ratio which could evoke an excessive peak diuretic effect, a common liability associated with loop diuretics such as furosemide. This report describes the discovery of a new ROMK inhibitor 22e (MK-8153), with a longer projected human half-life (∼14 h), which should lead to a reduced peak-to-trough ratio, potentially extrapolating to more extended and better tolerated diuretic effects.


Natriuretic Agents/chemistry , Potassium Channel Blockers/chemistry , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Action Potentials/drug effects , Animals , Benzofurans/chemistry , Blood Pressure/drug effects , Diuretics/chemistry , Diuretics/metabolism , Diuretics/pharmacology , Dogs , Half-Life , Haplorhini , Humans , Male , Natriuretic Agents/metabolism , Natriuretic Agents/pharmacology , Piperazines/chemistry , Potassium/urine , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/metabolism , Rats , Rats, Inbred SHR
9.
Toxins (Basel) ; 12(12)2020 12 16.
Article En | MEDLINE | ID: mdl-33339256

Recently developed fluorescent protein-scorpion toxin chimeras (FP-Tx) show blocking activities for potassium voltage-gated channels of Kv1 family and retain almost fully pharmacological profiles of the parental peptide toxins (Kuzmenkov et al., Sci Rep. 2016, 6, 33314). Here we report on N-terminally green fluorescent protein (GFP)-tagged agitoxin 2 (GFP-L2-AgTx2) with high affinity and selectivity for the binding site of Kv1.3 channel involved in the pathogenesis of various (primarily of autoimmune origin) diseases. The basis for this selectivity relates to N-terminal location of GFP, since transposition of GFP to the C-terminus of AgTx2 recovered specific interactions with the Kv1.1 and Kv1.6 binding sites. Competitive binding experiments revealed that the binding site of GFP-L2-AgTx2 overlaps that of charybdotoxin, kaliotoxin 1, and agitoxin 2, the known Kv1.3-channel pore blockers. GFP-L2-AgTx2 was demonstrated to be applicable as a fluorescent probe to search for Kv1.3 pore blockers among individual compounds and in complex mixtures, to measure blocker affinities, and to visualize Kv1.3 distribution at the plasma membrane of Kv1.3-expressing HEK293 cells. Our studies show that definite combinations of fluorescent proteins and peptide blockers can result in considerable modulation of the natural blocker-channel binding profile yielding selective fluorescent ligands of certain channels.


Green Fluorescent Proteins/metabolism , Kv1.3 Potassium Channel/metabolism , Potassium Channel Blockers/metabolism , Scorpion Venoms/metabolism , Amino Acid Sequence , Binding Sites/physiology , Green Fluorescent Proteins/chemistry , HEK293 Cells , Humans , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/chemistry , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Protein Structure, Secondary , Scorpion Venoms/analysis , Scorpion Venoms/chemistry
10.
ChemMedChem ; 15(24): 2462-2469, 2020 12 15.
Article En | MEDLINE | ID: mdl-33043595

The Ca2+ activated potassium channel 3.1 (KCa 3.1) is involved in critical steps of the metastatic cascade, such as proliferation, migration, invasion and extravasation. Therefore, a fast and efficient protocol for imaging of KCa 3.1 channels was envisaged. The novel fluorescently labeled small molecule imaging probes 1 and 2 were synthesized by connecting a dimethylpyrrole-based BODIPY dye with a derivative of the KCa 3.1 channel inhibitor senicapoc via linkers of different length. Patch-clamp experiments revealed the inhibition of KCa 3.1 channels by the probes confirming interaction with the channel. Both probes 1 and 2 were able to stain KCa 3.1 channels in non-small-cell lung cancer (NSCLC) cells following a simple, fast and efficient protocol. Pre-incubation with unlabeled senicapoc removed the punctate staining pattern showing the specificity of the new probes 1 and 2. Staining of the channel with the fluorescently labeled senicapoc derivatives 1 or 2 or with antibody-based indirect immunofluorescence yielded identical or very similar densities of stained KCa 3.1 channels. However, co-staining using both methods did not lead to the expected overlapping punctate staining pattern. This observation was explained by docking studies showing that the antibody used for indirect immunofluorescence and the probes 1 and 2 label different channel populations. Whereas the antibody binds at the closed channel conformation, the probes 1 and 2 bind within the open channel.


Acetamides/pharmacology , Boron Compounds/pharmacology , Fluorescent Dyes/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Trityl Compounds/pharmacology , A549 Cells , Acetamides/metabolism , Animals , Antibodies/immunology , Antibodies/metabolism , Boron Compounds/metabolism , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes/metabolism , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/immunology , Mice , Patch-Clamp Techniques , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Protein Binding , Staining and Labeling , Trityl Compounds/metabolism
11.
Biomed Pharmacother ; 129: 110383, 2020 Sep.
Article En | MEDLINE | ID: mdl-32563149

Withaferin A (WFA), a C5,C6-epoxy steroidal lactone isolated from the medicinal plant Withania somnifera (L.) Dunal, inhibits growth of tumor cells in different cancer types. However, the mechanisms underlying the effect of WFA on tumor cells are not fully understood. In the present study, we evaluated the blockade of TASK-3 channels by WFA in TASK-3-expressing HEK-293 cells. Explore if the WFA-mediated TASK-3 blockade can be used as a pharmacological tool to decrease the cell viability in cancer cells. A combination of functional experiments (patch-clamp, gene downregulation, overexpression and pharmacological inhibition) and molecular docking analysis were used to get insights into the mechanism by which the inhibition of TASK-3 by WFA affects the growth and viability of cancer cells. Withaferin A was found to inhibit the activity of TASK-3 channels. The inhibitory effect of Withaferin A on TASK-3 potassium currents was dose-dependent and independent of voltage. Molecular modeling studies identified putative WFA-binding sites in TASK-3 channel involved the channel blockade. In agreements with the molecular modeling predictions, mutation of residues F125 to A (F125A), L197 to V (L197 V) and the double mutant F125A-L197 V markedly decreased the WFA-induced inhibition of TASK-3. Finally, the cytotoxic effect of WFA was tested in MDA-MB-231 human breast cancer cells transfected with TASK-3 or shRNA that decreases TASK-3 expression. Together, our results show that the cytotoxic effect of WFA on fully transformed MDA-MB-231 cells depends on the expression of TASK-3. Herein, we also provide insights into the mechanism of TASK-3 inhibition by WFA.


Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Withanolides/pharmacology , Antineoplastic Agents, Phytogenic/metabolism , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Membrane Potentials , Potassium Channel Blockers/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Protein Binding , Signal Transduction , Withanolides/metabolism
12.
BMC Mol Cell Biol ; 21(1): 31, 2020 Apr 19.
Article En | MEDLINE | ID: mdl-32306897

BACKGROUND: Cytoprotection afforded by mitochondrial ATP-sensitive K+-channel (mKATP-channel) opener diazoxide (DZ) largely depends on the activation of potassium cycle with eventual modulation of mitochondrial functions and ROS production. However, generally these effects were studied in the presence of Mg∙ATP known to block K+ transport. Thus, the purpose of our work was the estimation of DZ effects on K+ transport, K+ cycle and ROS production in rat liver mitochondria in the absence of Mg∙ATP. RESULTS: Without Mg·ATP, full activation of native mKATP-channel, accompanied by the increase in ATP-insensitive K+ uptake, activation of K+-cycle and respiratory uncoupling, was reached at ≤0.5 µM of DZ,. Higher diazoxide concentrations augmented ATP-insensitive K+ uptake, but not mKATP-channel activity. mKATP-channel was blocked by Mg·ATP, reactivated by DZ, and repeatedly blocked by mKATP-channel blockers glibenclamide and 5-hydroxydecanoate, whereas ATP-insensitive potassium transport was blocked by Mg2+ and was not restored by DZ. High sensitivity of potassium transport to DZ in native mitochondria resulted in suppression of mitochondrial ROS production caused by the activation of K+-cycle on sub-micromolar scale. Based on the oxygen consumption study, the share of mKATP-channel in respiratory uncoupling by DZ was found. CONCLUSIONS: The study of mKATP-channel activation by diazoxide in the absence of MgATP discloses novel, not described earlier, aspects of mKATP-channel interaction with this drug. High sensitivity of mKATP-channel to DZ results in the modulation of mitochondrial functions and ROS production by DZ on sub-micromolar concentration scale. Our experiments led us to the hypothesis that under the conditions marked by ATP deficiency affinity of mKATP-channel to DZ can increase, which might contribute to the high effectiveness of this drug in cardio- and neuroprotection.


Adenosine Triphosphate/metabolism , Diazoxide/pharmacology , Mitochondria, Liver/drug effects , Potassium Channels/metabolism , Potassium/metabolism , Animals , Decanoic Acids/pharmacology , Energy Metabolism/drug effects , Female , Glyburide/pharmacology , Hydroxy Acids/pharmacology , Ion Transport/drug effects , Ion Transport/genetics , KATP Channels/metabolism , Magnesium/metabolism , Mitochondria, Liver/metabolism , Oxygen Consumption/drug effects , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels/genetics , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
13.
Eur J Pharmacol ; 878: 173097, 2020 Jul 05.
Article En | MEDLINE | ID: mdl-32278853

It is well demonstrated that antidepressant fluoxetine has significant inhibitory effects on voltage-gated potassium channels. So far, the concise regulation of fluoxetine on Kv2.1, the predominant delayed rectifier potassium channel subtype in the central nervous system, are rarely reported. Here patch-clamp recording was used to investigate the inhibitory effects of fluoxetine on Kv2.1 potassium channels stably expressed in HEK293 cells. The results showed fluoxetine dose-dependently suppressed Kv2.1 currents with an IC50 of 51.3 µM. At the test potential positive to +50 mV, fluoxetine 50 µM voltage-dependently suppressed Kv2.1 currents with an electrical distance δ of 0.28. Moreover, fluoxetine 50 µM did not affect the activation process of Kv2.1, but reduced the decay time constant τinact and obviously accelerated the inactivation process of Kv2.1 and left-shifted the half-maximal inactivation potential of Kv2.1 potassium channel by 9.8 mV. Fluoxetine 50 µM notably delayed the recovery process of Kv2.1 from inactivation with increased time constants. In addition, fluoxetine 50 µM use-dependently inhibited Kv2.1 currents at different frequencies. In conclusion, the inhibition of Kv2.1 by fluoxetine was concentration-dependent, voltage-dependent and use-dependent. The accelerated steady-state inactivation of Kv2.1 channels induced by fluoxetine might be ascribed to the delay of the recovery process of Kv2.1.


Antidepressive Agents/pharmacology , Fluoxetine/pharmacology , Potassium Channel Blockers/metabolism , Shab Potassium Channels/antagonists & inhibitors , Dose-Response Relationship, Drug , Gene Expression Regulation , HEK293 Cells , Humans , Ion Channel Gating , Kinetics , Membrane Potentials , Patch-Clamp Techniques/methods
14.
Am J Physiol Renal Physiol ; 318(6): F1369-F1376, 2020 06 01.
Article En | MEDLINE | ID: mdl-32308018

Cytochrome P-450 (Cyp) epoxygenase-dependent metabolites of arachidonic acid (AA) have been shown to inhibit renal Na+ transport, and inhibition of Cyp-epoxygenase is associated with salt-sensitive hypertension. We used the patch-clamp technique to examine whether Cyp-epoxygenase-dependent AA metabolites inhibited the basolateral 40-pS K+ channel (Kir4.1/Kir5.1) in the distal convoluted tubule (DCT). Application of AA inhibited the basolateral 40-pS K+ channel in the DCT. The inhibitory effect of AA on the 40-pS K+ channel was specific because neither linoleic nor oleic acid was able to mimic the effect of AA on the K+ channel. Inhibition of Cyp-monooxygenase with N-methylsulfonyl-12,12-dibromododec-11-enamide or inhibition of cyclooxygenase with indomethacin failed to abolish the inhibitory effect of AA on the 40-pS K+ channel. However, the inhibition of Cyp-epoxygenase with N-methylsulfonyl-6-(propargyloxyphenyl)hexanamide abolished the effect of AA on the 40-pS K+ channel in the DCT. Moreover, addition of either 11,12-epoxyeicosatrienoic acid (EET) or 14,15-EET also inhibited the 40-pS K+ channel in the DCT. Whole cell recording demonstrated that application of AA decreased, whereas N-methylsulfonyl-6-(propargyloxyphenyl)hexanamide treatment increased, Ba2+-sensitive K+ currents in the DCT. Finally, application of 14,15-EET but not AA was able to inhibit the basolateral 40-pS K+ channel in the DCT of Cyp2c44-/- mice. We conclude that Cyp-epoxygenase-dependent AA metabolites inhibit the basolateral Kir4.1/Kir5.1 in the DCT and that Cyp2c44-epoxygenase plays a role in the regulation of the basolateral K+ channel in the mouse DCT.


8,11,14-Eicosatrienoic Acid/analogs & derivatives , Arachidonic Acid/pharmacology , Cytochrome P450 Family 2/metabolism , Kidney Tubules, Distal/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , 8,11,14-Eicosatrienoic Acid/metabolism , 8,11,14-Eicosatrienoic Acid/pharmacology , Amides/pharmacology , Animals , Arachidonic Acid/metabolism , Cytochrome P450 Family 2/antagonists & inhibitors , Cytochrome P450 Family 2/genetics , Enzyme Inhibitors/pharmacology , Kidney Tubules, Distal/metabolism , Male , Membrane Potentials , Mice, 129 Strain , Mice, Knockout , Potassium Channel Blockers/metabolism , Potassium Channels, Inwardly Rectifying/metabolism
15.
J Nanobiotechnology ; 18(1): 21, 2020 Jan 28.
Article En | MEDLINE | ID: mdl-31992314

BACKGROUND: Carbon nanoparticles (CNPs) have been reported to boost plant growth, while the mechanism that CNPs enhanced potassium uptake for plant growth has not been reported so far. RESULTS: In this study, the function that CNPs promoted potassium uptake in BY-2 cells was established and the potassium accumulated in cells had a significant correlation with the fresh biomass of BY-2 cells. The K+ accumulation in cells increased with the increasing concentration of CNPs. The K+ influx reached high level after treatment with CNPs and was significantly higher than that of the control group and the negative group treated with K+ channels blocker, tetraethylammonium chloride (TEA+). The K+ accumulation was not reduced in the presence of CNPs inhibitors. In the presence of potassium channel blocker TEA+ or CNPs inhibitors, the NKT1 gene expression was changed compared with the control group. The CNPs were found to preferentially transport K+ than other cations determined by rectification of ion current assay (RIC) in a conical nanocapillary. CONCLUSIONS: These results indicated that CNPs upregulated potassium gene expression to enhance K+ accumulation in BY-2 cells. Moreover, it was speculated that the CNPs simulated protein of ion channels via bulk of carboxyl for K+ permeating. These findings will provide support for improving plant growth by carbon nanoparticles.


Carbon/chemistry , Nanoparticles/chemistry , Nanoparticles/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Potassium/metabolism , Amino Acids/analysis , Amino Acids/metabolism , Cell Membrane Permeability , Gene Expression Regulation/drug effects , Genetic Enhancement , Humans , Membrane Potentials , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Tetraethylammonium/chemistry , Tetraethylammonium/metabolism , Up-Regulation/drug effects
16.
Sci Rep ; 10(1): 52, 2020 01 09.
Article En | MEDLINE | ID: mdl-31919372

4-Aminopyridine (4AP) is a specific blocker of voltage-gated potassium channels (KV1 family) clinically approved for the symptomatic treatment of patients with multiple sclerosis (MS). It has recently been shown that [18F]3F4AP, a radiofluorinated analog of 4AP, also binds to KV1 channels and can be used as a PET tracer for the detection of demyelinated lesions in rodent models of MS. Here, we investigate four novel 4AP derivatives containing methyl (-CH3), methoxy (-OCH3) as well as trifluoromethyl (-CF3) in the 2 and 3 position as potential candidates for PET imaging and/or therapy. We characterized the physicochemical properties of these compounds (basicity and lipophilicity) and analyzed their ability to block Shaker K+ channel under different voltage and pH conditions. Our results demonstrate that three of the four derivatives are able to block voltage-gated potassium channels. Specifically, 3-methyl-4-aminopyridine (3Me4AP) was found to be approximately 7-fold more potent than 4AP and 3F4AP; 3-methoxy- and 3-trifluoromethyl-4-aminopyridine (3MeO4AP and 3CF34AP) were found to be about 3- to 4-fold less potent than 4AP; and 2-trifluoromethyl-4-AP (2CF34AP) was found to be about 60-fold less active. These results suggest that these novel derivatives are potential candidates for therapy and imaging.


4-Aminopyridine/analogs & derivatives , Drosophila Proteins/metabolism , Potassium Channel Blockers/metabolism , Potassium Channels, Voltage-Gated/metabolism , 4-Aminopyridine/metabolism , Action Potentials/drug effects , Animals , Drosophila/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Hydrogen-Ion Concentration , Kinetics , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/chemistry , Potassium Channels, Voltage-Gated/genetics , Structure-Activity Relationship , Xenopus laevis/growth & development
17.
Bioorg Med Chem Lett ; 30(4): 126928, 2020 02 15.
Article En | MEDLINE | ID: mdl-31889664

One of the most commonly used strategies to reduce hERG (human ether-a-go-go) activity in the drug candidates is introduction of a carboxylic acid group. During the optimization of PPARδ modulators, some of the compounds containing a carboxylic acid were found to inhibit the hERG channel in a patch clamp assay. By modifying the basicity of the imidazole core, potent and selective PPARδ modulators that do not inhibit hERG channel were identified. Some of the modulators have excellent pharmacokinetic profiles in mice.


Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , PPAR delta/chemistry , Potassium Channel Blockers/chemistry , Drug Design , Ether-A-Go-Go Potassium Channels/metabolism , Half-Life , Humans , Kinetics , PPAR delta/genetics , PPAR delta/metabolism , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Structure-Activity Relationship , Transcriptional Activation/drug effects
18.
Eur J Pharmacol ; 870: 172921, 2020 Mar 05.
Article En | MEDLINE | ID: mdl-31935397

We explored the effect of the muscarinic receptor antagonist tolterodine on voltage-dependent K+ (Kv) channels using the patch-clamp technique in coronary arterial smooth muscle cells freshly isolated from rabbits. Tolterodine inhibited Kv channels in a concentration-dependent manner, with an IC50 of 1.71 ± 0.33 µM and Hill coefficient of 0.69 ± 0.03. Tolterodine accelerated the decay rate of Kv channel inactivation. The apparent rate constants of association and dissociation for tolterodine were 1.79 ± 0.13 µM-1s-1, and 3.13 ± 0.96 s-1, respectively. Although 3 µM tolterodine had no effect on the steady-state activation of the Kv current, it shifted the steady-state inactivation curve towards a negative potential. Application of consecutive train steps (1 or 2 Hz) progressively decreased the Kv current and promoted its inhibition. Furthermore, the recovery time constant was augmented in the presence of tolterodine, indicating that tolterodine-induced Kv channel blockade is use (state) dependent. Pretreatment with inhibitors of the Kv1.5, Kv2.1, and Kv7 subtypes (DPO-1, guangxitoxin, and linopirdine) partially reduced the inhibitory effect of tolterodine on Kv channels. The alternative muscarinic receptor antagonist atropine did not inhibit the Kv current nor influence tolterodine-induced inhibition of the Kv current. Tolterodine induced vasoconstriction and membrane depolarization. Based on these results, we conclude that tolterodine inhibits Kv channels in concentration-, time-, and use (state)-dependent manners, irrespective of its antagonism of muscarinic receptors.


Muscarinic Antagonists/metabolism , Myocytes, Smooth Muscle/metabolism , Potassium Channel Blockers/metabolism , Potassium Channels, Voltage-Gated/metabolism , Tolterodine Tartrate/metabolism , Animals , Cells, Cultured , Coronary Vessels/metabolism , Male , Membrane Potentials/drug effects , Muscle, Smooth, Vascular/cytology , Patch-Clamp Techniques , Rabbits
19.
J Med Chem ; 63(1): 216-230, 2020 01 09.
Article En | MEDLINE | ID: mdl-31838846

Gain-of-function of BK channels or knockout of their ß4 subunit is associated with spontaneous epilepsy. Currently, efficacy of BK (α + ß4) channel modulators in preventing epilepsy was never reported. Here, we show that martentoxin selectively inhibits BK (α + ß4) channels by interaction with the extracellular loop of the BK ß4 subunit (hß4-loop) at a molar ratio 4:1 (hß4-loop vs martentoxin). Residues Glu104, Glu122, Gln124, Lys125, and Glu128 of the hß4-loop form hydrogen bonds with residues Asp5, Glu13, Lys20, Ser24, Gln26, Lys28, and Arg35 of martentoxin, by which martentoxin reduces the neuronal spiking frequency and increases interspike intervals. Intrahippocampal infusion of martentoxin significantly increases the latency time of seizure, reduces seizure duration and seizure numbers on pentylenetetrazole-induced presensitized rats, inhibits hippocampal hyperexcitability and c-Fos expression, and displays neuroprotective effects on hippocampal neurons. These results suggest that the BK (α + ß4) channel is a novel therapeutic target of intractable epilepsy and martentoxin contributes to the rational drug design for epilepsy treatment.


Anticonvulsants/therapeutic use , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Potassium Channel Blockers/therapeutic use , Scorpion Venoms/therapeutic use , Seizures/prevention & control , Animals , Anticonvulsants/metabolism , GABA-A Receptor Antagonists/pharmacology , Humans , Male , Neuroprotective Agents/metabolism , Neuroprotective Agents/therapeutic use , Pentylenetetrazole/pharmacology , Potassium Channel Blockers/metabolism , Protein Binding , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Scorpion Venoms/metabolism
20.
Sci Rep ; 9(1): 16586, 2019 11 12.
Article En | MEDLINE | ID: mdl-31719645

The human ether-a-go-go-related gene (hERG) encodes a voltage-gated potassium channel that plays an essential role in the repolarization of action potentials in cardiac muscle. However, various drugs can block the ion current by binding to the hERG channel, resulting in potentially lethal cardiac arrhythmia. Accordingly, in silico studies are necessary to clarify the mechanisms of how these drugs bind to the hERG channel. Here, we used the experimental structure of the hERG channel, determined by cryo-electron microscopy, to perform docking simulations to predict the complex structures that occur between the hERG channel and structurally diverse drugs. The absolute binding free energies for the models were calculated using the MP-CAFEE method; calculated values were well correlated with experimental ones. By applying the regression equation obtained here, the affinity of a drug for the hERG channel can be accurately predicted from the calculated value of the absolute binding free energy.


Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/metabolism , Models, Molecular , Molecular Dynamics Simulation , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Action Potentials , Binding Sites , Humans , Protein Conformation , Thermodynamics
...