Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 263
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Prostaglandins Other Lipid Mediat ; 169: 106782, 2023 12.
Article in English | MEDLINE | ID: mdl-37741358

ABSTRACT

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.


Subject(s)
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
2.
Biochem Genet ; 60(2): 504-526, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34286408

ABSTRACT

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.


Subject(s)
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
3.
Int J Mol Sci ; 23(3)2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35163644

ABSTRACT

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.


Subject(s)
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
4.
J Mol Cell Cardiol ; 158: 163-177, 2021 09.
Article in English | MEDLINE | ID: mdl-34062207

ABSTRACT

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.


Subject(s)
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
5.
J Biol Chem ; 294(48): 18349-18359, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31533989

ABSTRACT

Tk-hefu is an artificial peptide designed based on the α-hairpinin scaffold, which selectively blocks voltage-gated potassium channels Kv1.3. Here we present its spatial structure resolved by NMR spectroscopy and analyze its interaction with channels using computer modeling. We apply protein surface topography to suggest mutations and increase Tk-hefu affinity to the Kv1.3 channel isoform. We redesign the functional surface of Tk-hefu to better match the respective surface of the channel pore vestibule. The resulting peptide Tk-hefu-2 retains Kv1.3 selectivity and displays ∼15 times greater activity compared with Tk-hefu. We verify the mode of Tk-hefu-2 binding to the channel outer vestibule experimentally by site-directed mutagenesis. We argue that scaffold engineering aided by protein surface topography represents a reliable tool for design and optimization of specific ion channel ligands.


Subject(s)
Kv1.3 Potassium Channel/chemistry , Peptides/chemistry , Potassium Channel Blockers/chemistry , Proteins/chemistry , Amino Acid Sequence , Animals , Humans , Kv1.3 Potassium Channel/metabolism , Ligands , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Dynamics Simulation , Mutation , Peptides/genetics , Peptides/metabolism , Potassium Channel Blockers/metabolism , Protein Binding , Protein Conformation , Proteins/metabolism , Surface Properties
6.
Am J Physiol Renal Physiol ; 318(6): F1369-F1376, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32308018

ABSTRACT

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.


Subject(s)
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
7.
Bioorg Med Chem Lett ; 30(4): 126928, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31889664

ABSTRACT

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.


Subject(s)
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
8.
J Nanobiotechnology ; 18(1): 21, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31992314

ABSTRACT

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.


Subject(s)
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
9.
Biotechnol Appl Biochem ; 66(1): 119-129, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30380177

ABSTRACT

Scorpine is a cationic protein from the venom of Pandinus imperator, belonging to potassium channel blocker family, which has been shown to have antibacterial, antiviral, and antiplasmodia activities. In the present study, a pET-44a vector containing scorpine synthetic gene with T7 Promoter (pET 44a-His6-Nus-His6-tev-scorpine) was transferred into Escherichia coli Rosetta-gami B (DE3) for soluble expression of the protein in the cytoplasm and its overproduction. After confirming recombinant scorpine peptide expression using SDS-PAGE and Western blot, augmentation of production was performed during two stages. At first, effects of three parameters including carbon source concentration of medium, temperature, and induction time were investigated in terrific broth (TB) medium. Afterward, the overexpression was performed by response surface methodology in TB + glucose. Under the optimized conditions, the highest production of 3.5 g/L in the TB + glucose medium (7.5 g/L glucose, induction at OD600 = 3.5 and 25 °C) was increased to 4.1 g/L in TB medium (2.5 g/L glycerol, induction at OD600 = 0.7 and 25 °C). Then, in order to increase the amount of protein production, effects of carbon concentration in the fermenter under the primary optimized condition was investigated. The amount of produced recombinant protein increased from 0.12 to 2.1 g/L.H. The results were similar to previous studies on optimizing and increasing the production of recombinant protein and in particular recombinant scorpine.


Subject(s)
Defensins , Escherichia coli/metabolism , Gene Expression , Potassium Channel Blockers , Defensins/biosynthesis , Defensins/genetics , Defensins/isolation & purification , Escherichia coli/genetics , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/isolation & purification , Potassium Channel Blockers/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
10.
Pharm Res ; 35(8): 159, 2018 Jun 18.
Article in English | MEDLINE | ID: mdl-29915999

ABSTRACT

PURPOSE: The renal clearance of fampridine (Fampyra®, or Ampyra®) significantly exceeds the glomerular filtration rate, suggesting active renal secretion is likely the major elimination pathway. The goal of this study was to identify the renal transporters that are involved in the renal active secretion, and elucidate the active renal secretion mechanism of fampridine. METHODS: The uptake of fampridine to HEK-293 cells overexpressing human OCT2, MATE1 or MATE2K was determined in the absence and presence of Cimetidine, the prototypical inhibitor of the transporters. The inhibition potential of fampridine on the renal transporters was evaluated by determining the uptake of TEA and Metformin, the probe substrates of the transporters of OCT2 and MATEs, respectively, in the absence or presence of fampridine. RESULTS: Significant time- and concentration-dependent uptake of fampridine by human OCT2 was observed. The Km and Vmax were determined as 51.0 ± 17.1 µM and 1107 ± 136 pmole/min/106 cells, respectively. Fampridine also inhibited OCT2 mediated uptake of Metformin with estimated IC50 of 66.8 µM. In contrast, there was not significant uptake of fampridine by human MATE1 or MATE2K, and fampridine did not inhibit MATE1 or MATE2K mediated uptake of TEA. CONCLUSION: The studies indicated fampridine is a substrate and inhibitor of OCT2, but not MATE1 or MATE2K. Results from the study suggested the active renal secretion of fampridine is mediated by human OCT2 but not MATE1 or MATE2K. To our knowledge, fampridine is the first reported substrate specific to OCT2 but not to MATE1 or MATE2K.


Subject(s)
4-Aminopyridine/pharmacokinetics , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/metabolism , Potassium Channel Blockers/pharmacokinetics , 4-Aminopyridine/metabolism , 4-Aminopyridine/pharmacology , Biological Transport/drug effects , HEK293 Cells , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacokinetics , Metformin/metabolism , Metformin/pharmacokinetics , Organic Cation Transporter 2/antagonists & inhibitors , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology
11.
J Cardiovasc Pharmacol ; 71(1): 10-18, 2018 01.
Article in English | MEDLINE | ID: mdl-29283926

ABSTRACT

Propofol blocks the voltage-gated human Kv1.5 (hKv1.5) channel by preferentially affecting in its open state. A previous mutational study suggested that several amino acids within the pore region of the hKv1.5 channel are involved in mediating the blocking action of propofol. The present investigation was undertaken to elucidate the predicted binding modes of propofol within the pore cavity of the open-state hKv1.5 channel, using computational docking and mutagenesis approaches. The docking simulation using a homology model of the hKv1.5 channel, constructed based on the crystal structure of the Kv1.2 channel, predicted that propofol was positioned at the base of the pore cavity of hKv1.5 channel, adjacent to 4 amino acids Thr479, Thr480, Val505, and Ile508, and formed arene-H interactions with Val505. The patch-clamp experiments on wild-type and mutant hKv1.5 channels constructed by site-directed mutagenesis revealed that the blocking potency of propofol was significantly reduced in T480A, V505A, and I508A but not in T479A mutants compared with wild-type hKv1.5 channel. These computational docking and experimental mutational analyses suggest that propofol is positioned at the base of the pore cavity and forms functional contact with Thr480, Val505, and Ile508 to directly block the hKv1.5 channel.


Subject(s)
Ion Channel Gating/drug effects , Kv1.5 Potassium Channel/antagonists & inhibitors , Molecular Docking Simulation , Mutagenesis, Site-Directed , Potassium Channel Blockers/pharmacology , Propofol/pharmacology , Animals , Binding Sites , CHO Cells , Cricetulus , Humans , Kv1.5 Potassium Channel/chemistry , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Membrane Potentials/drug effects , Mutation , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Propofol/chemistry , Propofol/metabolism , Protein Binding , Protein Conformation , Structure-Activity Relationship
12.
Molecules ; 23(6)2018 06 20.
Article in English | MEDLINE | ID: mdl-29925780

ABSTRACT

The human defensins are recently discovered to inhibit potassium channels, which are classical targets of the animal toxins. Whether other vertebrate defensins are potassium channel inhibitors remains unknown. In this work, we reported that the mouse ß-defensin 3 (mBD3) was a novel inhibitor of both endogenous and exogenous potassium channels. The structural analysis showed that mBD3 is the most identical to human Kv1.3 channel-sensitive human ß-defensin 2 (hBD2). However, the pharmacological profiles indicated that the recombinant mBD3 (rmBD3) weakly inhibited the mouse and human Kv1.3 channels. Different from the pharmacological features of human ß-defensins, mBD3 more selectively inhibited the mouse Kv1.6 and human KCNQ1/KCNE1 channels with IC50 values of 0.6 ± 0.4 µM and 1.2 ± 0.8 µM, respectively. The site directed mutagenesis experiments indicated that the extracellular pore region of mouse Kv1.6 channel was the interaction site of rmBD3. In addition, the minor effect on the channel conductance-voltage relationship curves implied that mBD3 might bind the extracellular transmembrane helices S1-S2 linker and/or S3-S4 linker of mouse Kv1.6 channel. Together, these findings not only revealed mBD3 as a novel inhibitor of both endogenous and exogenous potassium channels, but also provided a clue to investigate the role of mBD3-Kv1.6 channel interaction in the physiological and pathological field in the future.


Subject(s)
Potassium Channel Blockers/metabolism , beta-Defensins/metabolism , Animals , Binding Sites , Escherichia coli , HEK293 Cells , Humans , Ion Channel Gating , Mice , Protein Binding , Protein Conformation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , beta-Defensins/genetics
13.
J Biol Chem ; 291(13): 7097-106, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26817841

ABSTRACT

The structural similarity between defensins and scorpion neurotoxins suggests that they might have evolved from a common ancestor. However, there is no direct experimental evidence demonstrating a functional link between scorpion neurotoxins and defensins. The scorpion defensin BmKDfsin4 from Mesobuthus martensiiKarsch contains 37 amino acid residues and a conserved cystine-stabilized α/ß structural fold. The recombinant BmKDfsin4, a classical defensin, has been found to have inhibitory activity against Gram-positive bacteria such as Staphylococcus aureus, Bacillus subtilis, and Micrococcus luteusas well as methicillin-resistant Staphylococcus aureus Interestingly, electrophysiological experiments showed that BmKDfsin4,like scorpion potassium channel neurotoxins, could effectively inhibit Kv1.1, Kv1.2, and Kv1.3 channel currents, and its IC50value for the Kv1.3 channel was 510.2 nm Similar to the structure-function relationships of classical scorpion potassium channel-blocking toxins, basic residues (Lys-13 and Arg-19) of BmKDfsin4 play critical roles in peptide-Kv1.3 channel interactions. Furthermore, mutagenesis and electrophysiological experiments demonstrated that the channel extracellular pore region is the binding site of BmKDfsin4, indicating that BmKDfsin4 adopts the same mechanism for blocking potassium channel currents as classical scorpion toxins. Taken together, our work identifies scorpion BmKDfsin4 as the first invertebrate defensin to block potassium channels. These findings not only demonstrate that defensins from invertebrate animals are a novel type of potassium channel blockers but also provide evidence of a functional link between defensins and neurotoxins.


Subject(s)
Anti-Bacterial Agents/chemistry , Defensins/chemistry , Neurotoxins/chemistry , Potassium Channel Blockers/chemistry , Scorpion Venoms/chemistry , Amino Acid Sequence , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacillus subtilis/drug effects , Bacillus subtilis/growth & development , Defensins/genetics , Defensins/metabolism , Defensins/pharmacology , Gene Expression , Humans , Kv1.1 Potassium Channel/antagonists & inhibitors , Kv1.1 Potassium Channel/genetics , Kv1.1 Potassium Channel/metabolism , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/genetics , Kv1.2 Potassium Channel/metabolism , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/growth & development , Mice , Micrococcus luteus/drug effects , Micrococcus luteus/growth & development , Models, Molecular , Molecular Sequence Data , Neurotoxins/genetics , Neurotoxins/metabolism , Neurotoxins/pharmacology , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Scorpion Venoms/biosynthesis , Scorpions/chemistry , Scorpions/physiology , Sequence Alignment , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Structural Homology, Protein , Structure-Activity Relationship
14.
J Cell Physiol ; 232(8): 2019-2032, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27255432

ABSTRACT

Several reports credit mibefradil with tumor suppressing properties arising from its known inhibition of Ca2+ currents. Given that mibefradil (Mb) is also known to inhibit K+ channels, we decided to study the interaction between this organic compound and the tumor-related Kv10.1 channel. Here we report that Mb modulates the gating of Kv10.1. Mb induces an apparent inactivation from both open and early closed states where the channels dwell at hyperpolarized potentials. Additionally, Mb accelerates the kinetics of current activation, in a manner that depends on initial conditions. Our observations suggest that Mb binds to the voltage sensor domain of Kv10.1 channels, thereby modifying the gating of the channels in a way that in some, but not all, aspects opposes to the gating effects exerted by divalent cations. J. Cell. Physiol. 232: 2019-2032, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ion Channel Gating/drug effects , Mibefradil/pharmacology , Potassium Channel Blockers/pharmacology , Binding Sites , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , HEK293 Cells , Humans , Kinetics , Membrane Potentials , Mibefradil/metabolism , Models, Biological , Potassium Channel Blockers/metabolism , Protein Binding , Transfection
15.
Cell Physiol Biochem ; 44(1): 172-184, 2017.
Article in English | MEDLINE | ID: mdl-29131061

ABSTRACT

BACKGROUND/AIMS: The human-voltage gated Kv1.3 channel (hKv1.3) is expressed in T- and B lymphocytes. Verapamil is able to block hKv1.3 channels. We characterized the effect of verapamil on currents through hKv1.3 channels paying special attention to the on-rate (kon) of verapamil. By comparing on-rates obtained in wild-type (wt) and mutant channels a binding pocket for verapamil and impacts of different amino acid residues should be investigated. METHODS: Using the whole-cell patch clamp technique the action of verapamil on currents through wild-type and six hKv1.3 mutant channels in the open state was investigated by measuring the time course of the open channel block in order to calculate kon of verapamil. RESULTS: The on-rate of verapamil to block current through hKv1.3_T419C mutant channels is similar to that obtained for hKv1.3_wt channels whereas the on-rate of verapamil to block currents through hKv1.3_L417C and hKv1.3_L418C mutant channels was ∼ 3 times slower compared to in wt channels. The on-rate of verapamil to block currents through hKv1.3_L346C and the double mutant hKv1.3_L346C_L418C channel was ∼ 2 times slower compared to that obtained in the wt channel. The hKv1.3_I420C mutant channel reduced the on-rate of verapamil to block currents ∼ 6 fold. CONCLUSIONS: We conclude that position 420 in hKv1.3 channels maximally interferes with verapamil reaching its binding site to block the channel. Positions 417 and 418 in hKv1.3 channels partially hinder verapamil reaching its binding site to block the channel whereas position 419 may not interfere with verapamil at all. Mutant hKv1.3_L346C and hKv1.3_L346C_L418C mutant channels might indirectly influence the ability of verapamil reaching its binding site to block current.


Subject(s)
Kv1.3 Potassium Channel/metabolism , Potassium Channel Blockers/metabolism , Verapamil/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Humans , Kinetics , Kv1.3 Potassium Channel/genetics , Membrane Potentials/drug effects , Molecular Docking Simulation , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Protein Binding , Protein Structure, Tertiary , Verapamil/chemistry , Verapamil/pharmacology
16.
Biochim Biophys Acta Proteins Proteom ; 1865(5): 465-472, 2017 May.
Article in English | MEDLINE | ID: mdl-28179135

ABSTRACT

We report isolation, sequencing, and electrophysiological characterization of OSK3 (α-KTx 8.8 in Kalium and Uniprot databases), a potassium channel blocker from the scorpion Orthochirus scrobiculosus venom. Using the voltage clamp technique, OSK3 was tested on a wide panel of 11 voltage-gated potassium channels expressed in Xenopus oocytes, and was found to potently inhibit Kv1.2 and Kv1.3 with IC50 values of ~331nM and ~503nM, respectively. OdK1 produced by the scorpion Odontobuthus doriae differs by just two C-terminal residues from OSK3, but shows marked preference to Kv1.2. Based on the charybdotoxin-potassium channel complex crystal structure, a model was built to explain the role of the variable residues in OdK1 and OSK3 selectivity.


Subject(s)
Potassium Channel Blockers/chemistry , Protein Conformation , Scorpion Venoms/metabolism , Amino Acid Sequence/genetics , Animals , Crystallography, X-Ray , Electrophysiology , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/chemistry , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/chemistry , Oocytes/metabolism , Patch-Clamp Techniques , Potassium/chemistry , Potassium/metabolism , Potassium Channel Blockers/isolation & purification , Potassium Channel Blockers/metabolism , Scorpion Venoms/chemistry , Scorpion Venoms/genetics , Scorpion Venoms/isolation & purification , Scorpions/chemistry , Scorpions/metabolism , Xenopus/genetics
17.
Amino Acids ; 49(11): 1895-1906, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28900735

ABSTRACT

The effects of 4-chloro-3-nitro-N-butylbenzenesulfonamide (SMD2) on KV3.1 channels, heterologous expressed in L-929 cells, were studied with the whole cell patch-clamp technique. SMD2 blocks KV3.1 in a reversible and use-dependent manner, with IC50 around 10 µM, and a Hill coefficient around 2. Although the conductance vs. voltage relationship in control condition can be described by a single Boltzmann function, two terms are necessary to describe the data in the presence of SMD2. The activation and deactivation time constants are weakly voltage dependent both for control and in the presence of SMD2. SMD2 does not change the channel selectivity and tail currents show a typical crossover phenomenon. The time course of inactivation has a fast and a slow component, and SMD2 significantly decreased their values. Steady-state inactivation is best described by a Boltzmann equation with V 1/2 (the voltage where the probability to find the channels in the inactivated state is 50%) and K (slope factor) equals to -22.9 ± 1.5 mV and 5.3 ± 0.9 mV for control, and -30.3 ± 1.3 mV and 6 ± 0.8 mV for SMD2, respectively. The action of SMD2 is enhanced by high frequency stimulation, and by the time the channel stays open. Taken together, our results suggest that SMD2 blocks the open conformation of KV3.1. From a pharmacological and therapeutic point of view, N-alkylsulfonamides may constitute a new class of pharmacological modulators of KV3.1.


Subject(s)
Potassium Channel Blockers/pharmacology , Shaw Potassium Channels/drug effects , Sulfonamides/pharmacology , Action Potentials/physiology , Animals , Cell Line , Dose-Response Relationship, Drug , Fibroblasts , Mice , Patch-Clamp Techniques , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacokinetics , Shaw Potassium Channels/metabolism , Shaw Potassium Channels/physiology , Sulfonamides/metabolism , Sulfonamides/pharmacokinetics
18.
Pflugers Arch ; 467(8): 1733-46, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25220134

ABSTRACT

Absence seizures are manifestations of abnormal thalamocortical oscillations characterized by spike-and-wave complexes in EEG. Ethosuximide (ETX) is one of the principal medications against absence seizures. We investigate the effect of ETX on the Kir2.1 channel, a prototypical inward rectifier K(+) channel possibly playing an important role in the setting of neuronal membrane potential. We demonstrate that the outward currents of Kir2.1 channels are significantly inhibited by intracellular ETX. We further show that the movement of neutral molecule ETX in the Kir2.1 channel is accompanied by ∼1.2 K(+), giving rise to the vivid voltage dependence of ETX unbinding rate. Moreover, the apparent affinity (K d ) of ETX in the channels are decreased by single-point mutations involving M183, E224, and S165, and especially by double mutations involving T141/S165, which always also disrupt the flux-coupling feature of ETX block. Molecular dynamics simulation demonstrates narrowing of the pore at ∼D172 by binding of ETX to S165 or T141. ETX block of the Kir2.1 channels may cause a modest but critical depolarization of the relevant neurons, decreasing available T-type Ca(2+) channels and consequently lessening pathological thalamocortical burst discharges.


Subject(s)
Anticonvulsants/pharmacology , Ethosuximide/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium/metabolism , Animals , Anticonvulsants/chemistry , Anticonvulsants/metabolism , Binding Sites , Calcium Channels, T-Type/metabolism , Dose-Response Relationship, Drug , Ethosuximide/chemistry , Ethosuximide/metabolism , Kinetics , Membrane Potentials , Molecular Dynamics Simulation , Molecular Structure , Mutagenesis, Site-Directed , Oocytes , Point Mutation , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Protein Binding , Structure-Activity Relationship , Xenopus
19.
Mol Biol Evol ; 31(3): 546-59, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24425781

ABSTRACT

Scorpion K(+) channel toxins and insect defensins share a conserved three-dimensional structure and related biological activities (defense against competitors or invasive microbes by disrupting their membrane functions), which provides an ideal system to study how functional evolution occurs in a conserved structural scaffold. Using an experimental approach, we show that the deletion of a small loop of a parasitoid venom defensin possessing the "scorpion toxin signature" (STS) can remove steric hindrance of peptide-channel interactions and result in a neurotoxin selectively inhibiting K(+) channels with high affinities. This insect defensin-derived toxin adopts a hallmark scorpion toxin fold with a common cysteine-stabilized α-helical and ß-sheet motif, as determined by nuclear magnetic resonance analysis. Mutations of two key residues located in STS completely diminish or significantly decrease the affinity of the toxin on the channels, demonstrating that this toxin binds to K(+) channels in the same manner as scorpion toxins. Taken together, these results provide new structural and functional evidence supporting the predictability of toxin evolution. The experimental strategy is the first employed to establish an evolutionary relationship of two distantly related protein families.


Subject(s)
Defensins/chemistry , Evolution, Molecular , Neurotoxins/chemistry , Neurotoxins/toxicity , Scorpion Venoms/chemistry , Amino Acid Sequence , Animals , Insecta/metabolism , Ion Channel Gating/drug effects , Models, Molecular , Molecular Sequence Data , Neurotoxins/metabolism , Neurotoxins/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Peptides/chemistry , Peptides/metabolism , Phylogeny , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/toxicity , Potassium Channels/chemistry , Potassium Channels/metabolism , Protein Binding/drug effects , Scorpion Venoms/metabolism , Xenopus
20.
Toxicol Appl Pharmacol ; 288(2): 203-12, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26216464

ABSTRACT

The antimalarial drug mefloquine, is known to be a potassium channel blocker, although its mechanism of action has not being elucidated and its effects on the transient outward current (Ito) and the molecular correlate, the Kv4.3 channel has not being studied. Here, we describe the mefloquine-induced inhibition of the rat ventricular Ito and of CHO cells co-transfected with human Kv4.3 and its accessory subunit hKChIP2C by whole-cell voltage-clamp. Mefloquine inhibited rat Ito and hKv4.3+KChIP2C currents in a concentration-dependent manner with a limited voltage dependence and similar potencies (IC50=8.9µM and 10.5µM for cardiac myocytes and Kv4.3 channels, respectively). In addition, mefloquine did not affect the activation of either current but significantly modified the hKv4.3 steady-state inactivation and recovery from inactivation. The effects of this drug was compared with that of 4-aminopyridine (4-AP), a well-known potassium channel blocker and its binding site does not seem to overlap with that of 4-AP.


Subject(s)
Action Potentials/drug effects , Antimalarials/toxicity , Ion Channel Gating/drug effects , Mefloquine/toxicity , Myocytes, Cardiac/drug effects , Potassium Channel Blockers/toxicity , Shal Potassium Channels/antagonists & inhibitors , 4-Aminopyridine/pharmacology , Animals , Antimalarials/metabolism , Binding Sites , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Female , Kv Channel-Interacting Proteins/genetics , Kv Channel-Interacting Proteins/metabolism , Mefloquine/metabolism , Molecular Docking Simulation , Myocytes, Cardiac/metabolism , Potassium/metabolism , Potassium Channel Blockers/metabolism , Protein Binding , Rats, Wistar , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL