Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.111
Filter
1.
Molecules ; 29(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38931004

ABSTRACT

Potassium channels have recently emerged as suitable target for the treatment of epileptic diseases. Among potassium channels, KCNT1 channels are the most widely characterized as responsible for several epileptic and developmental encephalopathies. Nevertheless, the medicinal chemistry of KCNT1 blockers is underdeveloped so far. In the present review, we describe and analyse the papers addressing the issue of KCNT1 blockers' development and identification, also evidencing the pros and the cons of the scientific approaches therein described. After a short introduction describing the epileptic diseases and the structure-function of potassium channels, we provide an extensive overview of the chemotypes described so far as KCNT1 blockers, and the scientific approaches used for their identification.


Subject(s)
Chemistry, Pharmaceutical , Epilepsy , Potassium Channel Blockers , Humans , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/therapeutic use , Potassium Channel Blockers/pharmacology , Chemistry, Pharmaceutical/methods , Epilepsy/drug therapy , Epilepsy/metabolism , Structure-Activity Relationship , Animals , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/metabolism , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Potassium Channels, Voltage-Gated/metabolism , Potassium Channels, Sodium-Activated
2.
Molecules ; 29(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38893312

ABSTRACT

Gain-of-function mutations in the KCNT1 gene, which encodes the sodium-activated potassium channel known as SLACK, are associated with the rare but devastating developmental and epileptic encephalopathy known as epilepsy of infancy with migrating focal seizures (EIMFS). The design of small molecule inhibitors of SLACK channels represents a potential therapeutic approach to the treatment of EIMFS, other childhood epilepsies, and developmental disorders. Herein, we describe a hit optimization effort centered on a xanthine SLACK inhibitor (8) discovered via a high-throughput screen. Across three distinct regions of the chemotype, we synthesized 58 new analogs and tested each one in a whole-cell automated patch-clamp assay to develop structure-activity relationships for inhibition of SLACK channels. We further evaluated selected analogs for their selectivity versus a variety of other ion channels and for their activity versus clinically relevant SLACK mutants. Selectivity within the series was quite good, including versus hERG. Analog 80 (VU0948578) was a potent inhibitor of WT, A934T, and G288S SLACK, with IC50 values between 0.59 and 0.71 µM across these variants. VU0948578 represents a useful in vitro tool compound from a chemotype that is distinct from previously reported small molecule inhibitors of SLACK channels.


Subject(s)
Potassium Channel Blockers , Structure-Activity Relationship , Humans , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels, Sodium-Activated , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Xanthine/chemistry , Xanthine/pharmacology , Patch-Clamp Techniques , HEK293 Cells , Molecular Structure , Xanthines/chemistry , Xanthines/pharmacology
3.
Europace ; 26(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38788213

ABSTRACT

AIMS: Human induced pluripotent stem cell-derived atrial cardiomyocytes (hiPSC-aCM) could be a helpful tool to study the physiology and diseases of the human atrium. To fulfil this expectation, the electrophysiology of hiPSC-aCM should closely resemble the situation in the human atrium. Data on the contribution of the slowly activating delayed rectifier currents (IKs) to repolarization are lacking for both human atrium and hiPSC-aCM. METHODS AND RESULTS: Human atrial tissues were obtained from patients with sinus rhythm (SR) or atrial fibrillation (AF). Currents were measured in human atrial cardiomyocytes (aCM) and compared with hiPSC-aCM and used to model IKs contribution to action potential (AP) shape. Action potential was recorded by sharp microelectrodes. HMR-1556 (1 µM) was used to identify IKs and to estimate IKs contribution to repolarization. Less than 50% of hiPSC-aCM and aCM possessed IKs. Frequency of occurrence, current densities, activation/deactivation kinetics, and voltage dependency of IKs did not differ significantly between hiPSC-aCM and aCM, neither in SR nor AF. ß-Adrenoceptor stimulation with isoprenaline did not increase IKs neither in aCM nor in hiPSC-aCM. In tissue from SR, block of IKs with HMR-1556 did not lengthen the action potential duration, even when repolarization reserve was reduced by block of the ultra-rapid repolarizing current with 4-aminopyridine or the rapidly activating delayed rectifier potassium outward current with E-4031. CONCLUSION: I Ks exists in hiPSC-aCM with biophysics not different from aCM. As in adult human atrium (SR and AF), IKs does not appear to relevantly contribute to repolarization in hiPSC-aCM.


Subject(s)
Action Potentials , Atrial Fibrillation , Delayed Rectifier Potassium Channels , Heart Atria , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Myocytes, Cardiac/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Induced Pluripotent Stem Cells/metabolism , Heart Atria/physiopathology , Delayed Rectifier Potassium Channels/metabolism , Atrial Fibrillation/physiopathology , Atrial Fibrillation/metabolism , Female , Cells, Cultured , Male , Middle Aged , Kinetics , Aged , Cell Differentiation , Models, Cardiovascular , Potassium Channel Blockers/pharmacology
4.
Chem Res Toxicol ; 37(6): 910-922, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38781421

ABSTRACT

The human Ether-à-go-go-Related Gene (hERG) is a transmembrane protein that regulates cardiac action potential, and its inhibition can induce a potentially deadly cardiac syndrome. In vitro tests help identify hERG blockers at early stages; however, the high cost motivates searching for alternative, cost-effective methods. The primary goal of this study was to enhance the Pred-hERG tool for predicting hERG blockage. To achieve this, we developed new QSAR models that incorporated additional data, updated existing classificatory and multiclassificatory models, and introduced new regression models. Notably, we integrated SHAP (SHapley Additive exPlanations) values to offer a visual interpretation of these models. Utilizing the latest data from ChEMBL v30, encompassing over 14,364 compounds with hERG data, our binary and multiclassification models outperformed both the previous iteration of Pred-hERG and all publicly available models. Notably, the new version of our tool introduces a regression model for predicting hERG activity (pIC50). The optimal model demonstrated an R2 of 0.61 and an RMSE of 0.48, surpassing the only available regression model in the literature. Pred-hERG 5.0 now offers users a swift, reliable, and user-friendly platform for the early assessment of chemically induced cardiotoxicity through hERG blockage. The tool provides versatile outcomes, including (i) classificatory predictions of hERG blockage with prediction reliability, (ii) multiclassificatory predictions of hERG blockage with reliability, (iii) regression predictions with estimated pIC50 values, and (iv) probability maps illustrating the contribution of chemical fragments for each prediction. Furthermore, we implemented explainable AI analysis (XAI) to visualize SHAP values, providing insights into the contribution of each feature to binary classification predictions. A consensus prediction calculated based on the predictions of the three developed models is also present to assist the user's decision-making process. Pred-hERG 5.0 has been designed to be user-friendly, making it accessible to users without computational or programming expertise. The tool is freely available at http://predherg.labmol.com.br.


Subject(s)
Ether-A-Go-Go Potassium Channels , Quantitative Structure-Activity Relationship , Humans , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Risk Assessment , Regression Analysis , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry
5.
J Med Chem ; 67(11): 9731-9744, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38807539

ABSTRACT

Recent literature reports highlight the importance of the renal outer medullary potassium (ROMK) channel in renal sodium and potassium homeostasis and emphasize the potential impact that ROMK inhibitors could have as a novel mechanism diuretic in heart failure patients. A series of piperazine-based ROMK inhibitors were designed and optimized to achieve excellent ROMK potency, hERG selectivity, and ADME properties, which led to the identification of compound 28 (BMS-986308). BMS-986308 demonstrated efficacy in the volume-loaded rat diuresis model as well as promising in vitro and in vivo profiles and was therefore advanced to clinical development.


Subject(s)
Heart Failure , Potassium Channel Blockers , Animals , Heart Failure/drug therapy , Humans , Rats , Potassium Channel Blockers/therapeutic use , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacokinetics , Potassium Channel Blockers/chemical synthesis , Structure-Activity Relationship , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/metabolism , Drug Discovery , Diuresis/drug effects , Piperazines/pharmacology , Piperazines/chemistry , Piperazines/therapeutic use , Piperazines/chemical synthesis , Piperazines/pharmacokinetics , Male , Rats, Sprague-Dawley
6.
Mar Drugs ; 22(5)2024 May 13.
Article in English | MEDLINE | ID: mdl-38786608

ABSTRACT

We identified a new human voltage-gated potassium channel blocker, NnK-1, in the jellyfish Nemopilema nomurai based on its genomic information. The gene sequence encoding NnK-1 contains 5408 base pairs, with five introns and six exons. The coding sequence of the NnK-1 precursor is 894 nucleotides long and encodes 297 amino acids containing five presumptive ShK-like peptides. An electrophysiological assay demonstrated that the fifth peptide, NnK-1, which was chemically synthesized, is an effective blocker of hKv1.3, hKv1.4, and hKv1.5. Multiple-sequence alignment with cnidarian Shk-like peptides, which have Kv1.3-blocking activity, revealed that three residues (3Asp, 25Lys, and 34Thr) of NnK-1, together with six cysteine residues, were conserved. Therefore, we hypothesized that these three residues are crucial for the binding of the toxin to voltage-gated potassium channels. This notion was confirmed by an electrophysiological assay with a synthetic peptide (NnK-1 mu) where these three peptides were substituted with 3Glu, 25Arg, and 34Met. In conclusion, we successfully identified and characterized a new voltage-gated potassium channel blocker in jellyfish that interacts with three different voltage-gated potassium channels. A peptide that interacts with multiple voltage-gated potassium channels has many therapeutic applications in various physiological and pathophysiological contexts.


Subject(s)
Peptides , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Scyphozoa , Animals , Humans , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Peptides/pharmacology , Peptides/chemistry , Amino Acid Sequence , Cnidarian Venoms/pharmacology , Cnidarian Venoms/chemistry , Sequence Alignment
7.
Sci Rep ; 14(1): 11105, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38750155

ABSTRACT

4-aminopyridine (4AP) is a potassium (K+) channel blocker used clinically to improve walking in people with multiple sclerosis (MS). 4AP binds to exposed K+ channels in demyelinated axons, reducing the leakage of intracellular K+ and enhancing impulse conduction. Multiple derivatives of 4AP capable of blocking K+ channels have been reported including three radiolabeled with positron emitting isotopes for imaging demyelinated lesions using positron emission tomography (PET). However, there remains a demand for novel molecules with suitable physicochemical properties and binding affinity that can potentially be radiolabeled and used as PET radiotracers. In this study, we introduce 3-fluoro-5-methylpyridin-4-amine (5Me3F4AP) as a novel trisubstituted K+ channel blocker with potential application in PET. 5Me3F4AP has comparable potency to 4AP and the PET tracer 3-fluoro-4-aminopyridine (3F4AP). Compared to 3F4AP, 5Me3F4AP exhibits comparable basicity (pKa = 7.46 ± 0.01 vs. 7.37 ± 0.07, P-value = 0.08), greater lipophilicity (logD = 0.664 ± 0.005 vs. 0.414 ± 0.002, P-value < 0.0001) and higher permeability to an artificial brain membrane (Pe = 88.1 ± 18.3 vs. 31.1 ± 2.9 nm/s, P-value = 0.03). 5Me3F4AP is also more stable towards oxidation in vitro by the cytochrome P450 enzyme CYP2E1 (IC50 = 36.2 ± 2.5 vs. 15.4 ± 5.1, P-value = 0.0003); the enzyme responsible for the metabolism of 4AP and 3F4AP. Taken together, 5Me3F4AP has promising properties as a candidate for PET imaging warranting additional investigation.


Subject(s)
Positron-Emission Tomography , Potassium Channel Blockers , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry , Humans , Positron-Emission Tomography/methods , 4-Aminopyridine/pharmacology , 4-Aminopyridine/chemistry , 4-Aminopyridine/analogs & derivatives , Amifampridine/metabolism
8.
Biomed Pharmacother ; 175: 116651, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692062

ABSTRACT

Voltage-gated potassium channel 1.3 (Kv1.3) has emerged as a pivotal player in numerous biological processes and pathological conditions, sparking considerable interest as a potential therapeutic target across various diseases. In this review, we present a comprehensive examination of Kv1.3 channels, highlighting their fundamental characteristics and recent advancements in utilizing Kv1.3 inhibitors for treating autoimmune disorders, neuroinflammation, and cancers. Notably, Kv1.3 is prominently expressed in immune cells and implicated in immune responses and inflammation associated with autoimmune diseases and chronic inflammatory conditions. Moreover, its aberrant expression in certain tumors underscores its role in cancer progression. While preclinical studies have demonstrated the efficacy of Kv1.3 inhibitors, their clinical translation remains pending. Molecular imaging techniques offer promising avenues for tracking Kv1.3 inhibitors and assessing their therapeutic efficacy, thereby facilitating their development and clinical application. Challenges and future directions in Kv1.3 inhibitor research are also discussed, emphasizing the significant potential of targeting Kv1.3 as a promising therapeutic strategy across a spectrum of diseases.


Subject(s)
Kv1.3 Potassium Channel , Neoplasms , Humans , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/metabolism , Animals , Neoplasms/drug therapy , Neoplasms/metabolism , Potassium Channel Blockers/therapeutic use , Potassium Channel Blockers/pharmacology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/metabolism , Molecular Targeted Therapy , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism
9.
J Med Chem ; 67(11): 9124-9149, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38782404

ABSTRACT

Gain-of-function (GoF) variants in KCNT1 channels cause severe, drug-resistant forms of epilepsy. Quinidine is a known KCNT1 blocker, but its clinical use is limited due to severe drawbacks. To identify novel KCNT1 blockers, a homology model of human KCNT1 was built and used to screen an in-house library of compounds. Among the 20 molecules selected, five (CPK4, 13, 16, 18, and 20) showed strong KCNT1-blocking ability in an in vitro fluorescence-based assay. Patch-clamp experiments confirmed a higher KCNT1-blocking potency of these compounds when compared to quinidine, and their selectivity for KCNT1 over hERG and Kv7.2 channels. Among identified molecules, CPK20 displayed the highest metabolic stability; this compound also blocked KCNT2 currents, although with a lower potency, and counteracted GoF effects prompted by 2 recurrent epilepsy-causing KCNT1 variants (G288S and A934T). The present results provide solid rational basis for future design of novel compounds to counteract KCNT1-related neurological disorders.


Subject(s)
Epilepsy , Humans , Epilepsy/drug therapy , Epilepsy/metabolism , Potassium Channels, Voltage-Gated/metabolism , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemical synthesis , Potassium Channel Blockers/chemistry , Animals , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Structure-Activity Relationship , HEK293 Cells , Computer Simulation , Potassium Channels, Sodium-Activated
10.
J Ethnopharmacol ; 330: 118218, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38677570

ABSTRACT

ETHNOPHARMACOLOGY RELEVANCE: Zanthoxylum bungeanum Maxim. (Z. bungeanum), a member of the Rutaceae family, has a rich history of traditional use in Asia for treating arthritis and toothache conditions. As characteristic chemical components, numerous kinds of alkaloids have been extracted from plants and their diverse biological activities have been reported. However, research on the isoquinoline alkaloid, a specific type of alkaloids, in Z. bungeanum was scarce. AIM OF THE STUDY: The study aimed to isolate a novel isoquinoline alkaloid from Z. bungeanum and explore its pharmacological activity in vitro and analgesic activity in vivo. MATERIALS AND METHODS: Isoquinoline alkaloid isolation and identification from Z. bungeanum were conducted using chromatographic and spectroscopic methods. The whole-cell patch-clamp technique was applied to assess its impact on neuronal excitability, and endogenous voltage-gated potassium (Kv) and sodium (Nav) currents in acutely isolated mouse small-diameter dorsal root ganglion (DRG) neurons. Its inhibitory impacts on channels were further validated with HEK293 cells stably expressing Nav1.7 and Nav1.8, and Chinese hamster ovary (CHO) cells transiently expressing Kv2.1. The formalin inflammatory pain model was utilized to evaluate the potential analgesic activity in vivo. RESULTS: A novel isoquinoline alkaloid named HJ-69 (N-13-(3-methoxyprop-1-yl)rutaecarpine) was isolated and identified from Z. bungeanum for the first time. HJ-69 significantly suppressed the firing frequency and amplitudes of action potentials in DRG neurons. Consistently, it state-dependently inhibited endogenous Nav currents of DRG neurons, with half maximal inhibitory concentration (IC50) values of 13.06 ± 2.06 µM and 30.19 ± 2.07 µM for the inactivated and resting states, respectively. HJ-69 significantly suppressed potassium currents in DRG neurons, which notably inhibited the delayed rectifier potassium (IK) currents (IC50 = 6.95 ± 1.29 µM) and slightly affected the transient outward potassium (IA) currents (IC50 = 523.50 ± 39.16 µM). Furtherly, HJ-69 exhibited similar potencies on heterologously expressed Nav1.7, Nav1.8, and Kv2.1 channels, which correspondingly represent the main components in neurons. Notably, intraperitoneal administration of 30 mg/kg and 100 mg/kg HJ-69 significantly alleviated pain behaviors in the mouse inflammatory pain model induced by formalin. CONCLUSION: The study concluded that HJ-69 is a novel and active isoquinoline alkaloid, and the inhibition of Nav and Kv channels contributes to its analgesic activity. HJ-69 may be a promising prototype for future analgesic drug discovery based on the isoquinoline alkaloid.


Subject(s)
Analgesics , Ganglia, Spinal , Pain , Zanthoxylum , Animals , Zanthoxylum/chemistry , Humans , HEK293 Cells , Analgesics/pharmacology , Analgesics/chemistry , Analgesics/isolation & purification , Analgesics/therapeutic use , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Mice , Male , Pain/drug therapy , Isoquinolines/pharmacology , Isoquinolines/isolation & purification , Isoquinolines/chemistry , Alkaloids/pharmacology , Alkaloids/isolation & purification , Alkaloids/chemistry , Alkaloids/therapeutic use , Potassium Channel Blockers/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Inflammation/drug therapy , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/isolation & purification , Potassium Channels, Voltage-Gated/metabolism , Potassium Channels, Voltage-Gated/drug effects , Neurons/drug effects , Neurons/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/chemistry , Mice, Inbred C57BL , Cricetulus
11.
Eur J Pharmacol ; 973: 176610, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38663541

ABSTRACT

Aripiprazole, a third-generation antipsychotic, has been widely used to treat schizophrenia. In this study, we evaluated the effect of aripiprazole on voltage-gated potassium (Kv) channels in rabbit coronary arterial smooth muscle cells using the patch clamp technique. Aripiprazole reduced the Kv current in a concentration-dependent manner with a half-maximal inhibitory concentration of 0.89 ± 0.20 µM and a Hill coefficient of 1.30 ± 0.25. The inhibitory effect of aripiprazole on Kv channels was voltage-dependent, and an additional aripiprazole-induced decrease in the Kv current was observed in the voltage range of full channel activation. The decay rate of Kv channel inactivation was accelerated by aripiprazole. Aripiprazole shifted the steady-state activation curve to the right and the inactivation curve to the left. Application of a repetitive train of pulses (1 and 2 Hz) promoted inhibition of the Kv current by aripiprazole. Furthermore, the recovery time constant from inactivation increased in the presence of aripiprazole. Pretreatment of Kv1.5 subtype inhibitor reduced the inhibitory effect of aripiprazole. However, pretreatment with Kv 7 and Kv2.1 subtype inhibitors did not change the degree of aripiprazole-induced inhibition of the Kv current. We conclude that aripiprazole inhibits Kv channels in a concentration-, voltage-, time-, and use (state)-dependent manner by affecting the gating properties of the channels.


Subject(s)
Aripiprazole , Coronary Vessels , Myocytes, Smooth Muscle , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Animals , Aripiprazole/pharmacology , Rabbits , Potassium Channels, Voltage-Gated/metabolism , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Coronary Vessels/drug effects , Coronary Vessels/cytology , Potassium Channel Blockers/pharmacology , Male , Antipsychotic Agents/pharmacology , Dose-Response Relationship, Drug
12.
Cardiovasc Res ; 120(7): 735-744, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38442735

ABSTRACT

AIMS: While variants in KCNQ1 are the commonest cause of the congenital long QT syndrome, we and others find only a small IKs in cardiomyocytes from human-induced pluripotent stem cells (iPSC-CMs) or human ventricular myocytes. METHODS AND RESULTS: We studied population control iPSC-CMs and iPSC-CMs from a patient with Jervell and Lange-Nielsen (JLN) syndrome due to compound heterozygous loss-of-function (LOF) KCNQ1 variants. We compared the effects of pharmacologic IKs block to those of genetic KCNQ1 ablation, using JLN cells, cells homozygous for the KCNQ1 LOF allele G643S, or siRNAs reducing KCNQ1 expression. We also studied the effects of two blockers of IKr, the other major cardiac repolarizing current, in the setting of pharmacologic or genetic ablation of KCNQ1: moxifloxacin, associated with a very low risk of drug-induced long QT, and dofetilide, a high-risk drug. In control cells, a small IKs was readily recorded but the pharmacologic IKs block produced no change in action potential duration at 90% repolarization (APD90). In contrast, in cells with genetic ablation of KCNQ1 (JLN), baseline APD90 was markedly prolonged compared with control cells (469 ± 20 vs. 310 ± 16 ms). JLN cells displayed increased sensitivity to acute IKr block: the concentration (µM) of moxifloxacin required to prolong APD90 100 msec was 237.4 [median, interquartile range (IQR) 100.6-391.6, n = 7] in population cells vs. 23.7 (17.3-28.7, n = 11) in JLN cells. In control cells, chronic moxifloxacin exposure (300 µM) mildly prolonged APD90 (10%) and increased IKs, while chronic exposure to dofetilide (5 nM) produced greater prolongation (67%) and no increase in IKs. However, in the siRNA-treated cells, moxifloxacin did not increase IKs and markedly prolonged APD90. CONCLUSION: Our data strongly suggest that KCNQ1 expression modulates baseline cardiac repolarization, and the response to IKr block, through mechanisms beyond simply generating IKs.


Subject(s)
Action Potentials , Induced Pluripotent Stem Cells , Jervell-Lange Nielsen Syndrome , KCNQ1 Potassium Channel , Moxifloxacin , Myocytes, Cardiac , Phenethylamines , Sulfonamides , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Action Potentials/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Moxifloxacin/pharmacology , Phenethylamines/pharmacology , Sulfonamides/pharmacology , Jervell-Lange Nielsen Syndrome/genetics , Jervell-Lange Nielsen Syndrome/metabolism , Jervell-Lange Nielsen Syndrome/physiopathology , Potassium Channel Blockers/pharmacology , Fluoroquinolones/pharmacology
13.
J Biol Chem ; 300(4): 107155, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38479597

ABSTRACT

Despite significant advances in the development of therapeutic interventions targeting autoimmune diseases and chronic inflammatory conditions, lack of effective treatment still poses a high unmet need. Modulating chronically activated T cells through the blockade of the Kv1.3 potassium channel is a promising therapeutic approach; however, developing selective Kv1.3 inhibitors is still an arduous task. Phage display-based high throughput peptide library screening is a rapid and robust approach to develop promising drug candidates; however, it requires solid-phase immobilization of target proteins with their binding site preserved. Historically, the KcsA bacterial channel chimera harboring only the turret region of the human Kv1.3 channel was used for screening campaigns. Nevertheless, literature data suggest that binding to this type of chimera does not correlate well with blocking potency on the native Kv1.3 channels. Therefore, we designed and successfully produced advanced KcsA-Kv1.3, KcsA-Kv1.1, and KcsA-Kv1.2 chimeric proteins in which both the turret and part of the filter regions of the human Kv1.x channels were transferred. These T+F (turret-filter) chimeras showed superior peptide ligand-binding predictivity compared to their T-only versions in novel phage ELISA assays. Phage ELISA binding and competition results supported with electrophysiological data confirmed that the filter region of KcsA-Kv1.x is essential for establishing adequate relative affinity order among selected peptide toxins (Vm24 toxin, Hongotoxin-1, Kaliotoxin-1, Maurotoxin, Stichodactyla toxin) and consequently obtaining more reliable selectivity data. These new findings provide a better screening tool for future drug development efforts and offer insight into the target-ligand interactions of these therapeutically relevant ion channels.


Subject(s)
Kv1.3 Potassium Channel , Potassium Channel Blockers , Recombinant Fusion Proteins , Animals , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/antagonists & inhibitors , Binding Sites , Kv1.3 Potassium Channel/metabolism , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/chemistry , Ligands , Peptide Library , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Potassium Channels/chemistry , Potassium Channels/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Cell Line
14.
Nat Chem Biol ; 20(7): 857-866, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38355723

ABSTRACT

Major depressive disorder, a prevalent and severe psychiatric condition, necessitates development of new and fast-acting antidepressants. Genetic suppression of astrocytic inwardly rectifying potassium channel 4.1 (Kir4.1) in the lateral habenula ameliorates depression-like phenotypes in mice. However, Kir4.1 remains an elusive drug target for depression. Here, we discovered a series of Kir4.1 inhibitors through high-throughput screening. Lys05, the most potent one thus far, effectively suppressed native Kir4.1 channels while displaying high selectivity against established targets for rapid-onset antidepressants. Cryogenic-electron microscopy structures combined with electrophysiological characterizations revealed Lys05 directly binds in the central cavity of Kir4.1. Notably, a single dose of Lys05 reversed the Kir4.1-driven depression-like phenotype and exerted rapid-onset (as early as 1 hour) antidepressant actions in multiple canonical depression rodent models with efficacy comparable to that of (S)-ketamine. Overall, we provided a proof of concept that Kir4.1 is a promising target for rapid-onset antidepressant effects.


Subject(s)
Antidepressive Agents , Potassium Channels, Inwardly Rectifying , Antidepressive Agents/pharmacology , Antidepressive Agents/chemistry , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/metabolism , Animals , Mice , Male , Rats , Humans , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Depression/drug therapy , Depression/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry
15.
Expert Opin Ther Targets ; 28(1-2): 67-82, 2024.
Article in English | MEDLINE | ID: mdl-38316438

ABSTRACT

INTRODUCTION: Kv1.3 is the main voltage-gated potassium channel of leukocytes from both the innate and adaptive immune systems. Channel function is required for common processes such as Ca2+ signaling but also for cell-specific events. In this context, alterations in Kv1.3 are associated with multiple immune disorders. Excessive channel activity correlates with numerous autoimmune diseases, while reduced currents result in increased cancer prevalence and immunodeficiencies. AREAS COVERED: This review offers a general view of the role of Kv1.3 in every type of leukocyte. Moreover, diseases stemming from dysregulations of the channel are detailed, as well as current advances in their therapeutic research. EXPERT OPINION: Kv1.3 arises as a potential immune target in a variety of diseases. Several lines of research focused on channel modulation have yielded positive results. However, among the great variety of specific channel blockers, only one has reached clinical trials. Future investigations should focus on developing simpler administration routes for channel inhibitors to facilitate their entrance into clinical trials. Prospective Kv1.3-based treatments will ensure powerful therapies while minimizing undesired side effects.


Subject(s)
Autoimmune Diseases , Potassium Channels, Voltage-Gated , Humans , Prospective Studies , Potassium Channels, Voltage-Gated/therapeutic use , Autoimmune Diseases/drug therapy , Signal Transduction , Kv1.3 Potassium Channel , Potassium Channel Blockers/pharmacology
16.
Naunyn Schmiedebergs Arch Pharmacol ; 397(7): 5093-5104, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38224347

ABSTRACT

Cavutilide (niferidil, refralon) is a new class III antiarrhythmic drug which effectively terminates persistent atrial fibrillation (AF; 84.6% of patients, mean AF duration 3 months) and demonstrates low risk of torsade de pointes (1.7%). ERG channels of rapid delayed rectifier current(IKr) are the primary target of cavutilide, but the particular reasons of higher effectiveness and lower proarrhythmic risk in comparison with other class III IKr blockers are unclear. The inhibition of hERG channels expressed in CHO-K1 cells by cavutilide was studied using whole-cell patch-clamp. The present study demonstrates high sensitivity of IhERG expressed in CHO-K1 cells to cavutilide (IC50 = 12.8 nM). Similarly to methanesulfonanilide class III agents, but unlike amiodarone and related drugs, cavutilide does not bind to hERG channels in their resting state. However, in contrast to dofetilide, cavutilide binds not only to opened, but also to inactivated channels. Moreover, at positive constantly set membrane potential (+ 60 mV) inhibition of IhERG by 100 nM cavutilide develops faster than at 0 mV and, especially, - 30 mV (τ of inhibition was 78.8, 103, and 153 ms, respectively). Thereby, cavutilide produces IhERG inhibition only when the cell is depolarized. During the same period of time, cavutilide produces greater block of IhERG when the cell is depolarized with 2 Hz frequency, if compared to 0.2 Hz. We suggest that, during the limited time after injection, cavutilide produces stronger inhibition of IKr in fibrillating atrium than in non-fibrillating ventricle. This leads to beneficial combination of antiarrhythmic effectiveness and low proarrhythmicity of cavutilide.


Subject(s)
Anti-Arrhythmia Agents , Cricetulus , Anti-Arrhythmia Agents/pharmacology , CHO Cells , Animals , Humans , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Potassium Channel Blockers/pharmacology , Sulfonamides/pharmacology , Patch-Clamp Techniques , Phenethylamines/pharmacology , Cricetinae , ERG1 Potassium Channel/antagonists & inhibitors , ERG1 Potassium Channel/metabolism
17.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 53(2): 194-200, 2024 Apr 25.
Article in English, Chinese | MEDLINE | ID: mdl-38268403

ABSTRACT

OBJECTIVES: To isolate a potassium ion channel Kv4.1 inhibitor from centipede venom, and to determine its sequence and structure. METHODS: Ion-exchange chromatography and reversed-phase high-performance liquid chromatography were performed to separate and purify peptide components of centipede venom, and their inhibiting effect on Kv4.1 channel was determined by whole-cell patch clamp recording. The molecular weight of isolated peptide Kv4.1 channel inhibitor was identified with matrix assisted laser desorption ionization-time-of-flight mass spectrometry; its primary sequence was determined by Edman degradation sequencing and two-dimensional mass spectrometry; its structure was established based on iterative thread assembly refinement online analysis. RESULTS: A peptide SsTx-P2 was separated from centipede venom with the molecular weight of 6122.8, and its primary sequence consists of 53 amino acid residues NH2-ELTWDFVRTCCKLFPDKSECTKACATEFTGGDESRLKDVWPRKLRSGDSRLKD-OH. Peptide SsTx-P2 potently inhibited the current of Kv4.1 channel transiently transfected in HEK293 cell, with 1.0 µmol/L SsTx-P2 suppressing 95% current of Kv4.1 channel. Its structure showed that SsTx-P2 shared a conserved helical structure. CONCLUSIONS: The study has isolated a novel peptide SsTx-P2 from centipede venom, which can potently inhibit the potassium ion channel Kv4.1 and displays structural conservation.


Subject(s)
Amino Acid Sequence , Arthropod Venoms , Shal Potassium Channels , Animals , Humans , Arthropod Venoms/chemistry , Arthropod Venoms/pharmacology , Molecular Sequence Data , Peptides/pharmacology , Peptides/isolation & purification , Peptides/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/isolation & purification , Potassium Channel Blockers/chemistry , Shal Potassium Channels/antagonists & inhibitors , Chilopoda/chemistry
18.
J Chem Inf Model ; 64(7): 2515-2527, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-37870574

ABSTRACT

In the field of drug discovery, there is a substantial challenge in seeking out chemical structures that possess desirable pharmacological, toxicological, and pharmacokinetic properties. Complications arise when drugs interfere with the functioning of cardiac ion channels, leading to serious cardiovascular consequences. The discontinuation and removal of numerous approved drugs from the market or at late development stages in the pipeline due to such inhibitory effects further highlight the urgency of addressing this issue. Consequently, the early prediction of potential blockers targeting cardiac ion channels during the drug discovery process is of paramount importance. This study introduces a deep learning framework that computationally determines the cardiotoxicity associated with the voltage-gated potassium channel (hERG), the voltage-gated calcium channel (Cav1.2), and the voltage-gated sodium channel (Nav1.5) for drug candidates. The predictive capabilities of three feature representations─molecular fingerprints, descriptors, and graph-based numerical representations─are rigorously benchmarked. Additionally, a novel training and evaluation data set framework is presented, enabling predictive model training of drug off-target cardiotoxicity using a comprehensive and large curated data set covering these three cardiac ion channels. To facilitate these predictions, a robust and comprehensive small molecule cardiotoxicity prediction tool named CToxPred has been developed. It is made available as open source under the permissive MIT license at https://github.com/issararab/CToxPred.


Subject(s)
Cardiotoxicity , Ether-A-Go-Go Potassium Channels , Humans , Benchmarking , Ion Channels , Drug Discovery , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry
19.
FASEB J ; 38(1): e23381, 2024 01.
Article in English | MEDLINE | ID: mdl-38102952

ABSTRACT

Dysfunction of the human voltage-gated K+ channel Kv1.1 has been associated with epilepsy, multiple sclerosis, episodic ataxia, myokymia, and cardiorespiratory dysregulation. We report here that AETX-K, a sea anemone type I (SAK1) peptide toxin we isolated from a phage display library, blocks Kv1.1 with high affinity (Ki ~ 1.6 pM) and notable specificity, inhibiting other Kv channels we tested a million-fold less well. Nuclear magnetic resonance (NMR) was employed both to determine the three-dimensional structure of AETX-K, showing it to employ a classic SAK1 scaffold while exhibiting a unique electrostatic potential surface, and to visualize AETX-K bound to the Kv1.1 pore domain embedded in lipoprotein nanodiscs. Study of Kv1.1 in Xenopus oocytes with AETX-K and point variants using electrophysiology demonstrated the blocking mechanism to employ a toxin-channel configuration we have described before whereby AETX-K Lys23 , two positions away on the toxin interaction surface from the classical blocking residue, enters the pore deeply enough to interact with K+ ions traversing the pathway from the opposite side of the membrane. The mutant channel Kv1.1-L296 F is associated with pharmaco-resistant multifocal epilepsy in infants because it significantly increases K+ currents by facilitating opening and slowing closure of the channels. Consistent with the therapeutic potential of AETX-K for Kv1.1 gain-of-function-associated diseases, AETX-K at 4 pM decreased Kv1.1-L296 F currents to wild-type levels; further, populations of heteromeric channels formed by co-expression Kv1.1 and Kv1.2, as found in many neurons, showed a Ki of ~10 nM even though homomeric Kv1.2 channels were insensitive to the toxin (Ki > 2000 nM).


Subject(s)
Epilepsy , Gain of Function Mutation , Humans , Peptides/genetics , Peptides/pharmacology , Epilepsy/genetics , Potassium Channel Blockers/pharmacology
20.
Int J Mol Sci ; 24(22)2023 Nov 13.
Article in English | MEDLINE | ID: mdl-38003453

ABSTRACT

Modulation of the human Ether-à-go-go-Related Gene (hERG) channel, a crucial voltage-gated potassium channel in the repolarization of action potentials in ventricular myocytes of the heart, has significant implications on cardiac electrophysiology and can be either antiarrhythmic or proarrhythmic. For example, hERG channel blockade is a leading cause of long QT syndrome and potentially life-threatening arrhythmias, such as torsades de pointes. Conversely, hERG channel blockade is the mechanism of action of Class III antiarrhythmic agents in terminating ventricular tachycardia and fibrillation. In recent years, it has been recognized that less proarrhythmic hERG blockers with clinical potential or Class III antiarrhythmic agents exhibit, in addition to their hERG-blocking activity, a second action that facilitates the voltage-dependent activation of the hERG channel. This facilitation is believed to reduce the proarrhythmic potential by supporting the final repolarizing of action potentials. This review covers the pharmacological characteristics of hERG blockers/facilitators, the molecular mechanisms underlying facilitation, and their clinical significance, as well as unresolved issues and requirements for research in the fields of ion channel pharmacology and drug-induced arrhythmias.


Subject(s)
Ether-A-Go-Go Potassium Channels , Potassium Channel Blockers , Humans , ERG1 Potassium Channel , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/therapeutic use , Anti-Arrhythmia Agents/adverse effects , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/drug therapy , Myocytes, Cardiac , Action Potentials
SELECTION OF CITATIONS
SEARCH DETAIL
...