1
|
Del Core L, Mirams GR. Parameter inference for stochastic reaction models of ion channel gating from whole-cell voltage-clamp data. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2025; 383:20240224. [PMID: 40078146 PMCID: PMC11904632 DOI: 10.1098/rsta.2024.0224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/28/2024] [Accepted: 12/01/2024] [Indexed: 03/14/2025]
Abstract
Mathematical models of ion channel gating describe the changes in ion channel configurations due to the electrical activity of the cell membrane. Experimental findings suggest that ion channels behave randomly, and therefore stochastic models of ion channel gating should be more realistic than deterministic counterparts. Whole-cell voltage-clamp data allow us to calibrate the parameters of ion channel models. However, standard methods for deterministic models do not distinguish between stochastic channel gating and measurement error noise, resulting in biased estimates, whereas conventional approaches for stochastic models are computationally demanding. We propose a state-space model of ion channel gating based on stochastic reaction networks, and a maximum likelihood inference procedure to estimate the unknown parameters. Simulation studies show that: (i) our proposed method infers the unknown parameters with low uncertainty and outperforms standard approaches whilst being computationally efficient, and (ii) considering stochastic mechanisms of flickering between conducting and non-conducting open states improves the estimates in the total number of ion channels. Finally, the application of our method to experimental data correctly distinguished the 50-Hz measurement error from noise due to stochastic gating. This method improves data-driven models of ion channel dynamics, by accounting for stochastic gating and measurement errors during inference.This article is part of the theme issue 'Uncertainty quantification for healthcare and biological systems (Part 1)'.
Collapse
Affiliation(s)
- Luca Del Core
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
2
|
Yasuda K, Berenger F, Amaike K, Ueda A, Nakagomi T, Hamasaki G, Li C, Otani NY, Kaitoh K, Tsuda K, Itami K, Yamanishi Y. De novo generation of dual-target compounds using artificial intelligence. iScience 2025; 28:111526. [PMID: 39801837 PMCID: PMC11721219 DOI: 10.1016/j.isci.2024.111526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/06/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Drugs that interact with multiple therapeutic targets are potential high-value products in polypharmacology-based drug discovery, but the rational design remains a formidable challenge. Here, we present artificial intelligence (AI)-based methods to design the chemical structures of compounds that interact with multiple therapeutic target proteins. The molecular structure generation is performed by a fragment-based approach using a genetic algorithm with chemical substructures and a deep learning approach using reinforcement learning with stochastic policy gradients in the framework of generative adversarial networks. Using the proposed methods, we designed the chemical structures of compounds that would interact with two therapeutic targets of bronchial asthma, i.e., adenosine A2a receptor (ADORA2A) and phosphodiesterase 4D (PDE4D). We then synthesized 10 compounds and evaluated their bioactivities via the binding assays of 39 target human proteins, including ADORA2A and PDE4D. Three of the 10 synthesized compounds successfully interacted with ADORA2A and PDE4D with high specificity.
Collapse
Affiliation(s)
- Kasumi Yasuda
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
| | - Francois Berenger
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwa-no-ha, Kashiwa, Chiba 277-8561, Japan
| | - Kazuma Amaike
- Graduate School of Science, Nagoya University, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Ayaka Ueda
- Graduate School of Science, Nagoya University, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Tomoya Nakagomi
- Graduate School of Science, Nagoya University, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Genki Hamasaki
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
| | - Chen Li
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
- Graduate School of Informatics, Nagoya University, Chikusa, Nagoya, Aichi 464-8601, Japan
| | - Noriko Yuyama Otani
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
- Graduate School of Informatics, Nagoya University, Chikusa, Nagoya, Aichi 464-8601, Japan
| | - Kazuma Kaitoh
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
- Graduate School of Informatics, Nagoya University, Chikusa, Nagoya, Aichi 464-8601, Japan
| | - Koji Tsuda
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwa-no-ha, Kashiwa, Chiba 277-8561, Japan
| | - Kenichiro Itami
- Graduate School of Science, Nagoya University, Chikusa, Nagoya, Aichi 464-8602, Japan
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Yoshihiro Yamanishi
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
- Graduate School of Informatics, Nagoya University, Chikusa, Nagoya, Aichi 464-8601, Japan
| |
Collapse
|
3
|
Zhang Y, El Harchi A, James AF, Oiki S, Dempsey CE, Hancox JC. Stereoselective block of the hERG potassium channel by the Class Ia antiarrhythmic drug disopyramide. Cell Mol Life Sci 2024; 81:466. [PMID: 39607488 PMCID: PMC11604869 DOI: 10.1007/s00018-024-05498-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024]
Abstract
Potassium channels encoded by human Ether-à-go-go-Related Gene (hERG) are inhibited by diverse cardiac and non-cardiac drugs. Disopyramide is a chiral Class Ia antiarrhythmic that inhibits hERG at clinical concentrations. This study evaluated effects of disopyramide enantiomers on hERG current (IhERG) from hERG expressing HEK 293 cells at 37 °C. S(+) and R(-) disopyramide inhibited wild-type (WT) IhERG with IC50 values of 3.9 µM and 12.9 µM respectively. The attenuated-inactivation mutant N588K had little effect on the action of S(+) disopyramide but the IC50 for the R(-) enantiomer was ~ 15-fold that for S(+) disopyramide. The enhanced inactivation mutant N588E only slightly increased the potency of R(-) disopyramide. S6 mutation Y652A reduced S(+) disopyramide potency more than that of R(-) disopyramide (respective IC50 values ~ 49-fold and 11-fold their WT controls). The F656A mutation also exerted a stronger effect on S(+) than R(-) disopyramide, albeit with less IC50 elevation. A WT-Y652A tandem dimer exhibited a sensitivity to the enantiomers that was intermediate between that of WT and Y652A, suggesting Y652 groups on adjacent subunits contribute to the binding. Moving the Y (normally at site 652) one residue in the N- terminal (up) direction in N588K hERG markedly increased the blocking potency of R(-) disopyramide. Molecular dynamics simulations using a hERG pore model produced different binding modes for S(+) and R(-) disopyramide consistent with the experimental observations. In conclusion, S(+) disopyramide interacts more strongly with S6 aromatic binding residues on hERG than does R(-) disopyramide, whilst optimal binding of the latter is more reliant on intact inactivation.
Collapse
Affiliation(s)
- Yihong Zhang
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Aziza El Harchi
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Andrew F James
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Shigetoshi Oiki
- Biomedical Imaging Research Centre, University of Fukui, 23-3 Matsuokashimoaizuki, Eiheiji-cho, Fukui, 910-1193, Japan
| | - Christopher E Dempsey
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Jules C Hancox
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
4
|
Johnson AA, Trudeau MC. Inhibition of hERG K channels by verapamil at physiological temperature: Implications for the CiPA initiative. J Pharmacol Toxicol Methods 2024; 130:107562. [PMID: 39332652 DOI: 10.1016/j.vascn.2024.107562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/12/2024] [Accepted: 09/22/2024] [Indexed: 09/29/2024]
Abstract
The Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative reassesses using the inhibition of hERG potassium channels by drugs as the major determinant for the potential to cause drug-induced Torsades de Pointes (TdP) cardiac arrhythmias. Here we report our findings on the next phase of CiPA: Determination of hERG inhibitory properties using the standard CiPA-defined data acquisition protocol, here called the standard protocol, at physiological temperature (37 degrees Celsius). To do this, we measured inhibition of hERG1a potassium channels stably expressed in HEK293 cells by the small molecule verapamil, using manual whole-cell patch-clamp electrophysiology recordings with the standard protocol, which is characterized, in part, by a series of 10 s duration voltage steps to 0 mV, ultimately leading to a cumulative recording time of approximately 30 min. Using the standard protocol, we measured an IC50 for verapamil of 225 nM, a Hill coefficient of 1, and time constant of inhibition at 0 mV of 0.64 s. But, using the standard protocol resulted in a very low (5 %) experimental success rate per cell, which had low practicality for future experiments. To address the 5 % success rate, we generated a revised protocol characterized, in part, by a series of 3 s duration voltage steps to 0 mV, leading to a cumulative recording time of approximately 10 min. Using the revised protocol, we found an IC50 for verapamil of 252 nM, a Hill coefficient of 0.8, and time constant of inhibition at 0 mV of 0.67 s. The values measured with the revised protocol were similar to those measured using the standard protocol and, furthermore, our success rate using the revised protocol rose to 25 %, an increase of 5-fold over the standard protocol, and more in line with the success rate for biophysical studies. In summary, we captured key pharmacological data for subsequent analysis in CiPA using a revised protocol with an increased success rate and an overall enhanced feasibility and practicality. We propose that the revised protocol may be more pragmatic for generation of some hERG channel drug inhibition data for CiPA and other regulatory sciences.
Collapse
Affiliation(s)
- Ashley A Johnson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Matthew C Trudeau
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America.
| |
Collapse
|
5
|
Chang G, Aroge FA, Venkateshappa R, Claydon TW, Sun B. Development of an Absolute Quantification Method for hERG Using PRM with Single Isotopologue in-Sample Calibration. ACS OMEGA 2024; 9:33972-33982. [PMID: 39130540 PMCID: PMC11308013 DOI: 10.1021/acsomega.4c04541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 08/13/2024]
Abstract
The human ether-à-go-go-related gene (KCNH2)-encoded protein hERG constitutes the α subunit of the Kv11.1 channel and contributes to the I kr current, which plays an important role in the cardiac action potential. Genetically and xenobiotically triggered malfunctions of hERG can cause arrhythmia. The expression of hERG in various study systems was assessed mainly as the fold change relative to the corresponding control. Here, we developed a simple and sensitive quantitation method using targeted mass spectrometry, i.e., the parallel reaction monitoring approach, to measure the absolute quantity of hERG in copy number. Such measurements do not require controls, and the obtained values can be compared with similar results for any other protein. To effectively avoid matrix effects, we used the heavy-match-light (HML) in-sample calibration approach that requires only a single isotopologue to achieve copy-number quantitation. No significant difference was observed in the results obtained by HML and by the classic standard addition in-sample calibration approach. Using four proteotypic peptides, we quantified the average number of copies of hERG in the HEK293T heterologous expression system as 3.6 ± 0.5 × 106 copies/cell, i.e., 1 million copies/cell for the fully assembled Kv11.1 channel.
Collapse
Affiliation(s)
- Ge Chang
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A1S6, Canada
| | - Fabusuyi A. Aroge
- School
of Mechatronic Systems Engineering, Simon
Fraser University, Surrey, British Columbia V3T0A3, Canada
| | - Ravichandra Venkateshappa
- Department
of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A1S6, Canada
| | - Tom W. Claydon
- Department
of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A1S6, Canada
| | - Bingyun Sun
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A1S6, Canada
| |
Collapse
|
6
|
Davis J, Cornwell JD, Campagna N, Guo J, Li W, Yang T, Wang T, Zhang S. Rescue of expression and function of long QT syndrome-causing mutant hERG channels by enhancing channel stability in the plasma membrane. J Biol Chem 2024; 300:107526. [PMID: 38960041 PMCID: PMC11325228 DOI: 10.1016/j.jbc.2024.107526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024] Open
Abstract
The human ether-a-go-go-related gene (hERG) encodes the Kv11.1 (or hERG) channel that conducts the rapidly activating delayed rectifier potassium current (IKr). Naturally occurring mutations in hERG impair the channel function and cause long QT syndrome type 2. Many missense hERG mutations lead to a lack of channel expression on the cell surface, representing a major mechanism for the loss-of-function of mutant channels. While it is generally thought that a trafficking defect underlies the lack of channel expression on the cell surface, in the present study, we demonstrate that the trafficking defective mutant hERG G601S can reach the plasma membrane but is unstable and quickly degrades, which is akin to WT hERG channels under low K+ conditions. We previously showed that serine (S) residue at 624 in the innermost position of the selectivity filter of hERG is involved in hERG membrane stability such that substitution of serine 624 with threonine (S624T) enhances hERG stability and renders hERG insensitive to low K+ culture. Here, we report that the intragenic addition of S624T substitution to trafficking defective hERG mutants G601S, N470D, and P596R led to a complete rescue of the function of these otherwise loss-of-function mutant channels to a level similar to the WT channel, representing the most effective rescue means for the function of mutant hERG channels. These findings not only provide novel insights into hERG mutation-mediated channel dysfunction but also point to the critical role of S624 in hERG stability on the plasma membrane.
Collapse
Affiliation(s)
- Jordan Davis
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - James D Cornwell
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Noah Campagna
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jun Guo
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Wentao Li
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Tonghua Yang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Tingzhong Wang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Shetuan Zhang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
7
|
Stevens-Sostre WA, Flores-Aldama L, Bustos D, Li J, Morais-Cabral JH, Delemotte L, Robertson GA. An intracellular hydrophobic nexus critical for hERG1 channel slow deactivation. Biophys J 2024; 123:2024-2037. [PMID: 38219015 PMCID: PMC11309987 DOI: 10.1016/j.bpj.2024.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/17/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024] Open
Abstract
Slow deactivation is a critical property of voltage-gated K+ channels encoded by the human Ether-à-go-go-Related Gene 1 (hERG). hERG1 channel deactivation is modulated by interactions between intracellular N-terminal Per-Arnt-Sim (PAS) and C-terminal cyclic nucleotide-binding homology (CNBh) domains. The PAS domain is multipartite, comprising a globular domain (gPAS; residues 26-135) and an N-terminal PAS-cap that is further subdivided into an initial unstructured "tip" (residues 1-12) and an amphipathic α-helical region (residues 13-25). Although the PAS-cap tip has long been considered the effector of slow deactivation, how its position near the gating machinery is controlled has not been elucidated. Here, we show that a triad of hydrophobic interactions among the gPAS, PAS-cap α helix, and the CNBh domains is required to support slow deactivation in hERG1. The primary sequence of this "hydrophobic nexus" is highly conserved among mammalian ERG channels but shows key differences to fast-deactivating Ether-à-go-go 1 (EAG1) channels. Combining sequence analysis, structure-directed mutagenesis, electrophysiology, and molecular dynamics simulations, we demonstrate that polar serine substitutions uncover an intermediate deactivation mode that is also mimicked by deletion of the PAS-cap α helix. Molecular dynamics simulation analyses of the serine-substituted channels show an increase in distance among the residues of the hydrophobic nexus, a rotation of the intracellular gating ring, and a retraction of the PAS-cap tip from its receptor site near the voltage sensor domain and channel gate. These findings provide compelling evidence that the hydrophobic nexus coordinates the respective components of the intracellular gating ring and positions the PAS-cap tip to control hERG1 deactivation gating.
Collapse
Affiliation(s)
- Whitney A Stevens-Sostre
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Lisandra Flores-Aldama
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Daniel Bustos
- Centro de Investigación de Estudios Avanzados Del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica Del Maule, Talca, Chile; Laboratorio de Bioinformática y Química Computacional (LBQC), Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica Del Maule, Talca, Chile
| | - Jin Li
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - João H Morais-Cabral
- Instituto de Investigação e Inovação Em Saude da Universidade Do Porto (i3S); Instituto de Biologia Molecular e Celular, Universidade Do Porto, Porto, Portugal
| | - Lucie Delemotte
- KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
| | - Gail A Robertson
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
8
|
Anees P, Saminathan A, Rozmus ER, Di A, Malik AB, Delisle BP, Krishnan Y. Detecting organelle-specific activity of potassium channels with a DNA nanodevice. Nat Biotechnol 2024; 42:1065-1074. [PMID: 37735264 PMCID: PMC11021130 DOI: 10.1038/s41587-023-01928-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 07/31/2023] [Indexed: 09/23/2023]
Abstract
Cell surface potassium ion (K+) channels regulate nutrient transport, cell migration and intercellular communication by controlling K+ permeability and are thought to be active only at the plasma membrane. Although these channels transit the trans-Golgi network, early and recycling endosomes, whether they are active in these organelles is unknown. Here we describe a pH-correctable, ratiometric reporter for K+ called pHlicKer, use it to probe the compartment-specific activity of a prototypical voltage-gated K+ channel, Kv11.1, and show that this cell surface channel is active in organelles. Lumenal K+ in organelles increased in cells expressing wild-type Kv11.1 channels but not after treatment with current blockers. Mutant Kv11.1 channels, with impaired transport function, failed to increase K+ levels in recycling endosomes, an effect rescued by pharmacological correction. By providing a way to map the organelle-specific activity of K+ channels, pHlicKer technology could help identify new organellar K+ channels or channel modulators with nuanced functions.
Collapse
Affiliation(s)
- Palapuravan Anees
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
- Grossman Center for Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, USA
- Institute of Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
| | - Anand Saminathan
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
- Grossman Center for Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, USA
| | - Ezekiel R Rozmus
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Anke Di
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of Medicine, Chicago, IL, USA
| | - Asrar B Malik
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of Medicine, Chicago, IL, USA
| | - Brian P Delisle
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA.
| | - Yamuna Krishnan
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.
- Grossman Center for Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, USA.
- Institute of Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
9
|
Abramochkin DV, Pustovit OB, Mironov NY, Filatova TS, Nesterova T. Characterization of hERG K + channel inhibition by the new class III antiarrhythmic drug cavutilide. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5093-5104. [PMID: 38224347 DOI: 10.1007/s00210-023-02940-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/30/2023] [Indexed: 01/16/2024]
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.
Collapse
Affiliation(s)
- Denis V Abramochkin
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia.
| | - Oksana B Pustovit
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia
| | | | - Tatiana S Filatova
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia
- Chazov National Medical Research Center for Cardiology, Moscow, Russia
- Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova str., 1, Moscow, Russia
| | - Tatiana Nesterova
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, 620049, Ekaterinburg, Russia
- Institute of Natural Sciences and Mathematics, Ural Federal University, 620075, Ekaterinburg, Russia
| |
Collapse
|
10
|
Sawada R, Sakajiri Y, Shibata T, Yamanishi Y. Predicting therapeutic and side effects from drug binding affinities to human proteome structures. iScience 2024; 27:110032. [PMID: 38868195 PMCID: PMC11167438 DOI: 10.1016/j.isci.2024.110032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/08/2024] [Accepted: 05/16/2024] [Indexed: 06/14/2024] Open
Abstract
Evaluation of the binding affinities of drugs to proteins is a crucial process for identifying drug pharmacological actions, but it requires three dimensional structures of proteins. Herein, we propose novel computational methods to predict the therapeutic indications and side effects of drug candidate compounds from the binding affinities to human protein structures on a proteome-wide scale. Large-scale docking simulations were performed for 7,582 drugs with 19,135 protein structures revealed by AlphaFold (including experimentally unresolved proteins), and machine learning models on the proteome-wide binding affinity score (PBAS) profiles were constructed. We demonstrated the usefulness of the method for predicting the therapeutic indications for 559 diseases and side effects for 285 toxicities. The method enabled to predict drug indications for which the related protein structures had not been experimentally determined and to successfully extract proteins eliciting the side effects. The proposed method will be useful in various applications in drug discovery.
Collapse
Affiliation(s)
- Ryusuke Sawada
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka, Japan
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuko Sakajiri
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka, Japan
- Graduate School of Informatics, Nagoya University, Chikusa, Nagoya, Japan
| | - Tomokazu Shibata
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka, Japan
| | - Yoshihiro Yamanishi
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka, Japan
- Graduate School of Informatics, Nagoya University, Chikusa, Nagoya, Japan
| |
Collapse
|
11
|
Ihsan MF, Kawashima D, Li S, Ogasawara S, Murata T, Takei M. Non-invasive hERG channel screening based on electrical impedance tomography and extracellular voltage activation (EIT-EVA). LAB ON A CHIP 2024; 24:3183-3190. [PMID: 38828904 DOI: 10.1039/d4lc00230j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
hERG channel screening has been achieved based on electrical impedance tomography and extracellular voltage activation (EIT-EVA) to improve the non-invasive aspect of drug discovery. EIT-EVA screens hERG channels by considering the change in extracellular ion concentration which modifies the extracellular resistance in cell suspension. The rate of ion passing in cell suspension is calculated from the extracellular resistance Rex, which is obtained from the EIT measurement at a frequency of 500 kHz. In the experiment, non-invasive screening is applied by a novel integrated EIT-EVA printed circuit board (PCB) sensor to human embryonic kidney (HEK) 293 cells transfected with the human ether-a-go-go-related gene (hERG) ion channel, while the E-4031 antiarrhythmic drug is used for hERG channel inhibition. The extracellular resistance Rex of the HEK 293 cells suspension is measured by EIT as the hERG channels are activated by EVA over time. The Rex is reconstructed into extracellular conductivity distribution change Δσ to reflect the extracellular K+ ion concentration change Δc resulting from the activated hERG channel. Δc is increased rapidly during the hERG channel non-inhibition state while Δc is increased slower with increasing drug concentration cd. In order to evaluate the EIT-EVA system, the inhibitory ratio index (IR) was calculated based on the rate of Δc over time. Half-maximal inhibitory concentration (IC50) of 2.7 nM is obtained from the cd and IR dose-response relationship. The IR from EIT-EVA is compared with the results from the patch-clamp method, which gives R2 of 0.85. In conclusion, EIT-EVA is successfully applied to non-invasive hERG channel screening.
Collapse
Affiliation(s)
- Muhammad Fathul Ihsan
- Department of Mechanical Engineering, Graduate School of Science and Engineering, Division of Fundamental Engineering, Chiba University, Chiba 263-8522, Japan
| | - Daisuke Kawashima
- Graduate School of Engineering, Chiba University, Chiba 263-8522, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba 263-8522, Japan.
| | - Songshi Li
- Graduate School of Engineering, Chiba University, Chiba 263-8522, Japan
| | - Satoshi Ogasawara
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan
- Molecular Chirality Research Center, Chiba University, Chiba 263-8522, Japan
| | - Takeshi Murata
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan
- Molecular Chirality Research Center, Chiba University, Chiba 263-8522, Japan
| | - Masahiro Takei
- Graduate School of Engineering, Chiba University, Chiba 263-8522, Japan
| |
Collapse
|
12
|
Kadsanit N, Worsawat P, Sakonsinsiri C, McElroy CR, Macquarrie D, Noppawan P, Hunt AJ. Sustainable methods for the carboxymethylation and methylation of ursolic acid with dimethyl carbonate under mild and acidic conditions. RSC Adv 2024; 14:16921-16934. [PMID: 38799212 PMCID: PMC11124730 DOI: 10.1039/d4ra02122c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
Ursolic acid is a triterpene plant extract that exhibits significant potential as an anti-cancer, anti-tumour, and anti-inflammatory agent. Its direct use in the pharmaceutical industry is hampered by poor uptake of ursolic acid in the human body coupled with rapid metabolism causing a decrease in bioactivity. Modification of ursolic acid can overcome such issues, however, use of toxic reagents, unsustainable synthetic routes and poor reaction metrics have limited its potential. Herein, we demonstrate the first reported carboxymethylation and/or methylation of ursolic acid with dimethyl carbonate (DMC) as a green solvent and sustainable reagent under acidic conditions. The reaction of DMC with ursolic acid, in the presence of PTSA, ZnCl2, or H2SO4-SiO2 yielded the carboxymethylation product 3β-[[methoxy]carbonyl]oxyurs-12-en-28-oic acid, the methylation product 3β-methoxyurs-12-en-28-oic acid and the dehydration product urs-2,12-dien-28-oic acid. PTSA demonstrated high conversion and selectivity towards the previously unreported carboxymethylation of ursolic acid, while the application of formic acid in the system led to formylation of ursolic acid (3β-formylurs-12-en-28-oic acid) in quantitative yields via esterification, with DMC acting solely as a solvent. Meanwhile, the methylation product of ursolic acid, 3β-methoxyurs-12-en-28-oic acid, was successfully synthesised with FeCl3, demonstrating exceptional conversion and selectivity, >99% and 99%, respectively. Confirmed with the use of qualitative and quantitative green metrics, this result represents a significant improvement in conversion, selectivity, safety, and sustainability over previously reported methods of ursolic acid modification. It was demonstrated that these methods could be applied to other triterpenoids, including corosolic acid. The study also explored the potential pharmaceutical applications of ursolic acid, corosolic acid, and their derivatives, particularly in anti-inflammatory, anti-cancer, and anti-tumour treatments, using molecular ADMET and docking methods. The methods developed in this work have led to the synthesis of novel molecules, thus creating opportunities for the future investigation of biological activity and the modification of a wide range of triterpenoids applying acidic DMC systems to deliver novel active pharmaceutical intermediates.
Collapse
Affiliation(s)
- Nuttapong Kadsanit
- Materials Chemistry Research Center (MCRC), Department of Chemistry and Centre of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University Khon Kaen 40002 Thailand
| | - Pattamabhorn Worsawat
- Materials Chemistry Research Center (MCRC), Department of Chemistry and Centre of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University Khon Kaen 40002 Thailand
| | - Chadamas Sakonsinsiri
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University Khon Kaen 40002 Thailand
| | - Con R McElroy
- School of Chemistry, University of Lincoln Brayford Pool Campus Lincoln LN6 7TS UK
- Green Chemistry Centre of Excellence, Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Duncan Macquarrie
- Green Chemistry Centre of Excellence, Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Pakin Noppawan
- Department of Chemistry, Faculty of Science, Mahasarakham University Maha Sarakham 44150 Thailand
| | - Andrew J Hunt
- Materials Chemistry Research Center (MCRC), Department of Chemistry and Centre of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University Khon Kaen 40002 Thailand
| |
Collapse
|
13
|
Hossain MM, Khalid A, Akhter Z, Parveen S, Ayaz MO, Bhat AQ, Badesra N, Showket F, Dar MS, Ahmed F, Dhiman S, Kumar M, Singh U, Hussain R, Keshari P, Mustafa G, Nargorta A, Taneja N, Gupta S, Mir RA, Kshatri AS, Nandi U, Khan N, Ramajayan P, Yadav G, Ahmed Z, Singh PP, Dar MJ. Discovery of a novel and highly selective JAK3 inhibitor as a potent hair growth promoter. J Transl Med 2024; 22:370. [PMID: 38637842 PMCID: PMC11025159 DOI: 10.1186/s12967-024-05144-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/23/2024] [Indexed: 04/20/2024] Open
Abstract
JAK-STAT signalling pathway inhibitors have emerged as promising therapeutic agents for the treatment of hair loss. Among different JAK isoforms, JAK3 has become an ideal target for drug discovery because it only regulates a narrow spectrum of γc cytokines. Here, we report the discovery of MJ04, a novel and highly selective 3-pyrimidinylazaindole based JAK3 inhibitor, as a potential hair growth promoter with an IC50 of 2.03 nM. During in vivo efficacy assays, topical application of MJ04 on DHT-challenged AGA and athymic nude mice resulted in early onset of hair regrowth. Furthermore, MJ04 significantly promoted the growth of human hair follicles under ex-vivo conditions. MJ04 exhibited a reasonably good pharmacokinetic profile and demonstrated a favourable safety profile under in vivo and in vitro conditions. Taken together, we report MJ04 as a highly potent and selective JAK3 inhibitor that exhibits overall properties suitable for topical drug development and advancement to human clinical trials.
Collapse
Affiliation(s)
- Md Mehedi Hossain
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Arfan Khalid
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Zaheen Akhter
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Sabra Parveen
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Mir Owais Ayaz
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Aadil Qadir Bhat
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Neetu Badesra
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Farheen Showket
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Mohmmad Saleem Dar
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Farhan Ahmed
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute (CDRI), Lucknow, 226031, India
| | - Sumit Dhiman
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Mukesh Kumar
- Medicinal Product Chemistry, Sussex Drug Discovery Centre, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG, UK
| | - Umed Singh
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Razak Hussain
- Department of Entomology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Pankaj Keshari
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Ghulam Mustafa
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Amit Nargorta
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Neha Taneja
- Department of Dermatology and Venereology, All India Institute of Medical Sciences, New Delhi, India
| | - Somesh Gupta
- Department of Dermatology and Venereology, All India Institute of Medical Sciences, New Delhi, India
| | - Riyaz A Mir
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Aravind Singh Kshatri
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute (CDRI), Lucknow, 226031, India
| | - Utpal Nandi
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Nooruddin Khan
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, 500046, India
| | - P Ramajayan
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Govind Yadav
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Zabeer Ahmed
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Parvinder Pal Singh
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India.
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India.
| | - Mohd Jamal Dar
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India.
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India.
| |
Collapse
|
14
|
Wang Y, Shu J, Yang H, Hong K, Yang X, Guo W, Fang J, Li F, Liu T, Shan Z, Shi T, Cai S, Zhang J. Nav1.7 Modulator Bearing a 3-Hydroxyindole Backbone Holds the Potential to Reverse Neuropathic Pain. ACS Chem Neurosci 2024; 15:1063-1073. [PMID: 38449097 DOI: 10.1021/acschemneuro.3c00353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024] Open
Abstract
Chronic pain is a growing global health problem affecting at least 10% of the world's population. However, current chronic pain treatments are inadequate. Voltage-gated sodium channels (Navs) play a pivotal role in regulating neuronal excitability and pain signal transmission and thus are main targets for nonopioid painkiller development, especially those preferentially expressed in dorsal root ganglial (DRG) neurons, such as Nav1.6, Nav1.7, and Nav1.8. In this study, we screened in virtual hits from dihydrobenzofuran and 3-hydroxyoxindole hybrid molecules against Navs via a veratridine (VTD)-based calcium imaging method. The results showed that one of the molecules, 3g, could inhibit VTD-induced neuronal activity significantly. Voltage clamp recordings demonstrated that 3g inhibited the total Na+ currents of DRG neurons in a concentration-dependent manner. Biophysical analysis revealed that 3g slowed the activation, meanwhile enhancing the inactivation of the Navs. Additionally, 3g use-dependently blocked Na+ currents. By combining with selective Nav inhibitors and a heterozygous expression system, we demonstrated that 3g preferentially inhibited the TTX-S Na+ currents, specifically the Nav1.7 current, other than the TTX-R Na+ currents. Molecular docking experiments implicated that 3g binds to a known allosteric site at the voltage-sensing domain IV(VSDIV) of Nav1.7. Finally, intrathecal injection of 3g significantly relieved mechanical pain behavior in the spared nerve injury (SNI) rat model, suggesting that 3g is a promising candidate for treating chronic pain.
Collapse
Affiliation(s)
- Yuwei Wang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Jirong Shu
- Guangdong Chiral Drug Engineering Laboratory, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510000, China
| | - Haoyi Yang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Kemiao Hong
- Guangdong Chiral Drug Engineering Laboratory, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510000, China
| | - Xiangji Yang
- Guangdong Chiral Drug Engineering Laboratory, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510000, China
| | - Weijie Guo
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Jie Fang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Fuyi Li
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Tao Liu
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Zhiming Shan
- Department of Anesthesiology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen 518020, China
- Laboratory and Clinical Research Institute for Pain, Department of Anaesthesiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Taoda Shi
- Guangdong Chiral Drug Engineering Laboratory, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510000, China
| | - Song Cai
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Jian Zhang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
15
|
Abramochkin D, Li B, Zhang H, Kravchuk E, Nesterova T, Glukhov G, Shestak A, Zaklyazminskaya E, Sokolova OS. Novel Gain-of-Function Mutation in the Kv11.1 Channel Found in the Patient with Brugada Syndrome and Mild QTc Shortening. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:543-552. [PMID: 38648771 DOI: 10.1134/s000629792403012x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 04/25/2024]
Abstract
Brugada syndrome (BrS) is an inherited disease characterized by right precordial ST-segment elevation in the right precordial leads on electrocardiograms (ECG), and high risk of life-threatening ventricular arrhythmia and sudden cardiac death (SCD). Mutations in the responsible genes have not been fully characterized in the BrS patients, except for the SCN5A gene. We identified a new genetic variant, c.1189C>T (p.R397C), in the KCNH2 gene in the asymptomatic male proband diagnosed with BrS and mild QTc shortening. We hypothesize that this variant could alter IKr-current and may be causative for the rare non-SCN5A-related form of BrS. To assess its pathogenicity, we performed patch-clamp analysis on IKr reconstituted with this KCNH2 mutation in the Chinese hamster ovary cells and compared the phenotype with the wild type. It appeared that the R397C mutation does not affect the IKr density, but facilitates activation, hampers inactivation of the hERG channels, and increases magnitude of the window current suggesting that the p.R397C is a gain-of-function mutation. In silico modeling demonstrated that this missense mutation potentially leads to the shortening of action potential in the heart.
Collapse
Affiliation(s)
- Denis Abramochkin
- Shenzhen MSU-BIT University, Shenzhen, China.
- Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Bowen Li
- Shenzhen MSU-BIT University, Shenzhen, China.
| | - Han Zhang
- Shenzhen MSU-BIT University, Shenzhen, China.
| | | | - Tatiana Nesterova
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Ekaterinburg, 620049, Russia.
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg, 620075, Russia
| | - Grigory Glukhov
- Shenzhen MSU-BIT University, Shenzhen, China.
- Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Anna Shestak
- Petrovsky National Research Center of Surgery, Moscow, 119991, Russia.
| | | | - Olga S Sokolova
- Shenzhen MSU-BIT University, Shenzhen, China.
- Lomonosov Moscow State University, 119234, Moscow, Russia
| |
Collapse
|
16
|
Butler AS, Ascione R, Marrion NV, Harmer SC, Hancox JC. In situ monolayer patch clamp of acutely stimulated human iPSC-derived cardiomyocytes promotes consistent electrophysiological responses to SK channel inhibition. Sci Rep 2024; 14:3185. [PMID: 38326449 PMCID: PMC10850090 DOI: 10.1038/s41598-024-53571-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 02/02/2024] [Indexed: 02/09/2024] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) represent an in vitro model of cardiac function. Isolated iPSC-CMs, however, exhibit electrophysiological heterogeneity which hinders their utility in the study of certain cardiac currents. In the healthy adult heart, the current mediated by small conductance, calcium-activated potassium (SK) channels (ISK) is atrial-selective. Functional expression of ISK within atrial-like iPSC-CMs has not been explored thoroughly. The present study therefore aimed to investigate atrial-like iPSC-CMs as a model system for the study of ISK. iPSCs were differentiated using retinoic acid (RA) to produce iPSC-CMs which exhibited an atrial-like phenotype (RA-iPSC-CMs). Only 18% of isolated RA-iPSC-CMs responded to SK channel inhibition by UCL1684 and isolated iPSC-CMs exhibited substantial cell-to-cell electrophysiological heterogeneity. This variability was significantly reduced by patch clamp of RA-iPSC-CMs in situ as a monolayer (iPSC-ML). A novel method of electrical stimulation was developed to facilitate recording from iPSC-MLs via In situ Monolayer Patch clamp of Acutely Stimulated iPSC-CMs (IMPASC). Using IMPASC, > 95% of iPSC-MLs could be paced at a 1 Hz. In contrast to isolated RA-iPSC-CMs, 100% of RA-iPSC-MLs responded to UCL1684, with APD50 being prolonged by 16.0 ± 2.0 ms (p < 0.0001; n = 12). These data demonstrate that in conjunction with IMPASC, RA-iPSC-MLs represent an improved model for the study of ISK. IMPASC may be of wider value in the study of other ion channels that are inconsistently expressed in isolated iPSC-CMs and in pharmacological studies.
Collapse
Affiliation(s)
- Andrew S Butler
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Raimondo Ascione
- Bristol Heart Institute and Translational Biomedical Research Centre, Faculty of Health Science, University of Bristol, Bristol, BS2 8HW, UK
| | - Neil V Marrion
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Stephen C Harmer
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK.
| | - Jules C Hancox
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
17
|
Hancox JC, Copeland CS, Harmer SC, Henderson G. New synthetic cannabinoids and the potential for cardiac arrhythmia risk. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 6:100049. [PMID: 38143960 PMCID: PMC10739592 DOI: 10.1016/j.jmccpl.2023.100049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 12/26/2023]
Abstract
Synthetic cannabinoid receptor agonists (SCRAs) have been associated with QT interval prolongation. Limited preclinical information on SCRA effects on cardiac electrogenesis results from the rapid emergence of new compounds and restricted research availability. We used two machine-learning-based tools to evaluate seven novel SCRAs' interaction potential with the hERG potassium channel, an important drug antitarget. Five SCRAs were predicted to have the ability to block the hERG channel by both prediction tools; ADB-FUBIATA was predicted to be a strong hERG blocker. ADB-5Br-INACA and ADB-4en-PINACA showed varied predictions. These findings highlight potentially proarrhythmic hERG block by novel SCRAs, necessitating detailed safety evaluations.
Collapse
Affiliation(s)
- Jules C. Hancox
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Caroline S. Copeland
- Institute of Pharmaceutical Science, King's College London, UK
- Centre for Pharmaceutical Medicine Research, King's College London, UK
| | - Stephen C. Harmer
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Graeme Henderson
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| |
Collapse
|
18
|
Montalbano A, Sala C, Altadonna GC, Becchetti A, Arcangeli A. High throughput clone screening on overexpressed hERG1 and Kv1.3 potassium channels using ion channel reader (ICR) label free technology. Heliyon 2023; 9:e20112. [PMID: 37767500 PMCID: PMC10520782 DOI: 10.1016/j.heliyon.2023.e20112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/27/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Pharmacological studies aimed at the development of newly synthesized drugs directed against ion channels (as well as genetic studies of ion channel mutations) involve the development and use of transfected cells. However, the identification of the best clone, in terms of transfection efficiency, is often a time consuming procedure when performed through traditional methods such as manual patch-clamp. On the other hand, the use of other faster techniques, such as for example the IF, are not informative on the effective biological functionality of the transfected ion channel(s). In the present work, we used the high throughput automated ion channel reader (ICR) technology (ICR8000 Aurora Biomed Inc.) that combine atomic absorption spectroscopy with a patented microsampling process to accurately measure ion flux in cell-based screening assays. This technology indeed helped us to evaluate the transfection efficiency of hERG1 and hKv1.3 channels respectively on the HEK-293 and CHO cellular models. Moreover, as proof of the validity of this innovative method, we have corroborated these data with the functional characterization of the potassium currents carried out by the same clones through patch-clamp recordings. The results obtained in our study are promising and represent a valid methodological strategy to screen a large number of clones simultaneously and to pharmacologically evaluate their functionality within an extremely faster timeframe.
Collapse
Affiliation(s)
- Alberto Montalbano
- Department of Experimental and Clinical Medicine, University of Florence, I-50134, Florence, Italy
| | - Cesare Sala
- Department of Experimental and Clinical Medicine, University of Florence, I-50134, Florence, Italy
| | | | - Andrea Becchetti
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, I-20126, Milano, Italy
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence, I-50134, Florence, Italy
| |
Collapse
|
19
|
Campagna N, Wall E, Lee K, Guo J, Li W, Yang T, Baranchuk A, El-Diasty M, Zhang S. Differential Effects of Remdesivir and Lumacaftor on Homomeric and Heteromeric hERG Channels. Mol Pharmacol 2023; 104:164-173. [PMID: 37419691 DOI: 10.1124/molpharm.123.000708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/31/2023] [Accepted: 06/08/2023] [Indexed: 07/09/2023] Open
Abstract
The human ether-a-go-go-related gene (hERG) encodes for the pore-forming subunit of the channel that conducts the rapidly activating delayed K+ current (IKr) in the heart. The hERG channel is important for cardiac repolarization, and reduction of its expression in the plasma membrane due to mutations causes long QT syndrome type 2 (LQT2). As such, promoting hERG membrane expression is a strategy to rescue mutant channel function. In the present study, we applied patch clamp, western blots, immunocytochemistry, and quantitative reverse transcription polymerase chain reaction techniques to investigate the rescue effects of two drugs, remdesivir and lumacaftor, on trafficking-defective mutant hERG channels. As our group has recently reported that the antiviral drug remdesivir increases wild-type (WT) hERG current and surface expression, we studied the effects of remdesivir on trafficking-defective LQT2-causing hERG mutants G601S and R582C expressed in HEK293 cells. We also investigated the effects of lumacaftor, a drug used to treat cystic fibrosis, that promotes CFTR protein trafficking and has been shown to rescue membrane expression of some hERG mutations. Our results show that neither remdesivir nor lumacaftor rescued the current or cell-surface expression of homomeric mutants G601S and R582C. However, remdesivir decreased while lumacaftor increased the current and cell-surface expression of heteromeric channels formed by WT hERG and mutant G601S or R582C hERG. We concluded that drugs can differentially affect homomeric WT and heteromeric WT+G601S (or WT+R582C) hERG channels. These findings extend our understanding of drug-channel interaction and may have clinical implications for patients with hERG mutations. SIGNIFICANCE STATEMENT: Various naturally occurring mutations in a cardiac potassium channel called hERG can impair channel function by decreasing cell-surface channel expression, resulting in cardiac electrical disturbances and even sudden cardiac death. Promotion of cell-surface expression of mutant hERG channels represents a strategy to rescue channel function. This work demonstrates that drugs such as remdesivir and lumacaftor can differently affect homomeric and heteromeric mutant hERG channels, which have biological and clinical implications.
Collapse
Affiliation(s)
- Noah Campagna
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Erika Wall
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Kevin Lee
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Jun Guo
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Wentao Li
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Tonghua Yang
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Adrian Baranchuk
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Mohammad El-Diasty
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Shetuan Zhang
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
20
|
Helliwell MV, Zhang Y, El Harchi A, Dempsey CE, Hancox JC. Inhibition of the hERG Potassium Channel by a Methanesulphonate-Free E-4031 Analogue. Pharmaceuticals (Basel) 2023; 16:1204. [PMID: 37765012 PMCID: PMC10536391 DOI: 10.3390/ph16091204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 09/29/2023] Open
Abstract
hERG (human Ether-à-go-go Related Gene)-encoded potassium channels underlie the cardiac rapid delayed rectifier (IKr) potassium current, which is a major target for antiarrhythmic agents and diverse non-cardiac drugs linked to the drug-induced form of long QT syndrome. E-4031 is a high potency hERG channel inhibitor from the methanesulphonanilide drug family. This study utilized a methanesulphonate-lacking E-4031 analogue, "E-4031-17", to evaluate the role of the methanesulphonamide group in E-4031 inhibition of hERG. Whole-cell patch-clamp measurements of the hERG current (IhERG) were made at physiological temperature from HEK 293 cells expressing wild-type (WT) and mutant hERG constructs. For E-4031, WT IhERG was inhibited by a half-maximal inhibitory concentration (IC50) of 15.8 nM, whilst the comparable value for E-4031-17 was 40.3 nM. Both compounds exhibited voltage- and time-dependent inhibition, but they differed in their response to successive applications of a long (10 s) depolarisation protocol, consistent with greater dissociation of E-4031-17 than the parent compound between applied commands. Voltage-dependent inactivation was left-ward voltage shifted for E-4031 but not for E-4031-17; however, inhibition by both compounds was strongly reduced by attenuated-inactivation mutations. Mutations of S6 and S5 aromatic residues (F656V, Y652A, F557L) greatly attenuated actions of both drugs. The S624A mutation also reduced IhERG inhibition by both molecules. Overall, these results demonstrate that the lack of a methanesulphonate in E-4031-17 is not an impediment to high potency inhibition of IhERG.
Collapse
Affiliation(s)
- Matthew V. Helliwell
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK; (M.V.H.); (C.E.D.)
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK; (Y.Z.); (A.E.H.)
| | - Yihong Zhang
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK; (Y.Z.); (A.E.H.)
| | - Aziza El Harchi
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK; (Y.Z.); (A.E.H.)
| | - Christopher E. Dempsey
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK; (M.V.H.); (C.E.D.)
| | - Jules C. Hancox
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK; (Y.Z.); (A.E.H.)
| |
Collapse
|
21
|
Cheng D, Wei X, Zhang Y, Zhang Q, Xu J, Yang J, Yu J, Stalin A, Liu H, Wang J, Zhong D, Pan L, Zhao W, Chen Y. The Strength of hERG Inhibition by Erythromycin at Different Temperatures Might Be Due to Its Interacting Features with the Channels. Molecules 2023; 28:5176. [PMID: 37446837 DOI: 10.3390/molecules28135176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Erythromycin is one of the few compounds that remarkably increase ether-a-go-go-related gene (hERG) inhibition from room temperature (RT) to physiological temperature (PT). Understanding how erythromycin inhibits the hERG could help us to decide which compounds are needed for further studies. The whole-cell patch clamp technique was used to investigate the effects of erythromycin on hERG channels at different temperatures. While erythromycin caused a concentration-dependent inhibition of cardiac hERG channels, it also shifted the steady-state activation and steady-state inactivation of the channel to the left and significantly accelerated the onset of inactivation at both temperatures, although temperature itself caused a profound change in the dynamics of hERG channels. Our data also suggest that the binding pattern to S6 of the channels changes at PT. In contrast, cisapride, a well-known hERG blocker whose inhibition is not affected by temperature, does not change its critical binding sites after the temperature is raised to PT. Our data suggest that erythromycin is unique and that the shift in hERG inhibition may not apply to other compounds.
Collapse
Affiliation(s)
- Dongrong Cheng
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Xiaofeng Wei
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Yanting Zhang
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Qian Zhang
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Jianwei Xu
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Jiaxin Yang
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Junjie Yu
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610064, China
| | - Huan Liu
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Jintao Wang
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Dian Zhong
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Lanying Pan
- Shuren International Medical College, Zhejiang Shuren University, Hangzhou 310009, China
| | - Wei Zhao
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| | - Yuan Chen
- Chinese Herb Medicine Division, Zhejiang Agriculture and Forestry University, 666 Wusu Street, Hangzhou 311300, China
- The State Key Laboratory of Subtropical Silviculture, Zhejiang Agriculture and Forestry University, 666 Wusu St, Hangzhou 311300, China
| |
Collapse
|
22
|
Himmel H, Lagrutta A, Vömel M, Amin RP, Imredy JP, Johnson T, Vinzing M, Prescott J, Blaustein RO. Nonclinical Cardiovascular Assessment of the Soluble Guanylate Cyclase Stimulator Vericiguat. J Pharmacol Exp Ther 2023; 386:26-34. [PMID: 37068911 DOI: 10.1124/jpet.122.001368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 03/28/2023] [Accepted: 04/10/2023] [Indexed: 04/19/2023] Open
Abstract
Vericiguat and its metabolite M-1 were assessed for proarrhythmic risk in nonclinical in vitro and in vivo studies. In vitro manual voltage-clamp recordings at room temperature determined the effect of vericiguat on human Ether-a-go-go Related Gene (hERG) K+ channels. Effects of vericiguat and M-1 on hERG K+, Nav1.5, hCav1.2, hKvLQT1/1minK, and hKv4.3 channels were investigated via automated voltage-clamp recordings at ambient temperature. Effects of vericiguat and M-1 on hERG K+ and Nav1.5 channels at pathophysiological conditions were explored via manual voltage-clamp recordings at physiologic temperature. Single oral doses of vericiguat (0.6, 2.0, and 6.0 mg/kg) were assessed for in vivo proarrhythmic risk via administration to conscious telemetered dogs; electrocardiogram (ECG) and hemodynamic parameters were monitored. ECG recordings were included in 4- and 39-week dog toxicity studies. In manual voltage-clamp recordings, vericiguat inhibited hERG K+-mediated tail currents in a concentration-dependent manner (20% threshold inhibitory concentration ∼1.9 µM). In automated voltage-clamp recordings, neither vericiguat nor M-1 were associated with biologically relevant inhibition (>20%) of hNav1.5, hCav1.2, hKvLQT1, and hKv4.3. No clinically relevant observations were made for hNav1.5 and hKvLQT1 under simulated pathophysiological conditions. Vericiguat was associated with expected mode-of-action-related dose-dependent changes in systolic arterial blood pressure (up to -20%) and heart rate (up to +53%). At maximum vericiguat dose, corrected QT (QTc) interval changes from baseline varied slightly (-6 to +1%) depending on correction formula. Toxicity studies confirmed absence of significant QTc interval changes. There was no evidence of an increased proarrhythmic risk from nonclinical studies with vericiguat or M-1. SIGNIFICANCE STATEMENT: There was no evidence of an increased proarrhythmic risk from in vitro and in vivo nonclinical studies with vericiguat or M-1. The integrated risk assessment of these nonclinical data combined with existing clinical data demonstrate administration of vericiguat 10 mg once daily in patients with heart failure with reduced ejection fraction is not associated with a proarrhythmic risk.
Collapse
Affiliation(s)
- Herbert Himmel
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany (H.H., Mat.V., May.V.) and Merck & Co., Inc., Rahway, New Jersey (A.L., R.P.A., J.P.I., T.J., J.P., R.O.B.)
| | - Armando Lagrutta
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany (H.H., Mat.V., May.V.) and Merck & Co., Inc., Rahway, New Jersey (A.L., R.P.A., J.P.I., T.J., J.P., R.O.B.)
| | - Matthias Vömel
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany (H.H., Mat.V., May.V.) and Merck & Co., Inc., Rahway, New Jersey (A.L., R.P.A., J.P.I., T.J., J.P., R.O.B.)
| | - Rupesh P Amin
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany (H.H., Mat.V., May.V.) and Merck & Co., Inc., Rahway, New Jersey (A.L., R.P.A., J.P.I., T.J., J.P., R.O.B.)
| | - John P Imredy
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany (H.H., Mat.V., May.V.) and Merck & Co., Inc., Rahway, New Jersey (A.L., R.P.A., J.P.I., T.J., J.P., R.O.B.)
| | - Timothy Johnson
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany (H.H., Mat.V., May.V.) and Merck & Co., Inc., Rahway, New Jersey (A.L., R.P.A., J.P.I., T.J., J.P., R.O.B.)
| | - Maya Vinzing
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany (H.H., Mat.V., May.V.) and Merck & Co., Inc., Rahway, New Jersey (A.L., R.P.A., J.P.I., T.J., J.P., R.O.B.)
| | - Judith Prescott
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany (H.H., Mat.V., May.V.) and Merck & Co., Inc., Rahway, New Jersey (A.L., R.P.A., J.P.I., T.J., J.P., R.O.B.)
| | - Robert O Blaustein
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany (H.H., Mat.V., May.V.) and Merck & Co., Inc., Rahway, New Jersey (A.L., R.P.A., J.P.I., T.J., J.P., R.O.B.)
| |
Collapse
|
23
|
Meier S, Grundland A, Dobrev D, Volders PG, Heijman J. In silico analysis of the dynamic regulation of cardiac electrophysiology by K v 11.1 ion-channel trafficking. J Physiol 2023; 601:2711-2731. [PMID: 36752166 PMCID: PMC10313819 DOI: 10.1113/jp283976] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Cardiac electrophysiology is regulated by continuous trafficking and internalization of ion channels occurring over minutes to hours. Kv 11.1 (also known as hERG) underlies the rapidly activating delayed-rectifier K+ current (IKr ), which plays a major role in cardiac ventricular repolarization. Experimental characterization of the distinct temporal effects of genetic and acquired modulators on channel trafficking and gating is challenging. Computer models are instrumental in elucidating these effects, but no currently available model incorporates ion-channel trafficking. Here, we present a novel computational model that reproduces the experimentally observed production, forward trafficking, internalization, recycling and degradation of Kv 11.1 channels, as well as their modulation by temperature, pentamidine, dofetilide and extracellular K+ . The acute effects of these modulators on channel gating were also incorporated and integrated with the trafficking model in the O'Hara-Rudy human ventricular cardiomyocyte model. Supraphysiological dofetilide concentrations substantially increased Kv 11.1 membrane levels while also producing a significant channel block. However, clinically relevant concentrations did not affect trafficking. Similarly, severe hypokalaemia reduced Kv 11.1 membrane levels based on long-term culture data, but had limited effect based on short-term data. By contrast, clinically relevant elevations in temperature acutely increased IKr due to faster kinetics, while after 24 h, IKr was decreased due to reduced Kv 11.1 membrane levels. The opposite was true for lower temperatures. Taken together, our model reveals a complex temporal regulation of cardiac electrophysiology by temperature, hypokalaemia, and dofetilide through competing effects on channel gating and trafficking, and provides a framework for future studies assessing the role of impaired trafficking in cardiac arrhythmias. KEY POINTS: Kv 11.1 channels underlying the rapidly activating delayed-rectifier K+ current are important for ventricular repolarization and are continuously shuttled from the cytoplasm to the plasma membrane and back over minutes to hours. Kv 11.1 gating and trafficking are modulated by temperature, drugs and extracellular K+ concentration but experimental characterization of their combined effects is challenging. Computer models may facilitate these analyses, but no currently available model incorporates ion-channel trafficking. We introduce a new two-state ion-channel trafficking model able to reproduce a wide range of experimental data, along with the effects of modulators of Kv 11.1 channel functioning and trafficking. The model reveals complex dynamic regulation of ventricular repolarization by temperature, extracellular K+ concentration and dofetilide through opposing acute (millisecond) effects on Kv 11.1 gating and long-term (hours) modulation of Kv 11.1 trafficking. This in silico trafficking framework provides a tool to investigate the roles of acute and long-term processes on arrhythmia promotion and maintenance.
Collapse
Affiliation(s)
- Stefan Meier
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine, and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Adaïa Grundland
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine, and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Data Science and Knowledge Engineering, Faculty of Science and Engineering, Maastricht University, Maastricht, The Netherlands
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Quebec, Canada
| | - Paul G.A. Volders
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine, and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine, and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
24
|
Ma J, Wang NY, Jagani R, Wang HS. Proarrhythmic toxicity of low dose bisphenol A and its analogs in human iPSC-derived cardiomyocytes and human cardiac organoids through delay of cardiac repolarization. CHEMOSPHERE 2023; 328:138562. [PMID: 37004823 PMCID: PMC10121900 DOI: 10.1016/j.chemosphere.2023.138562] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/07/2023] [Accepted: 03/31/2023] [Indexed: 06/19/2023]
Abstract
Bisphenol A (BPA) and its analogs are common environmental chemicals with many potential adverse health effects. The impact of environmentally relevant low dose BPA on human heart, including cardiac electrical properties, is not understood. Perturbation of cardiac electrical properties is a key arrhythmogenic mechanism. In particular, delay of cardiac repolarization can cause ectopic excitation of cardiomyocytes and malignant arrhythmia. This can occur as a result of genetic mutations (i.e., long QT (LQT) syndrome), or cardiotoxicity of drugs and environmental chemicals. To define the impact of low dose BPA on electrical properties of cardiomyocytes in a human-relevant model system, we examined the rapid effects of 1 nM BPA in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) using patch-clamp and confocal fluorescence imaging. Acute exposure to BPA delayed repolarization and prolonged action potential duration (APD) in hiPSC-CMs through inhibition of the hERG K+ channel. In nodal-like hiPSC-CMs, BPA acutely increased pacing rate through stimulation of the If pacemaker channel. Existing arrhythmia susceptibility determines the response of hiPSC-CMs to BPA. BPA resulted in modest APD prolongation but no ectopic excitation in baseline condition, while rapidly promoted aberrant excitations and tachycardia-like events in myocytes that had drug-simulated LQT phenotype. In hiPSC-CM-based human cardiac organoids, the effects of BPA on APD and aberrant excitation were shared by its analog chemicals, which are often used in "BPA-free" products, with bisphenol AF having the largest effects. Our results reveal that BPA and its analogs have repolarization delay-associated pro-arrhythmic toxicity in human cardiomyocytes, particularly in myocytes that are prone to arrhythmias. The toxicity of these chemicals depends on existing pathophysiological conditions of the heart, and may be particularly pronounced in susceptible individuals. An individualized approach is needed in risk assessment and protection.
Collapse
Affiliation(s)
- Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | | | - Ravikumar Jagani
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hong-Sheng Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
25
|
Sakamoto K, Matsumoto S, Abe N, Sentoku M, Yasuda K. Importance of Spatial Arrangement of Cardiomyocyte Network for Precise and Stable On-Chip Predictive Cardiotoxicity Measurement. MICROMACHINES 2023; 14:854. [PMID: 37421087 DOI: 10.3390/mi14040854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/13/2023] [Accepted: 04/13/2023] [Indexed: 07/09/2023]
Abstract
One of the advantages of human stem cell-derived cell-based preclinical screening is the reduction of the false negative/positive misjudgment of lead compounds for predicting their effectiveness and risks during the early stage of development. However, as the community effect of cells was neglected in the conventional single cell-based in vitro screening, the potential difference in results caused by the cell number and their spatial arrangement differences has not yet been sufficiently evaluated. Here, we have investigated the effect of the community size and spatial arrangement difference for cardiomyocyte network response against the proarrhythmic compounds from the viewpoint of in vitro cardiotoxicity. Using three different typical types of cell networks of cardiomyocytes, small cluster, large square sheet, and large closed-loop sheet were formed in shaped agarose microchambers fabricated on a multielectrode array chip simultaneously, and their responses were compared against the proarrhythmic compound, E-4031. The interspike intervals (ISIs) in large square sheets and closed-loop sheets were durable and maintained stable against E-4031 even at a high dose of 100 nM. In contrast, those in the small cluster, which fluctuated even without E-4031, acquired stable beating reflecting the antiarrhythmic efficacy of E-4031 from a 10 nM medium dose administration. The repolarization index, field potential duration (FPD), was prolonged in closed-loop sheets with 10 nM E-4031, even though small clusters and large sheets remained normal at this concentration. Moreover, FPDs of large sheets were the most durable against E-4031 among the three geometries of cardiomyocyte networks. The results showed the apparent spatial arrangement dependence on the stability of their interspike intervals, and FPD prolongation, indicating the importance of the geometry control of cell networks for representing the appropriate response of cardiomyocytes against the adequate amount of compounds for in vitro ion channel measurement.
Collapse
Affiliation(s)
- Kazufumi Sakamoto
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Suguru Matsumoto
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Nanami Abe
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Mitsuru Sentoku
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Kenji Yasuda
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
- Department of Physics, School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan
| |
Collapse
|
26
|
Li E, Kool W, Woolschot L, van der Heyden MAG. Chronic Propafenone Application Increases Functional K IR2.1 Expression In Vitro. Pharmaceuticals (Basel) 2023; 16:ph16030404. [PMID: 36986503 PMCID: PMC10056987 DOI: 10.3390/ph16030404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Expression and activity of inwardly rectifying potassium (KIR) channels within the heart are strictly regulated. KIR channels have an important role in shaping cardiac action potentials, having a limited conductance at depolarized potentials but contributing to the final stage of repolarization and resting membrane stability. Impaired KIR2.1 function causes Andersen-Tawil Syndrome (ATS) and is associated with heart failure. Restoring KIR2.1 function by agonists of KIR2.1 (AgoKirs) would be beneficial. The class 1c antiarrhythmic drug propafenone is identified as an AgoKir; however, its long-term effects on KIR2.1 protein expression, subcellular localization, and function are unknown. Propafenone's long-term effect on KIR2.1 expression and its underlying mechanisms in vitro were investigated. KIR2.1-carried currents were measured by single-cell patch-clamp electrophysiology. KIR2.1 protein expression levels were determined by Western blot analysis, whereas conventional immunofluorescence and advanced live-imaging microscopy were used to assess the subcellular localization of KIR2.1 proteins. Acute propafenone treatment at low concentrations supports the ability of propafenone to function as an AgoKir without disturbing KIR2.1 protein handling. Chronic propafenone treatment (at 25-100 times higher concentrations than in the acute treatment) increases KIR2.1 protein expression and KIR2.1 current densities in vitro, which are potentially associated with pre-lysosomal trafficking inhibition.
Collapse
Affiliation(s)
- Encan Li
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Willy Kool
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Liset Woolschot
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Marcel A G van der Heyden
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| |
Collapse
|
27
|
Soepriatna AH, Navarrete-Welton A, Kim TY, Daley MC, Bronk P, Kofron CM, Mende U, Coulombe KLK, Choi BR. Action potential metrics and automated data analysis pipeline for cardiotoxicity testing using optically mapped hiPSC-derived 3D cardiac microtissues. PLoS One 2023; 18:e0280406. [PMID: 36745602 PMCID: PMC9901774 DOI: 10.1371/journal.pone.0280406] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/28/2022] [Indexed: 02/07/2023] Open
Abstract
Recent advances in human induced pluripotent stem cell (hiPSC)-derived cardiac microtissues provide a unique opportunity for cardiotoxic assessment of pharmaceutical and environmental compounds. Here, we developed a series of automated data processing algorithms to assess changes in action potential (AP) properties for cardiotoxicity testing in 3D engineered cardiac microtissues generated from hiPSC-derived cardiomyocytes (hiPSC-CMs). Purified hiPSC-CMs were mixed with 5-25% human cardiac fibroblasts (hCFs) under scaffold-free conditions and allowed to self-assemble into 3D spherical microtissues in 35-microwell agarose gels. Optical mapping was performed to quantify electrophysiological changes. To increase throughput, AP traces from 4x4 cardiac microtissues were simultaneously acquired with a voltage sensitive dye and a CMOS camera. Individual microtissues showing APs were identified using automated thresholding after Fourier transforming traces. An asymmetric least squares method was used to correct non-uniform background and baseline drift, and the fluorescence was normalized (ΔF/F0). Bilateral filtering was applied to preserve the sharpness of the AP upstroke. AP shape changes under selective ion channel block were characterized using AP metrics including stimulation delay, rise time of AP upstroke, APD30, APD50, APD80, APDmxr (maximum rate change of repolarization), and AP triangulation (APDtri = APDmxr-APD50). We also characterized changes in AP metrics under various ion channel block conditions with multi-class logistic regression and feature extraction using principal component analysis of human AP computer simulations. Simulation results were validated experimentally with selective pharmacological ion channel blockers. In conclusion, this simple and robust automated data analysis pipeline for evaluating key AP metrics provides an excellent in vitro cardiotoxicity testing platform for a wide range of environmental and pharmaceutical compounds.
Collapse
Affiliation(s)
- Arvin H. Soepriatna
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, United States of America
| | - Allison Navarrete-Welton
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Tae Yun Kim
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Mark C. Daley
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, United States of America
| | - Peter Bronk
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Celinda M. Kofron
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, United States of America
| | - Ulrike Mende
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Kareen L. K. Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, United States of America
| | - Bum-Rak Choi
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- * E-mail:
| |
Collapse
|
28
|
Ukachukwu CU, Jimenez-Vazquez EN, Jain A, Jones DK. hERG1 channel subunit composition mediates proton inhibition of rapid delayed rectifier potassium current (I Kr) in cardiomyocytes derived from hiPSCs. J Biol Chem 2023; 299:102778. [PMID: 36496073 PMCID: PMC9867984 DOI: 10.1016/j.jbc.2022.102778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/29/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
The voltage-gated channel, hERG1, conducts the rapid delayed rectifier potassium current (IKr) and is critical for human cardiac repolarization. Reduced IKr causes long QT syndrome and increases the risk for cardiac arrhythmia and sudden death. At least two subunits form functional hERG1 channels, hERG1a and hERG1b. Changes in hERG1a/1b abundance modulate IKr kinetics, magnitude, and drug sensitivity. Studies from native cardiac tissue suggest that hERG1 subunit abundance is dynamically regulated, but the impact of altered subunit abundance on IKr and its response to external stressors is not well understood. Here, we used a substrate-driven human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) maturation model to investigate how changes in relative hERG1a/1b subunit abundance impact the response of native IKr to extracellular acidosis, a known component of ischemic heart disease and sudden infant death syndrome. IKr recorded from immatured hiPSC-CMs displays a 2-fold greater inhibition by extracellular acidosis (pH 6.3) compared with matured hiPSC-CMs. Quantitative RT-PCR and immunocytochemistry demonstrated that hERG1a subunit mRNA and protein were upregulated and hERG1b subunit mRNA and protein were downregulated in matured hiPSC-CMs compared with immatured hiPSC-CMs. The shift in subunit abundance in matured hiPSC-CMs was accompanied by increased IKr. Silencing hERG1b's impact on native IKr kinetics by overexpressing a polypeptide identical to the hERG1a N-terminal Per-Arnt-Sim domain reduced the magnitude of IKr proton inhibition in immatured hiPSC-CMs to levels comparable to those observed in matured hiPSC-CMs. These data demonstrate that hERG1 subunit abundance is dynamically regulated and determines IKr proton sensitivity in hiPSC-CMs.
Collapse
Affiliation(s)
- Chiamaka U Ukachukwu
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Eric N Jimenez-Vazquez
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Abhilasha Jain
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - David K Jones
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA; Department of Internal Medicine, University of Michigan Medical School.
| |
Collapse
|
29
|
Tieu A, Phillips KG, Costa KD, Mayourian J. Computational design of custom therapeutic cells to correct failing human cardiomyocytes. FRONTIERS IN SYSTEMS BIOLOGY 2023; 3:1102467. [PMID: 36743445 PMCID: PMC9894098 DOI: 10.3389/fsysb.2023.1102467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Background Myocardial delivery of non-excitable cells-namely human mesenchymal stem cells (hMSCs) and c-kit+ cardiac interstitial cells (hCICs)-remains a promising approach for treating the failing heart. Recent empirical studies attempt to improve such therapies by genetically engineering cells to express specific ion channels, or by creating hybrid cells with combined channel expression. This study uses a computational modeling approach to test the hypothesis that custom hypothetical cells can be rationally designed to restore a healthy phenotype when coupled to human heart failure (HF) cardiomyocytes. Methods Candidate custom cells were simulated with a combination of ion channels from non-excitable cells and healthy human cardiomyocytes (hCMs). Using a genetic algorithm-based optimization approach, candidate cells were accepted if a root mean square error (RMSE) of less than 50% relative to healthy hCM was achieved for both action potential and calcium transient waveforms for the cell-treated HF cardiomyocyte, normalized to the untreated HF cardiomyocyte. Results Custom cells expressing only non-excitable ion channels were inadequate to restore a healthy cardiac phenotype when coupled to either fibrotic or non-fibrotic HF cardiomyocytes. In contrast, custom cells also expressing cardiac ion channels led to acceptable restoration of a healthy cardiomyocyte phenotype when coupled to fibrotic, but not non-fibrotic, HF cardiomyocytes. Incorporating the cardiomyocyte inward rectifier K+ channel was critical to accomplishing this phenotypic rescue while also improving single-cell action potential metrics associated with arrhythmias, namely resting membrane potential and action potential duration. The computational approach also provided insight into the rescue mechanisms, whereby heterocellular coupling enhanced cardiomyocyte L-type calcium current and promoted calcium-induced calcium release. Finally, as a therapeutically translatable strategy, we simulated delivery of hMSCs and hCICs genetically engineered to express the cardiomyocyte inward rectifier K+ channel, which decreased action potential and calcium transient RMSEs by at least 24% relative to control hMSCs and hCICs, with more favorable single-cell arrhythmia metrics. Conclusion Computational modeling facilitates exploration of customizable engineered cell therapies. Optimized cells expressing cardiac ion channels restored healthy action potential and calcium handling phenotypes in fibrotic HF cardiomyocytes and improved single-cell arrhythmia metrics, warranting further experimental validation studies of the proposed custom therapeutic cells.
Collapse
Affiliation(s)
- Andrew Tieu
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Katherine G. Phillips
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, United States
| | - Kevin D. Costa
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States,CORRESPONDENCE: Kevin D. Costa, Joshua Mayourian,
| | - Joshua Mayourian
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States,Department of Pediatrics, Harvard Medical School, Boston, MA, United States,Department of Pediatrics, Boston University, Boston, MA, United States,Department of Pediatrics, Boston Medical Center, Boston, MA, United States,CORRESPONDENCE: Kevin D. Costa, Joshua Mayourian,
| |
Collapse
|
30
|
Hwang M, Lee SJ, Lim CH, Shim EB, Lee HA. The three-dimensionality of the hiPSC-CM spheroid contributes to the variability of the field potential. Front Physiol 2023; 14:1123190. [PMID: 37025386 PMCID: PMC10070703 DOI: 10.3389/fphys.2023.1123190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/10/2023] [Indexed: 04/08/2023] Open
Abstract
Background: Field potential (FP) signals from human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) spheroid which are used for drug safety tests in the preclinical stage are different from action potential (AP) signals and require working knowledge of the multi-electrode array (MEA) system. In this study, we developed in silico three-dimensional (3-D) models of hiPSC-CM spheroids for the simulation of field potential measurement. We compared our model simulation results against in vitro experimental data under the effect of drugs E-4031 and nifedipine. Methods: In silico 3-D models of hiPSC-CM spheroids were constructed in spherical and discoidal shapes. Tetrahedral meshes were generated inside the models, and the propagation of the action potential in the model was obtained by numerically solving the monodomain reaction-diffusion equation. An electrical model of electrode was constructed and FPs were calculated using the extracellular potentials from the AP propagations. The effects of drugs were simulated by matching the simulation results with in vitro experimental data. Results: The simulated FPs from the 3-D models of hiPSC-CM spheroids exhibited highly variable shapes depending on the stimulation and measurement locations. The values of the IC50 of E-4031 and nifedipine calculated by matching the simulated FP durations with in vitro experimental data were in line with the experimentally measured ones reported in the literature. Conclusion: The 3-D in silico models of hiPSC-CM spheroids generated highly variable FPs similar to those observed in in vitro experiments. The in silico model has the potential to complement the interpretation of the FP signals obtained from in vitro experiments.
Collapse
Affiliation(s)
| | - Su-Jin Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | | | - Eun Bo Shim
- AI Medic, Inc., Seoul, Republic of Korea
- Department of Mechanical and Biomedical Engineering, Kangwon National University, Chuncheon, Republic of Korea
- *Correspondence: Eun Bo Shim, ; Hyang-Ae Lee,
| | - Hyang-Ae Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Republic of Korea
- *Correspondence: Eun Bo Shim, ; Hyang-Ae Lee,
| |
Collapse
|
31
|
Maly J, Emigh AM, DeMarco KR, Furutani K, Sack JT, Clancy CE, Vorobyov I, Yarov-Yarovoy V. Structural modeling of the hERG potassium channel and associated drug interactions. Front Pharmacol 2022; 13:966463. [PMID: 36188564 PMCID: PMC9523588 DOI: 10.3389/fphar.2022.966463] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
The voltage-gated potassium channel, KV11.1, encoded by the human Ether-à-go-go-Related Gene (hERG), is expressed in cardiac myocytes, where it is crucial for the membrane repolarization of the action potential. Gating of the hERG channel is characterized by rapid, voltage-dependent, C-type inactivation, which blocks ion conduction and is suggested to involve constriction of the selectivity filter. Mutations S620T and S641A/T within the selectivity filter region of hERG have been shown to alter the voltage dependence of channel inactivation. Because hERG channel blockade is implicated in drug-induced arrhythmias associated with both the open and inactivated states, we used Rosetta to simulate the effects of hERG S620T and S641A/T mutations to elucidate conformational changes associated with hERG channel inactivation and differences in drug binding between the two states. Rosetta modeling of the S641A fast-inactivating mutation revealed a lateral shift of the F627 side chain in the selectivity filter into the central channel axis along the ion conduction pathway and the formation of four lateral fenestrations in the pore. Rosetta modeling of the non-inactivating mutations S620T and S641T suggested a potential molecular mechanism preventing F627 side chain from shifting into the ion conduction pathway during the proposed inactivation process. Furthermore, we used Rosetta docking to explore the binding mechanism of highly selective and potent hERG blockers - dofetilide, terfenadine, and E4031. Our structural modeling correlates well with much, but not all, existing experimental evidence involving interactions of hERG blockers with key residues in hERG pore and reveals potential molecular mechanisms of ligand interactions with hERG in an inactivated state.
Collapse
Affiliation(s)
- Jan Maly
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
- Biophysics Graduate Group, University of California, Davis, Davis, CA, United States
| | - Aiyana M. Emigh
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
- Biophysics Graduate Group, University of California, Davis, Davis, CA, United States
| | - Kevin R. DeMarco
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
- Biophysics Graduate Group, University of California, Davis, Davis, CA, United States
| | - Kazuharu Furutani
- Department of Pharmacology, Tokushima Bunri University, Tokushima, Japan
| | - Jon T. Sack
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| | - Colleen E. Clancy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
32
|
Johnson AA, Crawford TR, Trudeau MC. The N-linker region of hERG1a upregulates hERG1b potassium channels. J Biol Chem 2022; 298:102233. [PMID: 35798139 PMCID: PMC9428852 DOI: 10.1016/j.jbc.2022.102233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 11/29/2022] Open
Abstract
A major physiological role of hERG1 (human Ether-á-go-go-Related Gene 1) potassium channels is to repolarize cardiac action potentials. Two isoforms, hERG1a and hERG1b, associate to form the potassium current IKr in cardiomyocytes. Inherited mutations in hERG1a or hERG1b cause prolonged cardiac repolarization, long QT syndrome, and sudden death arrhythmia. hERG1a subunits assemble with and enhance the number of hERG1b subunits at the plasma membrane, but the mechanism for the increase in hERG1b by hERG1a is not well understood. Here, we report that the hERG1a N-terminal region expressed in trans with hERG1b markedly increased hERG1b currents and increased biotin-labeled hERG1b protein at the membrane surface. hERG1b channels with a deletion of the N-terminal 1b domain did not have a measurable increase in current or biotinylated protein when coexpressed with hERG1a N-terminal regions, indicating that the 1b domain was required for the increase in hERG1b. Using a biochemical pull-down interaction assay and a FRET hybridization experiment, we detected a direct interaction between the hERG1a N-terminal region and the hERG1b N-terminal region. Using engineered deletions and alanine mutagenesis, we identified a short span of amino acids at positions 216 to 220 within the hERG1a "N-linker" region that were necessary for the upregulation of hERG1b. We propose that direct structural interactions between the hERG1a N-linker region and the hERG1b 1b domain increase hERG1b at the plasma membrane. Mechanisms regulating hERG1a and hERG1b are likely critical for cardiac function, may be disrupted by long QT syndrome mutants, and serve as potential targets for therapeutics.
Collapse
Affiliation(s)
- Ashley A Johnson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Taylor R Crawford
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Matthew C Trudeau
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
33
|
Furutani K, Kawano R, Ichiwara M, Adachi R, Clancy CE, Sack JT, Kita S. Pore opening, not voltage sensor movement, underpins the voltage-dependence of facilitation by a hERG blocker. Mol Pharmacol 2022; 102:MOLPHARM-AR-2022-000569. [PMID: 36041862 PMCID: PMC9595204 DOI: 10.1124/molpharm.122.000569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/19/2022] [Accepted: 08/04/2022] [Indexed: 11/07/2022] Open
Abstract
A drug that blocks the cardiac myocyte voltage-gated K+ channels encoded by the human Ether-à-go-go-Related Gene (hERG) carries a potential risk of long QT syndrome and life-threatening cardiac arrhythmia, including Torsade de Points Interestingly, certain hERG blockers can also facilitate hERG activation to increase hERG currents, which may reduce proarrhythmic potential. However, the molecular mechanism involved in the facilitation effect of hERG blockers remains unclear. The hallmark feature of the facilitation effect by hERG blockers is that a depolarizing preconditioning pulse shifts voltage-dependence of hERG activation to more negative voltages. Here we utilize a D540K hERG mutant to study the mechanism of the facilitation effect. D540K hERG is activated by not only depolarization but also hyperpolarization. This unusual gating property enables tests of the mechanism by which voltage induces facilitation of hERG by blockers. With D540K hERG, we find that nifekalant, a hERG blocker and Class III antiarrhythmic agent, blocks and facilitates not only current activation by depolarization but also current activation by hyperpolarization, suggesting a shared gating process upon depolarization and hyperpolarization. Moreover, in response to hyperpolarizing conditioning pulses, nifekalant facilitates D540K hERG currents but not wild-type currents. Our results indicate that induction of facilitation is coupled to pore opening, not voltage per se We propose that gated access to the hERG central cavity underlies the voltage-dependence of induction of facilitation. This study identifies hERG channel pore gate opening as the conformational change facilitated by nifekalant, a clinically important antiarrhythmic agent. Significance Statement Nifekalant is a clinically important antiarrhythmic agent and a hERG blocker which can also facilitate voltage-dependent activation of hERG channels after a preconditioning pulse. Here we show that the mechanism of action of the preconditioning pulse is to open a conductance gate to enable drug access to a facilitation site. Moreover, we find that facilitation increases hERG currents by altering pore dynamics, rather than acting through voltage sensors.
Collapse
Affiliation(s)
| | - Ryotaro Kawano
- Department of Pharmacology, Tokushima Bunri University, Japan
| | - Minami Ichiwara
- Department of Pharmacology, Tokushima Bunri University, Japan
| | - Ryo Adachi
- Department of Pharmacology, Tokushima Bunri University, Japan
| | | | - Jon T Sack
- UC Davis School of Medicine, United States
| | - Satomi Kita
- Department of Pharmacology, Tokushima Bunri University, Japan
| |
Collapse
|
34
|
Baron CA, Thiebaud N, Ren M, Viatchenko-Karpinski S, Indapurkar A, King T, Matta MK, Ismaiel OA, Patel V, Mashaee M, Vicente J, Wu WW. hERG block potencies for 5 positive control drugs obtained per ICH E14/S7B Q&As best practices: Impact of recording temperature and drug loss. J Pharmacol Toxicol Methods 2022; 117:107193. [DOI: 10.1016/j.vascn.2022.107193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/22/2022] [Accepted: 06/25/2022] [Indexed: 12/14/2022]
|
35
|
Metformin Reduces Potassium Currents and Prolongs Repolarization in Non-Diabetic Heart. Int J Mol Sci 2022; 23:ijms23116021. [PMID: 35682699 PMCID: PMC9181026 DOI: 10.3390/ijms23116021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/14/2022] [Accepted: 05/24/2022] [Indexed: 01/27/2023] Open
Abstract
Metformin is the first choice drug for the treatment of type 2 diabetes due to positive results in reducing hyperglycaemia and insulin resistance. However, diabetic patients have higher risk of ventricular arrhythmia and sudden cardiac death, and metformin failed to reduce ventricular arrhythmia in clinical trials. In order to explore the mechanisms responsible for the lack of protective effect, we investigated in vivo the effect of metformin on cardiac electrical activity in non-diabetic rats; and in vitro in isolated ventricular myocytes, HEK293 cells expressing the hERG channel and human induced pluripotent stem cells derived cardiomyocytes (hIPS-CMs). Surface electrocardiograms showed that long-term metformin treatment (7 weeks) at therapeutic doses prolonged cardiac repolarization, reflected as QT and QTc interval duration, and increased ventricular arrhythmia during the caffeine/dobutamine challenge. Patch-clamp recordings in ventricular myocytes isolated from treated animals showed that the cellular mechanism is a reduction in the cardiac transient outward potassium current (Ito). In vitro, incubation with metformin for 24 h also reduced Ito, prolonged action potential duration, and increased spontaneous contractions in ventricular myocytes isolated from control rats. Metformin incubation also reduced IhERG in HEK293 cells. Finally, metformin incubation prolonged action potential duration at 30% and 90% of repolarization in hIPS-CMs, which is compatible with the reduction of Ito and IhERG. Our results show that metformin directly modifies the electrical behavior of the normal heart. The mechanism consists in the inhibition of repolarizing currents and the subsequent decrease in repolarization capacity, which prolongs AP and QTc duration.
Collapse
|
36
|
Straface M, Koussai MA, Makwana R, Crawley E, Palmer A, Cai W, Gharibans A, Adebibe M, Loy J, O’Grady G, Andrews PLR, Sanger GJ. A multi-parameter approach to measurement of spontaneous myogenic contractions in human stomach: Utilization to assess potential modulators of myogenic contractions. Pharmacol Res 2022; 180:106247. [DOI: 10.1016/j.phrs.2022.106247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/22/2022] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
|
37
|
Al-Moubarak E, Shiels HA, Zhang Y, Du C, Hanington O, Harmer SC, Dempsey CE, Hancox JC. Inhibition of the hERG potassium channel by phenanthrene: a polycyclic aromatic hydrocarbon pollutant. Cell Mol Life Sci 2021; 78:7899-7914. [PMID: 34727194 PMCID: PMC8629796 DOI: 10.1007/s00018-021-03967-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/25/2021] [Accepted: 10/01/2021] [Indexed: 11/07/2022]
Abstract
The lipophilic polycyclic aromatic hydrocarbon (PAH) phenanthrene is relatively abundant in polluted air and water and can access and accumulate in human tissue. Phenanthrene has been reported to interact with cardiac ion channels in several fish species. This study was undertaken to investigate the ability of phenanthrene to interact with hERG (human Ether-à-go-go-Related Gene) encoded Kv11.1 K+ channels, which play a central role in human ventricular repolarization. Pharmacological inhibition of hERG can be proarrhythmic. Whole-cell patch clamp recordings of hERG current (IhERG) were made from HEK293 cells expressing wild-type (WT) and mutant hERG channels. WT IhERG1a was inhibited by phenanthrene with an IC50 of 17.6 ± 1.7 µM, whilst IhERG1a/1b exhibited an IC50 of 1.8 ± 0.3 µM. WT IhERG block showed marked voltage and time dependence, indicative of dependence of inhibition on channel gating. The inhibitory effect of phenanthrene was markedly impaired by the attenuated inactivation N588K mutation. Remarkably, mutations of S6 domain aromatic amino acids (Y652, F656) in the canonical drug binding site did not impair the inhibitory action of phenanthrene; the Y652A mutation augmented IhERG block. In contrast, the F557L (S5) and M651A (S6) mutations impaired the ability of phenanthrene to inhibit IhERG, as did the S624A mutation below the selectivity filter region. Computational docking using a cryo-EM derived hERG structure supported the mutagenesis data. Thus, phenanthrene acts as an inhibitor of the hERG K+ channel by directly interacting with the channel, binding to a distinct site in the channel pore domain.
Collapse
Affiliation(s)
- Ehab Al-Moubarak
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Holly A Shiels
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Yihong Zhang
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Chunyun Du
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Oliver Hanington
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Stephen C Harmer
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | | | - Jules C Hancox
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
38
|
Wortmann L, Bräuer N, Holton SJ, Irlbacher H, Weiske J, Lechner C, Meier R, Karén J, Siöberg CB, Pütter V, Christ CD, Ter Laak A, Lienau P, Lesche R, Nicke B, Cheung SH, Bauser M, Haegebarth A, von Nussbaum F, Mumberg D, Lemos C. Discovery and Characterization of the Potent and Highly Selective 1,7-Naphthyridine-Based Inhibitors BAY-091 and BAY-297 of the Kinase PIP4K2A. J Med Chem 2021; 64:15883-15911. [PMID: 34699202 DOI: 10.1021/acs.jmedchem.1c01245] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PIP4K2A is an insufficiently studied type II lipid kinase that catalyzes the conversion of phosphatidylinositol-5-phosphate (PI5P) into phosphatidylinositol 4,5-bisphosphate (PI4,5P2). The involvement of PIP4K2A/B in cancer has been suggested, particularly in the context of p53 mutant/null tumors. PIP4K2A/B depletion has been shown to induce tumor growth inhibition, possibly due to hyperactivation of AKT and reactive oxygen species-mediated apoptosis. Herein, we report the identification of the novel potent and highly selective inhibitors BAY-091 and BAY-297 of the kinase PIP4K2A by high-throughput screening and subsequent structure-based optimization. Cellular target engagement of BAY-091 and BAY-297 was demonstrated using cellular thermal shift assay technology. However, inhibition of PIP4K2A with BAY-091 or BAY-297 did not translate into the hypothesized mode of action and antiproliferative activity in p53-deficient tumor cells. Therefore, BAY-091 and BAY-297 serve as valuable chemical probes to study PIP4K2A signaling and its involvement in pathophysiological conditions such as cancer.
Collapse
Affiliation(s)
- Lars Wortmann
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Nico Bräuer
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Simon J Holton
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Horst Irlbacher
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Jörg Weiske
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Christian Lechner
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Robin Meier
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Jakob Karén
- Pelago Bioscience AB, Banvaktsvägen 20, 171 48 Solna, Sweden
| | | | - Vera Pütter
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Clara D Christ
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Antonius Ter Laak
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Philip Lienau
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Ralf Lesche
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Barbara Nicke
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Shing-Hu Cheung
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Marcus Bauser
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Andrea Haegebarth
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Franz von Nussbaum
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Dominik Mumberg
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Clara Lemos
- Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| |
Collapse
|
39
|
Cai S, Moutal A, Yu J, Chew LA, Isensee J, Chawla R, Gomez K, Luo S, Zhou Y, Chefdeville A, Madura C, Perez-Miller S, Bellampalli SS, Dorame A, Scott DD, François-Moutal L, Shan Z, Woodward T, Gokhale V, Hohmann AG, Vanderah TW, Patek M, Khanna M, Hucho T, Khanna R. Selective targeting of NaV1.7 via inhibition of the CRMP2-Ubc9 interaction reduces pain in rodents. Sci Transl Med 2021; 13:eabh1314. [PMID: 34757807 PMCID: PMC11729770 DOI: 10.1126/scitranslmed.abh1314] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The voltage-gated sodium NaV1.7 channel, critical for sensing pain, has been actively targeted by drug developers; however, there are currently no effective and safe therapies targeting NaV1.7. Here, we tested whether a different approach, indirect NaV1.7 regulation, could have antinociceptive effects in preclinical models. We found that preventing addition of small ubiquitin-like modifier (SUMO) on the NaV1.7-interacting cytosolic collapsin response mediator protein 2 (CRMP2) blocked NaV1.7 functions and had antinociceptive effects in rodents. In silico targeting of the SUMOylation site in CRMP2 (Lys374) identified >200 hits, of which compound 194 exhibited selective in vitro and ex vivo NaV1.7 engagement. Orally administered 194 was not only antinociceptive in preclinical models of acute and chronic pain but also demonstrated synergy alongside other analgesics—without eliciting addiction, rewarding properties, or neurotoxicity. Analgesia conferred by 194 was opioid receptor dependent. Our results demonstrate that 194 is a first-in-class protein-protein inhibitor that capitalizes on CRMP2-NaV1.7 regulation to deliver safe analgesia in rodents.
Collapse
Affiliation(s)
- Song Cai
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Jie Yu
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Lindsey A. Chew
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Jörg Isensee
- Department of Anesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital of Cologne, University Cologne, Joseph-Stelzmann-Str 9, Cologne D-50931, Germany
| | - Reena Chawla
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
| | - Kimberly Gomez
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Shizhen Luo
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Yuan Zhou
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Aude Chefdeville
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Cynthia Madura
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Samantha Perez-Miller
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Shreya Sai Bellampalli
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Angie Dorame
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - David D. Scott
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Liberty François-Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Zhiming Shan
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Taylor Woodward
- Department of Psychological and Brain Sciences, Program in Neuroscience and Gill Center for Biomolecular Science, Indiana University, Bloomington, IN 47405-2204, USA
| | - Vijay Gokhale
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
- College of Pharmacy, University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, USA
| | - Andrea G. Hohmann
- Department of Psychological and Brain Sciences, Program in Neuroscience and Gill Center for Biomolecular Science, Indiana University, Bloomington, IN 47405-2204, USA
| | - Todd W. Vanderah
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- Comprehensive Pain and Addiction Center, The University of Arizona, Tucson, AZ 85724, USA
| | - Marcel Patek
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
- Bright Rock Path LLC, Tucson, AZ 85724, USA
| | - May Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
| | - Tim Hucho
- Department of Anesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital of Cologne, University Cologne, Joseph-Stelzmann-Str 9, Cologne D-50931, Germany
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
- Comprehensive Pain and Addiction Center, The University of Arizona, Tucson, AZ 85724, USA
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
| |
Collapse
|
40
|
Berger M, Wortmann L, Buchgraber P, Lücking U, Zitzmann-Kolbe S, Wengner AM, Bader B, Bömer U, Briem H, Eis K, Rehwinkel H, Bartels F, Moosmayer D, Eberspächer U, Lienau P, Hammer S, Schatz CA, Wang Q, Wang Q, Mumberg D, Nising CF, Siemeister G. BAY-8400: A Novel Potent and Selective DNA-PK Inhibitor which Shows Synergistic Efficacy in Combination with Targeted Alpha Therapies. J Med Chem 2021; 64:12723-12737. [PMID: 34428039 DOI: 10.1021/acs.jmedchem.1c00762] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Eukaryotes have evolved two major pathways to repair potentially lethal DNA double-strand breaks. Homologous recombination represents a precise, DNA-template-based mechanism available during the S and G2 cell cycle phase, whereas non-homologous end joining, which requires DNA-dependent protein kinase (DNA-PK), allows for fast, cell cycle-independent but less accurate DNA repair. Here, we report the discovery of BAY-8400, a novel selective inhibitor of DNA-PK. Starting from a triazoloquinoxaline, which had been identified as a hit from a screen for ataxia telangiectasia and Rad3-related protein (ATR) inhibitors with inhibitory activity against ATR, ATM, and DNA-PK, lead optimization efforts focusing on potency and selectivity led to the discovery of BAY-8400. In in vitro studies, BAY-8400 showed synergistic activity of DNA-PK inhibition with DNA damage-inducing targeted alpha therapy. Combination of PSMA-targeted thorium-227 conjugate BAY 2315497 treatment of human prostate tumor-bearing mice with BAY-8400 oral treatment increased antitumor efficacy, as compared to PSMA-targeted thorium-227 conjugate monotherapy.
Collapse
Affiliation(s)
- Markus Berger
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Lars Wortmann
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Philipp Buchgraber
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Ulrich Lücking
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | | | - Antje M Wengner
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Benjamin Bader
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Ulf Bömer
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Hans Briem
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Knut Eis
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Hartmut Rehwinkel
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Florian Bartels
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Dieter Moosmayer
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Uwe Eberspächer
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Philip Lienau
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Stefanie Hammer
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Christoph A Schatz
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Qiuwen Wang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | - Qi Wang
- WuXi AppTec (Wuhan) Co., Ltd., 666 Gaoxin Road, East Lake High-tech Development Zone, Wuhan 430075, P. R. China
| | - Dominik Mumberg
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Carl F Nising
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| | - Gerhard Siemeister
- Research & Development, Pharmaceuticals, Bayer AG, Berlin 13353, Germany
| |
Collapse
|
41
|
Isogenic Sets of hiPSC-CMs Harboring Distinct KCNH2 Mutations Differ Functionally and in Susceptibility to Drug-Induced Arrhythmias. Stem Cell Reports 2021; 15:1127-1139. [PMID: 33176122 PMCID: PMC7664051 DOI: 10.1016/j.stemcr.2020.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/26/2022] Open
Abstract
Mutations in KCNH2 can lead to long QT syndrome type 2. Variable disease manifestation observed with this channelopathy is associated with the location and type of mutation within the protein, complicating efforts to predict patient risk. Here, we demonstrated phenotypic differences in cardiomyocytes derived from isogenic human induced pluripotent stem cells (hiPSC-CMs) genetically edited to harbor mutations either within the pore or tail region of the ion channel. Electrophysiological analysis confirmed that the mutations prolonged repolarization of the hiPSC-CMs, with differences between the mutations evident in monolayer cultures. Blocking the hERG channel revealed that the pore-loop mutation conferred greater susceptibility to arrhythmic events. These findings showed that subtle phenotypic differences related to KCNH2 mutations could be captured by hiPSC-CMs under genetically matched conditions. Moreover, the results support hiPSC-CMs as strong candidates for evaluating the underlying severity of individual KCNH2 mutations in humans, which could facilitate patient risk stratification. Mutation-specific differences detected in hiPSC-CMs with same genetic background APD and FPD in the hERG pore variant hiPSC-CMs more prolonged than the tail variant The pore variant was also more susceptible to drug-induced arrhythmic events Potential strategy to determine KCNH2 mutation-specific arrhythmic risk
Collapse
|
42
|
Bell DC, Fermini B. Use of automated patch clamp in cardiac safety assessment: Past, present & future perspectives. J Pharmacol Toxicol Methods 2021; 111:107114. [PMID: 34400309 DOI: 10.1016/j.vascn.2021.107114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
There is no doubt that automated patch clamp (APC) technology has revolutionized research in biomedical science. High throughput ion channel screening is now an integral part of the development and safety profiling of the majority of new chemical entities currently developed to address unmet medical needs. The increased throughput it provides has significantly improved the ability to overcome the time-consuming, low throughput bottlenecks resulting from the more conventional manual patch clamp method, considered the 'gold standard', for studying ion channel function and pharmacology. While systems offering the luxury of automation have only been commercially available for two decades, the road leading to this new technology is long and rich in seminal, hands-on, studies dating back as far as the 18th century. So where does this technology currently stand, and what will it look like in the future? In the current article, we review the scientific history leading to the development of APC systems, examine key drivers in the rapid development of this technology (such as failed ion channel programmes and the issue of drug-induced hERG inhibition and QT interval prolongation), highlight key capabilities and finally provide some perspective on the current and future impact of the technology on cardiac safety assessment and biomedical science.
Collapse
Affiliation(s)
- Damian C Bell
- Sophion Bioscience A/S, Ballerup, Copenhagen, Denmark.
| | | |
Collapse
|
43
|
Jenewein T, Kanner SA, Bauer D, Hertel B, Colecraft HM, Moroni A, Thiel G, Kauferstein S. The mutation L69P in the PAS domain of the hERG potassium channel results in LQTS by trafficking deficiency. Channels (Austin) 2021; 14:163-174. [PMID: 32253972 PMCID: PMC7188350 DOI: 10.1080/19336950.2020.1751522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The congenital long QT syndrome (LQTS) is a cardiac disorder characterized by a prolonged QT interval on the electrocardiogram and an increased susceptibility to ventricular arrhythmias and sudden cardiac death. A frequent cause for LQTS is mutations in the KCNH2 gene (also known as the human ether-a-go-go-related gene or hERG), which reduce or modulate the potassium current IKr and hence alter cardiac repolarization. In a patient with a clinically diagnosed LQTS, we identified the mutation L69P in the N-terminal PAS (Per-Arnt-Sim) domain of hERG. Functional expression in HEK293 cells shows that a homotetrameric hERG channel reconstituted with only mutant subunits exhibits a drastically reduced surface expression of the channel protein thus leading to a diminished hERG current. Unlike many other mutations in the hERG-PAS domain the negative impact of the L69P substitution cannot be rescued by facilitated protein folding at a lower incubation temperature. Further, co-expression of wt and mutant monomers does not restore either wt like surface expression or the full hERG current. These results indicate L69P is a dominant negative mutation, with deficits which most likely occurs at the level of protein folding and subsequently inhibits trafficking to the plasma membrane. The functional deficits of the mutant channel support the clinical diagnosis of a LQTS.
Collapse
Affiliation(s)
- Tina Jenewein
- Institute of Legal Medicine, University of Frankfurt, Frankfurt Am Main, Germany
| | - Scott A Kanner
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Daniel Bauer
- Computational Biology and Simulation Group, Department of Biology, Technische Universita ̈t Darmstadt, Darmstadt, Germany
| | - Brigitte Hertel
- Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Henry M Colecraft
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Anna Moroni
- Department of Biosciences and CNR IBF-Mi, University of Milano, Milano, Italy
| | - Gerhard Thiel
- Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Silke Kauferstein
- Institute of Legal Medicine, University of Frankfurt, Frankfurt Am Main, Germany
| |
Collapse
|
44
|
Ríos-Pérez EB, Liu F, Stevens-Sostre WA, Eichel CA, Silignavong J, Robertson GA. A stable cell line inducibly expressing hERG1a/1b heteromeric channels. J Pharmacol Toxicol Methods 2021; 110:107081. [PMID: 34058320 DOI: 10.1016/j.vascn.2021.107081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 04/07/2021] [Accepted: 05/20/2021] [Indexed: 11/24/2022]
Abstract
Heterologously expressed hERG channels represent a mainstay of in vitro drug safety screens intended to mitigate risk of cardiac IKr block and sudden cardiac death. This is true even as more channel types are adopted as part of the Comprehensive in vitro Proarrhythmia Assay (CiPA) intended to elevate specificity and thus enhance throughput of promising lead drugs. Until now, hERG1a homomeric channels have been used as a proxy for IKr despite a wealth of evidence showing that hERG1a/1b heteromers better represent native channels in terms of protein abundance and channel biophysical and pharmacological properties. Past efforts to create a stable hERG1a/1b cell line were met with unpredictable silencing of hERG1b expression despite stable integration of the gene into the HEK293 cell genome. Here we report a new cell line stably expressing hERG1a, with hERG1b reliably controlled by an inducible promoter sensitive to doxycycline. Co-immunoprecipitation, Western blot analysis and patch-clamp electrophysiology confirm the heteromeric composition of the expressed channels. Association with hERG1b was found to promote hERG1a protein levels and enhance membrane current levels. Optimal conditions for drug screening and experimental investigation were achieved at 24 h exposure to 100 ng/ml doxycycline. Differences in pharmacological sensitivity between homomeric and heteromeric channels were observed for dofetilide and ebastine, but not fluoxetine, as evaluated by their IC50 values. Using these values in the O'Hara-Rudy-CiPA in silico model revealed discrepancies in pro-arrhythmia risk, implying the hERG1a homomeric platform overestimates risk for these two drugs. Dofetilide block was use-dependent and faster for hERG1a/1b than hERG1a channels, whereas ebastine showed considerable block at rest and had a slower progression for hERG1a/1b channels. The hERG1a/1b cell line thus represents an advanced model for contemporary drug safety screening assays such as CiPA that employ IC50 values to estimate risk of proarrhythmia in computational models of ventricular cardiomyocytes. This novel technology fulfills an unmet need to enhance specificity and foster a safe yet expanded drug development pipeline.
Collapse
Affiliation(s)
- Erick B Ríos-Pérez
- Dept. of Neuroscience and Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave. #5505, Madison, WI 53705, United States of America
| | - Fang Liu
- Dept. of Neuroscience and Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave. #5505, Madison, WI 53705, United States of America
| | - Whitney A Stevens-Sostre
- Dept. of Neuroscience and Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave. #5505, Madison, WI 53705, United States of America
| | - Catherine A Eichel
- Dept. of Neuroscience and Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave. #5505, Madison, WI 53705, United States of America
| | - Jonathan Silignavong
- Dept. of Neuroscience and Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave. #5505, Madison, WI 53705, United States of America
| | - Gail A Robertson
- Dept. of Neuroscience and Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave. #5505, Madison, WI 53705, United States of America.
| |
Collapse
|
45
|
DeMarco KR, Yang PC, Singh V, Furutani K, Dawson JRD, Jeng MT, Fettinger JC, Bekker S, Ngo VA, Noskov SY, Yarov-Yarovoy V, Sack JT, Wulff H, Clancy CE, Vorobyov I. Molecular determinants of pro-arrhythmia proclivity of d- and l-sotalol via a multi-scale modeling pipeline. J Mol Cell Cardiol 2021; 158:163-177. [PMID: 34062207 PMCID: PMC8906354 DOI: 10.1016/j.yjmcc.2021.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/03/2021] [Accepted: 05/24/2021] [Indexed: 11/20/2022]
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.
Collapse
Affiliation(s)
- Kevin R DeMarco
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - Pei-Chi Yang
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - Vikrant Singh
- Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Kazuharu Furutani
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Tokushima 770-8514, Japan
| | - John R D Dawson
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Biophysics Graduate Group, University of California Davis, Davis, CA 95616, USA
| | - Mao-Tsuen Jeng
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - James C Fettinger
- Department of Chemistry, University of California Davis, Davis, CA 95616, USA
| | - Slava Bekker
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Science and Engineering, American River College, Sacramento, CA 95841, USA
| | - Van A Ngo
- Centre for Molecular Simulation and Biochemistry Research Cluster, Department of Biological Sciences, University of Calgary, Calgary, AB T2N1N4, Canada
| | - Sergei Y Noskov
- Centre for Molecular Simulation and Biochemistry Research Cluster, Department of Biological Sciences, University of Calgary, Calgary, AB T2N1N4, Canada
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, CA 95616, USA
| | - Jon T Sack
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, CA 95616, USA
| | - Heike Wulff
- Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Colleen E Clancy
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Pharmacology, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
46
|
Huang M, Liao Z, Li X, Yang Z, Fan X, Li Y, Zhao Z, Lang S, Cyganek L, Zhou X, Akin I, Borggrefe M, El-Battrawy I. Effects of Antiarrhythmic Drugs on hERG Gating in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes From a Patient With Short QT Syndrome Type 1. Front Pharmacol 2021; 12:675003. [PMID: 34025432 PMCID: PMC8138577 DOI: 10.3389/fphar.2021.675003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Aims: The short QT syndrome type 1 (SQT1) is linked to hERG channel mutations (e.g., N588K). Drug effects on hERG channel gating kinetics in SQT1-cells have not been investigated. Methods: This study used hiPSC-CMs of a healthy donor and a SQT1-patient carrying the N588K mutation and patch clamp to examine the drug effects on hERG channel gating kinetics. Results: Ajmaline, amiodarone, ivabradine, flecainide, quinidine, mexiletine and ranolazine inhibited the hERG channel current (IKr) less strongly in hiPSC-CMs from the SQTS1-patient (SQT1-hiPSC-CMs) comparing with cells from the healthy donor (donor-hiPSC-CMs). Quinidine and mexiletine reduced, but ajmaline, amiodarone, ivabradine and ranolazine increased the time to peak of IKr similarly in SQT1-hiPSC-CMs and donor-hiPSC-CMs. Although regarding the shift of activation and inactivation curves, tested drugs showed differential effects in donor- and SQT1-hiPSC-CMs, quinidine, ajmaline, ivabradine and mexiletine but not amiodarone, flecainide and ranolazine reduced the window current in SQT1-hiPSC-CMs. Quinidine, ajmaline, ivabradine and mexiletine differentially changed the time constant of recovery from inactivation, but all of them increased the time constant of deactivation in SQT1-hiPSC-CMs. Conclusion: The window current-reducing and deactivation-slowing effects may be important for the antiarrhythmic effect of ajmaline, ivabradine, quinidine and mexiletine in SQT1-cells. This information may be helpful for selecting drugs for treating SQT1-patients with hERG channel mutation.
Collapse
Affiliation(s)
- Mengying Huang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Zhenxing Liao
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,North Sichuan Medical College, Nanchong, China
| | - Xin Li
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Yang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,North Sichuan Medical College, Nanchong, China
| | - Xuehui Fan
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yingrui Li
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Zhihan Zhao
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Siegfried Lang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany.,Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Xiaobo Zhou
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Ibrahim Akin
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Martin Borggrefe
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Ibrahim El-Battrawy
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| |
Collapse
|
47
|
Bell DC, Fermini B. Use of automated patch clamp in cardiac safety assessment: past, present and future perspectives. J Pharmacol Toxicol Methods 2021; 110:107072. [PMID: 33962018 DOI: 10.1016/j.vascn.2021.107072] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/23/2021] [Accepted: 05/02/2021] [Indexed: 12/14/2022]
Abstract
There is no doubt that automated patch clamp (APC) technology has revolutionized research in biomedical science. High throughput ion channel screening is now an integral part of the development and safety profiling of the majority of new chemical entities currently developed to address unmet medical needs. The increased throughput it provides has significantly improved the ability to overcome the time-consuming, low throughput bottlenecks resulting from the more conventional manual patch clamp method, considered the 'gold standard', for studying ion channel function and pharmacology. While systems offering the luxury of automation have only been commercially available for two decades, the road leading to this new technology is long and rich in seminal, hands-on, studies dating back as far as the 18th century. So where does this technology currently stand, and what will it look like in the future? In the current article, we review the scientific history leading to the development of APC systems, examine key drivers in the rapid development of this technology (such as failed ion channel programmes and the issue of drug-induced hERG inhibition and QT interval prolongation), highlight key capabilities and finally provide some perspective on the current and future impact of the technology on cardiac safety assessment and biomedical science.
Collapse
|
48
|
Szendrey M, Guo J, Li W, Yang T, Zhang S. COVID-19 Drugs Chloroquine and Hydroxychloroquine, but Not Azithromycin and Remdesivir, Block hERG Potassium Channels. J Pharmacol Exp Ther 2021; 377:265-272. [PMID: 33674391 DOI: 10.1124/jpet.120.000484] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/02/2021] [Indexed: 12/27/2022] Open
Abstract
Drug-induced long QT syndrome (LQTS) is an established cardiac side effect of a wide range of medications and represents a significant concern for drug safety. The rapidly and slowly activating delayed rectifier K+ currents, mediated by channels encoded by the human ether-a-go-go-related gene (hERG) and KCNQ1 + KCNE1, respectively, are two main currents responsible for ventricular repolarization. The common cause for drugs to induce LQTS is through impairing the hERG channel. For the recent emergence of COVID-19, caused by severe acute respiratory syndrome coronavirus 2, several drugs have been investigated as potential therapies; however, there are concerns about their QT prolongation risk. Here, we studied the effects of chloroquine, hydroxychloroquine, azithromycin, and remdesivir on hERG channels. Our results showed that although chloroquine acutely blocked hERG current (IhERG), with an IC50 of 3.0 µM, hydroxychloroquine acutely blocked IhERG 8-fold less potently, with an IC50 of 23.4 µM. Azithromycin and remdesivir did not acutely affect IhERG When these drugs were added at 10 µM to the cell culture medium for 24 hours, remdesivir increased IhERG by 2-fold, which was associated with an increased mature hERG channel expression. In addition, these four drugs did not acutely or chronically affect KCNQ1 + KCNE1 channels. Our data provide insight into COVID-19 drug-associated LQTS and cardiac safety concerns. SIGNIFICANCE STATEMENT: This work demonstrates that, among off-label potential COVID-19 treatment drugs chloroquine, hydroxychloroquine, azithromycin, and remdesivir, the former two drugs block hERG potassium channels, whereas the latter two drugs do not. All four drugs do not affect KCNQ1 + KCNE1. As hERG and KCNQ1 + KCNE1 are two main K+ channels responsible for ventricular repolarization, and most drugs that induce long QT syndrome (LQTS) do so by impairing hERG channels, these data provide insight into COVID-19 drug-associated LQTS and cardiac safety concerns.
Collapse
Affiliation(s)
- Mark Szendrey
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jun Guo
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Wentao Li
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Tonghua Yang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Shetuan Zhang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
49
|
Hu W, Clark RB, Giles WR, Shibata E, Zhang H. Physiological Roles of the Rapidly Activated Delayed Rectifier K + Current in Adult Mouse Heart Primary Pacemaker Activity. Int J Mol Sci 2021; 22:4761. [PMID: 33946248 PMCID: PMC8124469 DOI: 10.3390/ijms22094761] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 01/01/2023] Open
Abstract
Robust, spontaneous pacemaker activity originating in the sinoatrial node (SAN) of the heart is essential for cardiovascular function. Anatomical, electrophysiological, and molecular methods as well as mathematical modeling approaches have quite thoroughly characterized the transmembrane fluxes of Na+, K+ and Ca2+ that produce SAN action potentials (AP) and 'pacemaker depolarizations' in a number of different in vitro adult mammalian heart preparations. Possible ionic mechanisms that are responsible for SAN primary pacemaker activity are described in terms of: (i) a Ca2+-regulated mechanism based on a requirement for phasic release of Ca2+ from intracellular stores and activation of an inward current-mediated by Na+/Ca2+ exchange; (ii) time- and voltage-dependent activation of Na+ or Ca2+ currents, as well as a cyclic nucleotide-activated current, If; and/or (iii) a combination of (i) and (ii). Electrophysiological studies of single spontaneously active SAN myocytes in both adult mouse and rabbit hearts consistently reveal significant expression of a rapidly activating time- and voltage-dependent K+ current, often denoted IKr, that is selectively expressed in the leading or primary pacemaker region of the adult mouse SAN. The main goal of the present study was to examine by combined experimental and simulation approaches the functional or physiological roles of this K+ current in the pacemaker activity. Our patch clamp data of mouse SAN myocytes on the effects of a pharmacological blocker, E4031, revealed that a rapidly activating K+ current is essential for action potential (AP) repolarization, and its deactivation during the pacemaker potential contributes a small but significant component to the pacemaker depolarization. Mathematical simulations using a murine SAN AP model confirm that well known biophysical properties of a delayed rectifier K+ current can contribute to its role in generating spontaneous myogenic activity.
Collapse
Affiliation(s)
- Wei Hu
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester M13 9PL, UK;
| | - Robert B. Clark
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (R.B.C.); (W.R.G.)
| | - Wayne R. Giles
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (R.B.C.); (W.R.G.)
| | - Erwin Shibata
- Department of Physiology, Carver School of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | - Henggui Zhang
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester M13 9PL, UK;
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
50
|
Functional testing for variant prioritization in a family with long QT syndrome. Mol Genet Genomics 2021; 296:823-836. [PMID: 33876311 DOI: 10.1007/s00438-021-01780-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/29/2021] [Indexed: 01/08/2023]
Abstract
Next-generation sequencing platforms are being increasingly applied in clinical genetic settings for evaluation of families with suspected heritable disease. These platforms potentially improve the diagnostic yield beyond that of disease-specific targeted gene panels, but also increase the number of rare or novel genetic variants that may confound precise diagnostics. Here, we describe a functional testing approach used to interpret the results of whole exome sequencing (WES) in a family presenting with syncope and sudden death. One individual had a prolonged QT interval on electrocardiogram (ECG) and carried a diagnosis of long QT syndrome (LQTS), but a second individual did not meet criteria for LQTS. Filtering WES results for uncommon variants with arrhythmia association identified four for further analyses. In silico analyses indicated that two of these variants, KCNH2 p.(Cys555Arg) and KCNQ1 p.(Arg293Cys), were likely to be causal in this family's LQTS. We subsequently performed functional characterization of these variants in a heterologous expression system. The expression of KCNQ1-Arg293Cys did not show a deleterious phenotype but KCNH2-Cys555Arg demonstrated a loss-of-function phenotype that was partially dominant. Our stepwise approach identified a precise genetic etiology in this family, which resulted in the establishment of a LQTS diagnosis in the second individual as well as an additional asymptomatic family member, enabling personalized clinical management. Given its ability to aid in the diagnosis, the application of functional characterization should be considered as a value adjunct to in silico analyses of WES.
Collapse
|