1
|
Buonocunto M, Lyon A, Delhaas T, Heijman J, Lumens J. Electrophysiological effects of stretch-activated ion channels: a systematic computational characterization. J Physiol 2024; 602:4585-4604. [PMID: 37665242 DOI: 10.1113/jp284439] [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: 01/30/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Cardiac electrophysiology and mechanics are strongly interconnected. Their interaction is, among others, mediated by mechano-electric feedback through stretch-activated ion channels (SACs). The electrophysiological changes induced by SACs may contribute to arrhythmogenesis, but the precise SAC-induced electrophysiological changes remain incompletely understood. Here, we provide a systematic characterization of stretch effects through three distinguished SACs on cardiac electrophysiology using computational modelling. We implemented potassium-selective, calcium-selective and non-selective SACs in the Tomek-Rodriguez-O'Hara-Rudy model of human ventricular electrophysiology. The model was calibrated to experimental data from isolated cardiomyocytes undergoing stretch, considering inter-species differences, and disease-related remodelling of SACs. SAC-mediated effects on the action potential (AP) were analysed by varying stretch amplitude, application timing and/or duration. Afterdepolarizations of different amplitudes were observed with transient 10-ms stretch stimuli of 15-18% applied during phase 4, while stretch ≥18% during phase 4 elicited triggered APs. Longer stimuli shifted the threshold of AP trigger during phase 4 to lower amplitudes, while shorter stimuli increased it. Continuous stretch provoked electrophysiological remodelling. Furthermore, stretch shortened duration or changed morphology of a subsequent electrically evoked AP, and, if applied during a vulnerable time window with sufficient amplitude, prevented its occurrence because of stretch-induced modulation of sodium and L-type calcium channel gating. These effects were more pronounced with disease-related SAC remodelling due to increased stretch sensitivity of diseased hearts. We showed that SACs may induce afterdepolarizations and triggered activities, and prevent subsequent AP generation or change its morphology. These effects depend on cardiomyocyte stretch characteristics and disease-related SACs remodelling and may contribute to cardiac arrhythmogenesis. KEY POINTS: The interplay between cardiac electrophysiology and mechanics is mediated by mechano-electric feedback through stretch-activated ion channels (SACs). These channels may be pro-arrhythmic, but their precise effect on electrophysiology remains unclear. Here we present a systematic in silico characterization of stretch effects through three SACs by implementing inter-species differences as well as disease-related remodelling of SACs in a novel computational model of human ventricular cardiomyocyte electrophysiology. Our simulations showed that, at the cellular level, SACs may provoke electrophysiological remodelling, afterdepolarizations, triggered activities, change the morphology or shorten subsequent electrically evoked action potentials. The model further suggests that a vulnerable window exists in which stretch prevents the following electrically triggered beat occurrence. The pro-arrhythmic effects of stretch strongly depend on disease-related SAC remodelling as well as on stretch characteristics, such as amplitude, time of application and duration. Our study helps in understanding the role of stretch in cardiac arrhythmogenesis and revealing the underlying cellular mechanisms.
Collapse
Affiliation(s)
- Melania Buonocunto
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Aurore Lyon
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Tammo Delhaas
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Joost Lumens
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
2
|
Verkerk L, Verkerk AO, Wilders R. Zebrafish as a Model System for Brugada Syndrome. Rev Cardiovasc Med 2024; 25:313. [PMID: 39355588 PMCID: PMC11440409 DOI: 10.31083/j.rcm2509313] [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: 01/27/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 10/03/2024] Open
Abstract
Brugada syndrome (BrS) is an inheritable cardiac arrhythmogenic disease, associated with an increased risk of sudden cardiac death. It is most common in males around the age of 40 and the prevalence is higher in Asia than in Europe and the United States. The pathophysiology underlying BrS is not completely understood, but several hypotheses have been proposed. So far, the best effective treatment is the implantation of an implantable cardioverter-defibrillator (ICD), but device-related complications are not uncommon. Therefore, there is an urgent need to improve diagnosis and risk stratification and to find new treatment options. To this end, research should further elucidate the genetic basis and pathophysiological mechanisms of BrS. Several experimental models are being used to gain insight into these aspects. The zebrafish (Danio rerio) is a widely used animal model for the study of cardiac arrhythmias, as its cardiac electrophysiology shows interesting similarities to humans. However, zebrafish have only been used in a limited number of studies on BrS, and the potential role of zebrafish in studying the mechanisms of BrS has not been reviewed. Therefore, the present review aims to evaluate zebrafish as an animal model for BrS. We conclude that zebrafish can be considered as a valuable experimental model for BrS research, not only for gene editing technologies, but also for screening potential BrS drugs.
Collapse
Affiliation(s)
- Leonie Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
3
|
Mahardika T NQ, Qauli AI, Marcellinus A, Lim KM. Evaluation of cardiac pro-arrhythmic risks using the artificial neural network with ToR-ORd in silico model output. Front Physiol 2024; 15:1374355. [PMID: 38638275 PMCID: PMC11024991 DOI: 10.3389/fphys.2024.1374355] [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: 01/22/2024] [Accepted: 03/04/2024] [Indexed: 04/20/2024] Open
Abstract
Torsades de pointes (TdP) is a type of ventricular arrhythmia that can lead to sudden cardiac death. Drug-induced TdP has been an important concern for researchers and international regulatory boards. The Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative was proposed that integrates in vitro testing and computational models of cardiac ion channels and human cardiomyocyte cells to evaluate the proarrhythmic risk of drugs. The TdP risk classification performance using only a single TdP metric may require some improvements because of information limitations and the instability of generalizing results. This study evaluates the performance of TdP metrics from the in silico simulations of the Tomek-O'Hara Rudy (ToR-ORd) ventricular cell model for classifying the TdP risk of drugs. We utilized these metrics as an input to an artificial neural network (ANN)-based classifier. The ANN model was optimized through hyperparameter tuning using the grid search (GS) method to find the optimal model. The study outcomes show an area under the curve (AUC) value of 0.979 for the high-risk category, 0.791 for the intermediate-risk category, and 0.937 for the low-risk category. Therefore, this study successfully demonstrates the capability of the ToR-ORd ventricular cell model in classifying the TdP risk into three risk categories, providing new insights into TdP risk prediction methods.
Collapse
Affiliation(s)
- Nurul Qashri Mahardika T
- Computational Medicine Lab, Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, Republic of Korea
| | - Ali Ikhsanul Qauli
- Computational Medicine Lab, Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, Republic of Korea
- Department of Engineering, Faculty of Advanced Technology and Multidiscipline, Universitas Airlangga, Surabaya, Jawa Timur, Indonesia
| | - Aroli Marcellinus
- Computational Medicine Lab, Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, Republic of Korea
| | - Ki Moo Lim
- Computational Medicine Lab, Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, Republic of Korea
- Computational Medicine Lab, Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, Republic of Korea
- Meta Heart Co Ltd., Gumi, Republic of Korea
| |
Collapse
|
4
|
Guerrelli D, Pressman J, Posnack N. hiPSC-CM Electrophysiology: Impact of Temporal Changes and Study Parameters on Experimental Reproducibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560475. [PMID: 37873094 PMCID: PMC10592927 DOI: 10.1101/2023.10.02.560475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are frequently used for preclinical cardiotoxicity testing and remain an important tool for confirming model-based predictions of drug effects in accordance with the Comprehensive in Vitro Proarrhythmia Assay (CiPA) initiative. Despite the considerable benefits hiPSC-CMs provide, concerns surrounding experimental reproducibility have emerged. Our study aimed to investigate the effects of temporal changes and experimental parameters on hiPSC-CM electrophysiology. hiPSC-CMs (iCell cardiomyocyte 2 ) were cultured for 14 days and biosignals were acquired using a microelectrode array (MEA) system. Continuous recordings revealed a 22.6% increase in the beating rate and 7.7% decrease in the field potential duration (FPD) during a 20-minute equilibration period. Location specific differences across a multiwell plate were also observed, with hiPSC-CMs in the outer rows beating 8.8 beats per minute (BPM) faster than the inner rows. Cardiac endpoints were also impacted by cell culture duration; from 2-14 days the beating rate decreased (-12.7 BPM), FPD lengthened (+257 ms), and spike amplitude increased (+3.3 mV). Cell culture duration (4-10 days) also impacted hiPSC-CM drug responsiveness (E-4031, nifedipine, isoproterenol). Our study highlights multiple sources of variability that should be considered and addressed when performing hiPSC-CM MEA studies. To improve reproducibility and data interpretation, MEA-based studies should establish a standardized protocol and report key experimental conditions (e.g., culture time, equilibration time, electrical stimulation settings, report raw data values). New & Noteworthy We demonstrate that hiPSC-CM electrophysiology measurements are significantly impacted by slight deviations in experimental techniques including electrical stimulation protocols, equilibration time, well-to-well variability, and length of hiPSC-CM culture. Furthermore, our results indicate that hiPSC-CM drug responsiveness changes within the first two weeks following defrost.
Collapse
|
5
|
Galow AM, Brenmoehl J, Hoeflich A. Synergistic effects of hormones on structural and functional maturation of cardiomyocytes and implications for heart regeneration. Cell Mol Life Sci 2023; 80:240. [PMID: 37541969 PMCID: PMC10403476 DOI: 10.1007/s00018-023-04894-6] [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: 04/04/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/06/2023]
Abstract
The limited endogenous regenerative capacity of the human heart renders cardiovascular diseases a major health threat, thus motivating intense research on in vitro heart cell generation and cell replacement therapies. However, so far, in vitro-generated cardiomyocytes share a rather fetal phenotype, limiting their utility for drug testing and cell-based heart repair. Various strategies to foster cellular maturation provide some success, but fully matured cardiomyocytes are still to be achieved. Today, several hormones are recognized for their effects on cardiomyocyte proliferation, differentiation, and function. Here, we will discuss how the endocrine system impacts cardiomyocyte maturation. After detailing which features characterize a mature phenotype, we will contemplate hormones most promising to induce such a phenotype, the routes of their action, and experimental evidence for their significance in this process. Due to their pleiotropic effects, hormones might be not only valuable to improve in vitro heart cell generation but also beneficial for in vivo heart regeneration. Accordingly, we will also contemplate how the presented hormones might be exploited for hormone-based regenerative therapies.
Collapse
Affiliation(s)
- Anne-Marie Galow
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany.
| | - Julia Brenmoehl
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Andreas Hoeflich
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| |
Collapse
|
6
|
Seibertz F, Sutanto H, Dülk R, Pronto JRD, Springer R, Rapedius M, Liutkute A, Ritter M, Jung P, Stelzer L, Hüsgen LM, Klopp M, Rubio T, Fakuade FE, Mason FE, Hartmann N, Pabel S, Streckfuss-Bömeke K, Cyganek L, Sossalla S, Heijman J, Voigt N. Electrophysiological and calcium-handling development during long-term culture of human-induced pluripotent stem cell-derived cardiomyocytes. Basic Res Cardiol 2023; 118:14. [PMID: 37020075 PMCID: PMC10076390 DOI: 10.1007/s00395-022-00973-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 04/07/2023]
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are increasingly used for personalised medicine and preclinical cardiotoxicity testing. Reports on hiPSC-CM commonly describe heterogenous functional readouts and underdeveloped or immature phenotypical properties. Cost-effective, fully defined monolayer culture is approaching mainstream adoption; however, the optimal age at which to utilise hiPSC-CM is unknown. In this study, we identify, track and model the dynamic developmental behaviour of key ionic currents and Ca2+-handling properties in hiPSC-CM over long-term culture (30-80 days). hiPSC-CMs > 50 days post differentiation show significantly larger ICa,L density along with an increased ICa,L-triggered Ca2+-transient. INa and IK1 densities significantly increase in late-stage cells, contributing to increased upstroke velocity and reduced action potential duration, respectively. Importantly, our in silico model of hiPSC-CM electrophysiological age dependence confirmed IK1 as the key ionic determinant of action potential shortening in older cells. We have made this model available through an open source software interface that easily allows users to simulate hiPSC-CM electrophysiology and Ca2+-handling and select the appropriate age range for their parameter of interest. This tool, together with the insights from our comprehensive experimental characterisation, could be useful in future optimisation of the culture-to-characterisation pipeline in the field of hiPSC-CM research.
Collapse
Affiliation(s)
- Fitzwilliam Seibertz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Henry Sutanto
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Rebekka Dülk
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Julius Ryan D Pronto
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Robin Springer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | | | - Aiste Liutkute
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Melanie Ritter
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Philipp Jung
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Lea Stelzer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Luisa M Hüsgen
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Marie Klopp
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Tony Rubio
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Funsho E Fakuade
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Fleur E Mason
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Nico Hartmann
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
| | - Steffen Pabel
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Katrin Streckfuss-Bömeke
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
| | - Samuel Sossalla
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
7
|
Agrawal A, Wang K, Polonchuk L, Cooper J, Hendrix M, Gavaghan DJ, Mirams GR, Clerx M. Models of the cardiac L-type calcium current: A quantitative review. WIREs Mech Dis 2023; 15:e1581. [PMID: 36028219 PMCID: PMC10078428 DOI: 10.1002/wsbm.1581] [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: 03/25/2022] [Revised: 06/16/2022] [Accepted: 07/19/2022] [Indexed: 01/31/2023]
Abstract
The L-type calcium current (I CaL ) plays a critical role in cardiac electrophysiology, and models ofI CaL are vital tools to predict arrhythmogenicity of drugs and mutations. Five decades of measuring and modelingI CaL have resulted in several competing theories (encoded in mathematical equations). However, the introduction of new models has not typically been accompanied by a data-driven critical comparison with previous work, so that it is unclear which model is best suited for any particular application. In this review, we describe and compare 73 published mammalianI CaL models and use simulated experiments to show that there is a large variability in their predictions, which is not substantially diminished when grouping by species or other categories. We provide model code for 60 models, list major data sources, and discuss experimental and modeling work that will be required to reduce this huge list of competing theories and ultimately develop a community consensus model ofI CaL . This article is categorized under: Cardiovascular Diseases > Computational Models Cardiovascular Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Aditi Agrawal
- Computational Biology & Health Informatics, Department of Computer ScienceUniversity of OxfordOxfordUK
| | - Ken Wang
- Pharma Research and Early Development, Innovation Center BaselF. Hoffmann‐La Roche Ltd.BaselSwitzerland
| | - Liudmila Polonchuk
- Pharma Research and Early Development, Innovation Center BaselF. Hoffmann‐La Roche Ltd.BaselSwitzerland
| | - Jonathan Cooper
- Centre for Advanced Research ComputingUniversity College LondonLondonUK
| | - Maurice Hendrix
- Centre for Mathematical Medicine & Biology, School of Mathematical SciencesUniversity of NottinghamNottinghamUK
- Digital Research Service, Information SciencesUniversity of NottinghamNottinghamUK
| | - David J. Gavaghan
- Computational Biology & Health Informatics, Department of Computer ScienceUniversity of OxfordOxfordUK
| | - Gary R. Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical SciencesUniversity of NottinghamNottinghamUK
| | - Michael Clerx
- Centre for Mathematical Medicine & Biology, School of Mathematical SciencesUniversity of NottinghamNottinghamUK
| |
Collapse
|
8
|
Kim JE, Kim EM, Lee HA, Kim KS. Effective derivation of ventricular cardiomyocytes from hPSCs using ascorbic acid-containing maturation medium. Anim Cells Syst (Seoul) 2023; 27:82-92. [PMID: 36999134 PMCID: PMC10044166 DOI: 10.1080/19768354.2023.2189932] [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] [Indexed: 04/01/2023] Open
Abstract
Cardiomyocytes derived from human pluripotent stem cells (hPSCs) can be used in various applications including disease modeling, drug safety screening, and novel cell-based cardiac therapies. Here, we report an optimized selection and maturation method to induce maturation of cardiomyocytes into a specific subtype after differentiation driven by the regulation of Wnt signaling. The medium used to optimize selection and maturation was in a glucose starvation conditions, supplemented with either a nutrition complex or ascorbic acid. Following optimized selection and maturation, more cardiac Troponin T (cTnT)-positive cardiomyocytes were detected using albumin and ascorbic acid than B27. In addition, ascorbic acid enriched maturation of ventricular cardiomyocytes. We compared cardiomyocyte-specific gene expression patterns under different selection and maturation conditions by next-generation sequencing (NGS) analysis. Our optimized conditions will enable simple and efficient maturation and specification of the desired cardiomyocyte subtype, facilitating both biomedical research and clinical applications.
Collapse
Affiliation(s)
- Ji-eun Kim
- Dongguk University, Seoul, Republic of Korea
| | - Eun-Mi Kim
- Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Hyang-Ae Lee
- Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Ki-Suk Kim
- Korea Institute of Toxicology, Daejeon, Republic of Korea
- Ki-Suk Kim Korea Institute of Toxicolgoy, 141 Gajeong-ro, Yuseong-gu, Daejeon34114, Republic of Korea
| |
Collapse
|
9
|
Wang L, Wada Y, Ballan N, Schmeckpeper J, Huang J, Rau CD, Wang Y, Gepstein L, Knollmann BC. Triiodothyronine and dexamethasone alter potassium channel expression and promote electrophysiological maturation of human-induced pluripotent stem cell-derived cardiomyocytes. J Mol Cell Cardiol 2021; 161:130-138. [PMID: 34400182 PMCID: PMC9809541 DOI: 10.1016/j.yjmcc.2021.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have emerged as a promising tool for disease modeling and drug development. However, hiPSC-CMs remain functionally immature, which hinders their utility as a model of human cardiomyocytes. OBJECTIVE To improve the electrophysiological maturation of hiPSC-CMs. METHODS AND RESULTS On day 16 of cardiac differentiation, hiPSC-CMs were treated with 100 nmol/L triiodothyronine (T3) and 1 μmol/L Dexamethasone (Dex) or vehicle for 14 days. On day 30, vehicle- and T3 + Dex-treated hiPSC-CMs were dissociated and replated either as cell sheets or single cells. Optical mapping and patch-clamp technique were used to examine the electrophysiological properties of vehicle- and T3 + Dex-treated hiPSC-CMs. Compared to vehicle, T3 + Dex-treated hiPSC-CMs had a slower spontaneous beating rate, more hyperpolarized resting membrane potential, faster maximal upstroke velocity, and shorter action potential duration. Changes in spontaneous activity and action potential were mediated by decreased hyperpolarization-activated current (If) and increased inward rectifier potassium currents (IK1), sodium currents (INa), and the rapidly and slowly activating delayed rectifier potassium currents (IKr and IKs, respectively). Furthermore, T3 + Dex-treated hiPSC-CM cell sheets (hiPSC-CCSs) exhibited a faster conduction velocity and shorter action potential duration than the vehicle. Inhibition of IK1 by 100 μM BaCl2 significantly slowed conduction velocity and prolonged action potential duration in T3 + Dex-treated hiPSC-CCSs but had no effect in the vehicle group, demonstrating the importance of IK1 for conduction velocity and action potential duration. CONCLUSION T3 + Dex treatment is an effective approach to rapidly enhance electrophysiological maturation of hiPSC-CMs.
Collapse
Affiliation(s)
- Lili Wang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, Medical Research Building IV, Rm.1275, 2215B Garland Ave, Nashville, TN 37232, USA,Correspondence to: Lili Wang, Ph.D., Division of Clinical Pharmacology, Vanderbilt University Medical Center, Medical Research Building IV, Rm.1275, 2215B Garland Ave, Nashville, TN 37232-0575 Or Bjorn C. Knollmann, MD, Ph.D., Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Vanderbilt, University Medical Center, Medical Research Building IV, Rm. 1265, 2215B Garland Ave, Nashville, TN 37232-0575,
| | - Yuko Wada
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, Medical Research Building IV, Rm.1275, 2215B Garland Ave, Nashville, TN 37232, USA
| | - Nimer Ballan
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, POB 9649, Haifa 3109601, Israel
| | - Jeffrey Schmeckpeper
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, Medical Research Building IV, Rm.1275, 2215B Garland Ave, Nashville, TN 37232, USA
| | - Jijun Huang
- Department of Anesthesiology, Medicine and Physiology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Christoph Daniel Rau
- Department of Anesthesiology, Medicine and Physiology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Yibin Wang
- Department of Anesthesiology, Medicine and Physiology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Lior Gepstein
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, POB 9649, Haifa 3109601, Israel,Cardiology Department, Rambam Health Care Campus, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, 2 Efron St. POB 9649, Haifa, 3109601, Israel
| | - Bjorn C. Knollmann
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, Medical Research Building IV, Rm.1275, 2215B Garland Ave, Nashville, TN 37232, USA,Correspondence to: Lili Wang, Ph.D., Division of Clinical Pharmacology, Vanderbilt University Medical Center, Medical Research Building IV, Rm.1275, 2215B Garland Ave, Nashville, TN 37232-0575 Or Bjorn C. Knollmann, MD, Ph.D., Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Vanderbilt, University Medical Center, Medical Research Building IV, Rm. 1265, 2215B Garland Ave, Nashville, TN 37232-0575,
| |
Collapse
|
10
|
Van de Sande DV, Kopljar I, Maaike A, Teisman A, Gallacher DJ, Bart L, Snyders DJ, Leybaert L, Lu HR, Labro AJ. The resting membrane potential of hSC-CM in a syncytium is more hyperpolarised than that of isolated cells. Channels (Austin) 2021; 15:239-252. [PMID: 33465001 PMCID: PMC7817136 DOI: 10.1080/19336950.2021.1871815] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/20/2020] [Accepted: 12/31/2020] [Indexed: 01/11/2023] Open
Abstract
Human-induced pluripotent stem cell (hiPSC) and stem cell (hSC) derived cardiomyocytes (CM) are gaining popularity as in vitro model for cardiology and pharmacology studies. A remaining flaw of these cells, as shown by single-cell electrophysiological characterization, is a more depolarized resting membrane potential (RMP) compared to native CM. Most reports attribute this to a lower expression of the Kir2.1 potassium channel that generates the IK1 current. However, most RMP recordings are obtained from isolated hSC/hiPSC-CMs whereas in a more native setting these cells are interconnected with neighboring cells by connexin-based gap junctions, forming a syncytium. Hereby, these cells are electrically connected and the total pool of IK1 increases. Therefore, the input resistance (Ri) of interconnected cells is lower than that of isolated cells. During patch clamp experiments pipettes need to be well attached or sealed to the cell, which is reflected in the seal resistance (Rs), because a nonspecific ionic current can leak through this pipette-cell contact or seal and balance out small currents within the cell such as IK1. By recording the action potential of isolated hSC-CMs and that of hSC-CMs cultured in small monolayers, we show that the RMP of hSC-CMs in monolayer is approximately -20 mV more hyperpolarized compared to isolated cells. Accordingly, adding carbenoxolone, a connexin channel blocker, isolates the cell that is patch clamped from its neighboring cells of the monolayer and depolarizes the RMP. The presented data show that the recorded RMP of hSC-CMs in a syncytium is more negative than that determined from isolated hSC/hiPSC-CMs, most likely because the active pool of Kir2.1 channels increased.
Collapse
Affiliation(s)
| | - Ivan Kopljar
- Global Safety Pharmacology, Non-Clinical Safety, Janssen R&D, Beerse, Belgium
| | - Alaerts Maaike
- Centre of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Ard Teisman
- Global Safety Pharmacology, Non-Clinical Safety, Janssen R&D, Beerse, Belgium
| | - David J. Gallacher
- Global Safety Pharmacology, Non-Clinical Safety, Janssen R&D, Beerse, Belgium
| | - Loeys Bart
- Centre of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Dirk J. Snyders
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Luc Leybaert
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Hua Rong Lu
- Global Safety Pharmacology, Non-Clinical Safety, Janssen R&D, Beerse, Belgium
| | - Alain J. Labro
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
11
|
Altrocchi C, de Korte T, Bernardi J, Spätjens RLHMG, Braam SR, Heijman J, Zaza A, Volders PGA. Repolarization instability and arrhythmia by IKr block in single human-induced pluripotent stem cell-derived cardiomyocytes and 2D monolayers. Europace 2021; 22:1431-1441. [PMID: 32770183 PMCID: PMC7478319 DOI: 10.1093/europace/euaa111] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 04/18/2020] [Indexed: 01/14/2023] Open
Abstract
Aims Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have proven valuable for studies in drug discovery and safety, although limitations regarding their structural and electrophysiological characteristics persist. In this study, we investigated the electrophysiological properties of Pluricyte® CMs, a commercially available hiPSC-CMs line with a ventricular phenotype, and assessed arrhythmia incidence by IKr block at the single-cell and 2D monolayer level. Methods and results Action potentials were measured at different pacing frequencies, using dynamic clamp. Through voltage-clamp experiments, we determined the properties of INa, IKr, and ICaL. Intracellular Ca2+ measurements included Ca2+-transients at baseline and during caffeine perfusion. Effects of IKr block were assessed in single hiPSC-CMs and 2D monolayers (multi-electrode arrays). Action-potential duration (APD) and its rate dependence in Pluricyte® CMs were comparable to those reported for native human CMs. INa, IKr, and ICaL revealed amplitudes, kinetics, and voltage dependence of activation/inactivation similar to other hiPSC-CM lines and, to some extent, to native CMs. Near-physiological Ca2+-induced Ca2+ release, response to caffeine and excitation–contraction coupling gain characterized the cellular Ca2+-handling. Dofetilide prolonged the APD and field-potential duration, and induced early afterdepolarizations. Beat-to-beat variability of repolarization duration increased significantly before the first arrhythmic events in single Pluricyte® CMs and 2D monolayers, and predicted pending arrhythmias better than action-potential prolongation. Conclusion Taking their ion-current characteristics and Ca2+ handling into account, Pluricyte® CMs are suitable for in vitro studies on action potentials and field potentials. Beat-to-beat variability of repolarization duration proved useful to evaluate the dynamics of repolarization instability and demonstrated its significance as proarrhythmic marker in hiPSC-CMs during IKr block.
Collapse
Affiliation(s)
- Cristina Altrocchi
- Department of Cardiology, CARIM, Maastricht University Medical Center+, PO Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Tessa de Korte
- Ncardia, Leiden 2333 BD, The Netherlands.,Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZD Leiden, The Netherlands
| | - Joyce Bernardi
- Department of Cardiology, CARIM, Maastricht University Medical Center+, PO Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Roel L H M G Spätjens
- Department of Cardiology, CARIM, Maastricht University Medical Center+, PO Box 5800, 6202 AZ Maastricht, The Netherlands
| | | | - Jordi Heijman
- Department of Cardiology, CARIM, Maastricht University Medical Center+, PO Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Antonio Zaza
- Laboratory of Cardiac Cellular Physiology, Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy
| | - Paul G A Volders
- Department of Cardiology, CARIM, Maastricht University Medical Center+, PO Box 5800, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
12
|
Varró A, Tomek J, Nagy N, Virág L, Passini E, Rodriguez B, Baczkó I. Cardiac transmembrane ion channels and action potentials: cellular physiology and arrhythmogenic behavior. Physiol Rev 2020; 101:1083-1176. [PMID: 33118864 DOI: 10.1152/physrev.00024.2019] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiac arrhythmias are among the leading causes of mortality. They often arise from alterations in the electrophysiological properties of cardiac cells and their underlying ionic mechanisms. It is therefore critical to further unravel the pathophysiology of the ionic basis of human cardiac electrophysiology in health and disease. In the first part of this review, current knowledge on the differences in ion channel expression and properties of the ionic processes that determine the morphology and properties of cardiac action potentials and calcium dynamics from cardiomyocytes in different regions of the heart are described. Then the cellular mechanisms promoting arrhythmias in congenital or acquired conditions of ion channel function (electrical remodeling) are discussed. The focus is on human-relevant findings obtained with clinical, experimental, and computational studies, given that interspecies differences make the extrapolation from animal experiments to human clinical settings difficult. Deepening the understanding of the diverse pathophysiology of human cellular electrophysiology will help in developing novel and effective antiarrhythmic strategies for specific subpopulations and disease conditions.
Collapse
Affiliation(s)
- András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - Jakub Tomek
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Elisa Passini
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
13
|
Bartolucci C, Passini E, Hyttinen J, Paci M, Severi S. Simulation of the Effects of Extracellular Calcium Changes Leads to a Novel Computational Model of Human Ventricular Action Potential With a Revised Calcium Handling. Front Physiol 2020; 11:314. [PMID: 32351400 PMCID: PMC7174690 DOI: 10.3389/fphys.2020.00314] [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: 12/05/2019] [Accepted: 03/19/2020] [Indexed: 01/13/2023] Open
Abstract
The importance of electrolyte concentrations for cardiac function is well established. Electrolyte variations can lead to arrhythmias onset, due to their important role in the action potential (AP) genesis and in maintaining cell homeostasis. However, most of the human AP computer models available in literature were developed with constant electrolyte concentrations, and fail to simulate physiological changes induced by electrolyte variations. This is especially true for Ca2+, even in the O'Hara-Rudy model (ORd), one of the most widely used models in cardiac electrophysiology. Therefore, the present work develops a new human ventricular model (BPS2020), based on ORd, able to simulate the inverse dependence of AP duration (APD) on extracellular Ca2+ concentration ([Ca2+]o), and APD rate dependence at 4 mM extracellular K+. The main changes needed with respect to ORd are: (i) an increased sensitivity of L-type Ca2+ current inactivation to [Ca2+]o; (ii) a single compartment description of the sarcoplasmic reticulum; iii) the replacement of Ca2+ release. BPS2020 is able to simulate the physiological APD-[Ca2+]o relationship, while also retaining the well-reproduced properties of ORd (APD rate dependence, restitution, accommodation and current block effects). We also used BPS2020 to generate an experimentally-calibrated population of models to investigate: (i) the occurrence of repolarization abnormalities in response to hERG current block; (ii) the rate adaptation variability; (iii) the occurrence of alternans and delayed after-depolarizations at fast pacing. Our results indicate that we successfully developed an improved version of ORd, which can be used to investigate electrophysiological changes and pro-arrhythmic abnormalities induced by electrolyte variations and current block at multiple rates and at the population level.
Collapse
Affiliation(s)
- Chiara Bartolucci
- Computational Physiopathology Unit, Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi”, University of Bologna, Cesena, Italy
| | - Elisa Passini
- Computational Physiopathology Unit, Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi”, University of Bologna, Cesena, Italy
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Jari Hyttinen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Michelangelo Paci
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Stefano Severi
- Computational Physiopathology Unit, Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi”, University of Bologna, Cesena, Italy
| |
Collapse
|
14
|
Big endothelin-1 as a clinical marker for ventricular tachyarrhythmias in patients with post-infarction left ventricular aneurysm. Anatol J Cardiol 2020; 22:256-261. [PMID: 31674930 PMCID: PMC6955057 DOI: 10.14744/anatoljcardiol.2019.67862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Objective: Ventricular tachyarrhythmia is the leading cause of death in post-infarction patients. Big endothelin-1 (ET-1) is a potent vasoconstrictor peptide and plays a role in ventricular tachyarrhythmia development. The aim of this study was to investigate the association between the serum concentration of big ET-1 and ventricular tachyarrhythmia in post-infarction left ventricular aneurysm (PI-LVA) patients. Methods: A total of 222 consecutive PI-LVA patients who had received medical therapy were enrolled in the study. There were 43 (19%) patients who had ventricular tachycardia/ventricular fibrillation (VT/VF) at the time of admission. The clinical characteristics were observed and the plasma big ET-1 level was measured. Associations between big ET-1 and the presence of VT/VF were assessed. Patients were followed up to check for outcomes related to cardiovascular mortality, VT/VF attack, and all-cause mortality. Results: The median concentration of big ET-1 was 0.635 pg/mL. Patients with big ET-1 concentrations above the median were more likely to have higher risk clinical features. There was a positive correlation between the level of big ET-1 with VT/VF attack (r=0.354, p<0.001). In the multiple logistic regression analysis, big ET-1 (OR=4.06, 95%CI:1.77-9.28, p<0.001) appeared as an independent predictive factor for the presence of VT/VF. Multiple Cox regression analysis suggested that big ET-1 concentration was independently predictive of VT/VF attack (OR=2.5, 95% CI 1.4–4.5, p<0.001). NT-proBNP and left ventricular ejection fraction of ≤35% were demonstrated to be independently predictive of cardiovascular mortality and all-cause mortality. Conclusion: Increased big ET-1 concentration in PI-LVA patients is a valuable independent predictor for the prevalence of ventricular tachyarrhythmias and VT/VF attacks during follow-up after PI-LVA treatment.
Collapse
|
15
|
Tomek J, Bueno-Orovio A, Passini E, Zhou X, Minchole A, Britton O, Bartolucci C, Severi S, Shrier A, Virag L, Varro A, Rodriguez B. Development, calibration, and validation of a novel human ventricular myocyte model in health, disease, and drug block. eLife 2019; 8:48890. [PMID: 31868580 PMCID: PMC6970534 DOI: 10.7554/elife.48890] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/18/2019] [Indexed: 12/19/2022] Open
Abstract
Human-based modelling and simulations are becoming ubiquitous in biomedical science due to their ability to augment experimental and clinical investigations. Cardiac electrophysiology is one of the most advanced areas, with cardiac modelling and simulation being considered for virtual testing of pharmacological therapies and medical devices. Current models present inconsistencies with experimental data, which limit further progress. In this study, we present the design, development, calibration and independent validation of a human-based ventricular model (ToR-ORd) for simulations of electrophysiology and excitation-contraction coupling, from ionic to whole-organ dynamics, including the electrocardiogram. Validation based on substantial multiscale simulations supports the credibility of the ToR-ORd model under healthy and key disease conditions, as well as drug blockade. In addition, the process uncovers new theoretical insights into the biophysical properties of the L-type calcium current, which are critical for sodium and calcium dynamics. These insights enable the reformulation of L-type calcium current, as well as replacement of the hERG current model.
Collapse
Affiliation(s)
- Jakub Tomek
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Alfonso Bueno-Orovio
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Elisa Passini
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Xin Zhou
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Ana Minchole
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Oliver Britton
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Chiara Bartolucci
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Bologna, Italy
| | - Stefano Severi
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Bologna, Italy
| | - Alvin Shrier
- Department of Physiology, McGill University, Montreal, Canada
| | - Laszlo Virag
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Andras Varro
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
16
|
Abi-Gerges N, Miller PE, Ghetti A. Human Heart Cardiomyocytes in Drug Discovery and Research: New Opportunities in Translational Sciences. Curr Pharm Biotechnol 2019; 21:787-806. [PMID: 31820682 DOI: 10.2174/1389201021666191210142023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022]
Abstract
In preclinical drug development, accurate prediction of drug effects on the human heart is critically important, whether in the context of cardiovascular safety or for the purpose of modulating cardiac function to treat heart disease. Current strategies have significant limitations, whereby, cardiotoxic drugs can escape detection or potential life-saving therapies are abandoned due to false positive toxicity signals. Thus, new and more reliable translational approaches are urgently needed to help accelerate the rate of new therapy development. Renewed efforts in the recovery of human donor hearts for research and in cardiomyocyte isolation methods, are providing new opportunities for preclinical studies in adult primary cardiomyocytes. These cells exhibit the native physiological and pharmacological properties, overcoming the limitations presented by artificial cellular models, animal models and have great potential for providing an excellent tool for preclinical drug testing. Adult human primary cardiomyocytes have already shown utility in assessing drug-induced cardiotoxicity risk and helping in the identification of new treatments for cardiac diseases, such as heart failure and atrial fibrillation. Finally, strategies with actionable decision-making trees that rely on data derived from adult human primary cardiomyocytes will provide the holistic insights necessary to accurately predict human heart effects of drugs.
Collapse
Affiliation(s)
- Najah Abi-Gerges
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| | - Paul E Miller
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| | - Andre Ghetti
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| |
Collapse
|
17
|
Lemoine MD, Krause T, Koivumäki JT, Prondzynski M, Schulze ML, Girdauskas E, Willems S, Hansen A, Eschenhagen T, Christ T. Human Induced Pluripotent Stem Cell-Derived Engineered Heart Tissue as a Sensitive Test System for QT Prolongation and Arrhythmic Triggers. Circ Arrhythm Electrophysiol 2019; 11:e006035. [PMID: 29925535 DOI: 10.1161/circep.117.006035] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 05/08/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND Cardiac repolarization abnormalities in drug-induced and genetic long-QT syndrome may lead to afterdepolarizations and life-threatening ventricular arrhythmias. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) should help to overcome the limitations of animal models based on species differences in repolarization reserve. Here, we compared head-to-head the contribution of IKs (long QT1) and IKr (long QT2) on action potentials (APs) in human left ventricular (LV) tissue and hiPSC-CM-derived engineered heart tissue (EHT). METHODS APs were measured with sharp microelectrodes in EHT from 3 different control hiPSC-CM lines and in tissue preparations from failing LV. RESULTS EHT from hiPSC-CMs showed spontaneous diastolic depolarization and AP generation that were sensitive to low concentrations of ivabradine. IKr block by E-4031 prolonged AP duration at 90% repolarization with similar half-effective concentration in EHT and LV but larger effect size in EHT (+281 versus +110 ms in LV). Although IKr block alone evoked early afterdepolarizations and triggered activity in 50% of EHTs, slow pacing, reduced extracellular K+, and blocking of IKr, IKs, and IK1 were necessary to induce early afterdepolarizations in LV. In accordance with their clinical safety, moxifloxacin and verapamil did not induce early afterdepolarizations in EHT. In both EHT and LV, IKs block by HMR-1556 prolonged AP duration at 90% repolarization slightly in the combined presence of E-4031 and isoprenaline. CONCLUSIONS EHT from hiPSC-CMs shows a lower repolarization reserve than human LV working myocardium and could thereby serve as a sensitive and specific human-based model for repolarization studies and arrhythmia, similar to Purkinje fibers. In both human LV and EHT, IKs only contributed to repolarization under adrenergic stimulation.
Collapse
Affiliation(s)
- Marc D Lemoine
- Department of Cardiology-Electrophysiology (M.D.L., S.W.) .,Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Tobias Krause
- Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Jussi T Koivumäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (J.T.K.).,BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Finland (J.T.K.)
| | - Maksymilian Prondzynski
- Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Mirja L Schulze
- Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Evaldas Girdauskas
- Department of Cardiovascular Surgery (E.G.), University Heart Center, Hamburg-Eppendorf, Germany
| | - Stephan Willems
- Department of Cardiology-Electrophysiology (M.D.L., S.W.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Arne Hansen
- Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Torsten Christ
- Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.) .,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| |
Collapse
|
18
|
Ross GR, Rizvi F, Emelyanova L, Tajik AJ, Jahangir A. Prolonged post-differentiation culture influences the expression and biophysics of Na + and Ca 2+ channels in induced pluripotent stem cell-derived ventricular-like cardiomyocytes. Cell Tissue Res 2019; 378:59-66. [PMID: 31041505 DOI: 10.1007/s00441-019-03030-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/04/2019] [Indexed: 12/18/2022]
Abstract
Several studies have been reported in various domains from induction methods to utilities of somatic cell pluripotent reprogramming. However, one of the major struggles facing the research field of induced pluripotent stem cell (iPSC)-derived target cells is the lack of consistency in observations. This could be due to variety of reasons including varied culture periods post-differentiation. The cardiomyocytes (CMs) derived from iPSCs are commonly studied and proposed to be utilized in the comprehensive in vitro proarrhythmia initiative for drug safety screening. As the influence of varied culture periods on the electrophysiological properties of iPSC-CMs is not clearly known, using whole-cell patch clamp technique, we compared two groups of differentiated ventricular-like iPSC-CMs that are cultured for 10 to 15 days (D10-15) and more than 30 days (≥ D30) both under current and voltage clamps. The prolonged culture imparts increased excitability with high-frequency spontaneous action potentials, robust increase in the magnitude of peak Na+ current density, relatively shallow inactivation kinetics of Na+ channels, faster recovery from inactivation, and augmented Ca2+ current density. Quantitative real-time PCR studies of α-subunit transcripts showed enhanced mRNA expression of SCN1A, SCN5A Na+ channel subtypes, and CACNA1C, CACNA1G, and CACNA1I Ca2+ channel subtypes, in ≥ D30 group. Conclusively, the prolonged culture of differentiated iPSC-CMs affects the excitability, single-cell electrophysiological properties, and ion channel expressions. Therefore, following standard periods of culture across research studies while utilizing ventricular-like iPSC-CMs for in vitro health/disease modeling to study cellular functional mechanisms or test high-throughput drugs' efficacy and toxicity becomes crucial.
Collapse
Affiliation(s)
- Gracious R Ross
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, 2900 W Oklahoma Ave, Milwaukee, WI, 53215, USA.
| | - Farhan Rizvi
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, 2900 W Oklahoma Ave, Milwaukee, WI, 53215, USA
| | - Larisa Emelyanova
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, 2900 W Oklahoma Ave, Milwaukee, WI, 53215, USA
| | - A Jamil Tajik
- Aurora Cardiovascular Services, Aurora Sinai/St. Luke's Medical Centers, Milwaukee, WI, USA
| | - Arshad Jahangir
- Aurora Cardiovascular Services, Aurora Sinai/St. Luke's Medical Centers, Milwaukee, WI, USA
| |
Collapse
|
19
|
Zhang K, Wu WY, Li G, Zhang YH, Sun Y, Qiu F, Yang Q, Xiao GS, Li GR, Wang Y. Regulation of the TRPC1 channel by endothelin-1 in human atrial myocytes. Heart Rhythm 2019; 16:1575-1583. [PMID: 30954598 DOI: 10.1016/j.hrthm.2019.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND Our recent study demonstrated that the nonselective cation current mediated by the transient receptor potential canonical 1 (TRPC1) channel is activated by endothelin-1 (ET-1) in human atrial myocytes; however, the related signal molecules involved are unknown. OBJECTIVE The purpose of this study was to investigate how the TRPC1 channel is regulated by ET-1 and whether it is upregulated in human atria from patients with atrial fibrillation (AF). METHODS Whole-cell patch technique and molecular biology techniques were used in the study. RESULTS The ET-1-evoked TRPC1 current was inhibited by the ET-1 type A (ETA) receptor antagonist BQ123 and the ET-1 type B (ETB) receptor antagonist BQ788 as well as the protein kinase C inhibitor chelerythrine. ETA receptor-mediated TRPC1 channel activity was selectively inhibited by the phosphoinositide-3-kinase inhibitor wortmannin, while ETB receptor-mediated TRPC1 activity was inhibited by the phospholipase C inhibitor U73122. The messenger RNAs and proteins of the TRPC1 channel and ETA receptor, but not the ETB receptor, were significantly upregulated in atria from patients with AF. The basal TRPC1 current increased in AF myocytes, and the response to ET-1 was greater in AF myocytes than in sinus rhythm myocytes. ET-1 induced a delayed repolarization in 20% of AF myocytes. CONCLUSION These results demonstrate for the first time that TRPC1 activation by ET-1 is mediated by protein kinase C through the distinct phospholipids pathways phosphoinositide-3-kinase and phospholipase C and that the TRPC1 channel and ETA receptor are upregulated in AF atria, which are likely involved in atrial electrical remodeling in patients with AF.
Collapse
Affiliation(s)
- Kai Zhang
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Wei-Yin Wu
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Gang Li
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Yan-Hui Zhang
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Yong Sun
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Feng Qiu
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Qian Yang
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Guo-Sheng Xiao
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China.
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
20
|
Human iPSC-Derived Cardiomyocytes for Investigation of Disease Mechanisms and Therapeutic Strategies in Inherited Arrhythmia Syndromes: Strengths and Limitations. Cardiovasc Drugs Ther 2018; 31:325-344. [PMID: 28721524 PMCID: PMC5550530 DOI: 10.1007/s10557-017-6735-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During the last two decades, significant progress has been made in the identification of genetic defects underlying inherited arrhythmia syndromes, which has provided some clinical benefit through elucidation of gene-specific arrhythmia triggers and treatment. However, for most arrhythmia syndromes, clinical management is hindered by insufficient knowledge of the functional consequences of the mutation in question, the pro-arrhythmic mechanisms involved, and hence the most optimal treatment strategy. Moreover, disease expressivity and sensitivity to therapeutic interventions often varies between mutations and/or patients, underlining the need for more individualized strategies. The development of the induced pluripotent stem cell (iPSC) technology now provides the opportunity for generating iPSC-derived cardiomyocytes (CMs) from human material (hiPSC-CMs), enabling patient- and/or mutation-specific investigations. These hiPSC-CMs may furthermore be employed for identification and assessment of novel therapeutic strategies for arrhythmia syndromes. However, due to their relative immaturity, hiPSC-CMs also display a number of essential differences as compared to adult human CMs, and hence there are certain limitations in their use. We here review the electrophysiological characteristics of hiPSC-CMs, their use for investigating inherited arrhythmia syndromes, and their applicability for identification and assessment of (novel) anti-arrhythmic treatment strategies.
Collapse
|
21
|
Sun X, Nunes SS. Bioengineering Approaches to Mature Human Pluripotent Stem Cell-Derived Cardiomyocytes. Front Cell Dev Biol 2017; 5:19. [PMID: 28337437 PMCID: PMC5343210 DOI: 10.3389/fcell.2017.00019] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/21/2017] [Indexed: 11/26/2022] Open
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CM) represent a potential unlimited cell supply for cardiac tissue engineering and possibly regenerative medicine applications. However, hPSC-CMs produced by current protocols are not representative of native adult human cardiomyocytes as they display immature gene expression profile, structure and function. In order to improve hPSC-CM maturity and function, various approaches have been developed, including genetic manipulations to induce gene expression, delivery of biochemical factors, such as triiodothyronine and alpha-adrenergic agonist phenylephrine, induction of cell alignment in 3D tissues, mechanical stress as a mimic of cardiac load and electrical stimulation/pacing or a combination of these. In this mini review, we discuss biomimetic strategies for the maturation for hPSC-CMs with a particular focus on electromechanical conditioning methods.
Collapse
Affiliation(s)
- Xuetao Sun
- Toronto General Research Institute, University Health Network Toronto, ON, Canada
| | - Sara S Nunes
- Toronto General Research Institute, University Health NetworkToronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of TorontoToronto, ON, Canada; Heart & Stroke/Richard Lewar Centre of Excellence, University of TorontoToronto, ON, Canada
| |
Collapse
|
22
|
Chiamvimonvat N, Chen-Izu Y, Clancy CE, Deschenes I, Dobrev D, Heijman J, Izu L, Qu Z, Ripplinger CM, Vandenberg JI, Weiss JN, Koren G, Banyasz T, Grandi E, Sanguinetti MC, Bers DM, Nerbonne JM. Potassium currents in the heart: functional roles in repolarization, arrhythmia and therapeutics. J Physiol 2017; 595:2229-2252. [PMID: 27808412 DOI: 10.1113/jp272883] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/11/2016] [Indexed: 12/19/2022] Open
Abstract
This is the second of the two White Papers from the fourth UC Davis Cardiovascular Symposium Systems Approach to Understanding Cardiac Excitation-Contraction Coupling and Arrhythmias (3-4 March 2016), a biennial event that brings together leading experts in different fields of cardiovascular research. The theme of the 2016 symposium was 'K+ channels and regulation', and the objectives of the conference were severalfold: (1) to identify current knowledge gaps; (2) to understand what may go wrong in the diseased heart and why; (3) to identify possible novel therapeutic targets; and (4) to further the development of systems biology approaches to decipher the molecular mechanisms and treatment of cardiac arrhythmias. The sessions of the Symposium focusing on the functional roles of the cardiac K+ channel in health and disease, as well as K+ channels as therapeutic targets, were contributed by Ye Chen-Izu, Gideon Koren, James Weiss, David Paterson, David Christini, Dobromir Dobrev, Jordi Heijman, Thomas O'Hara, Crystal Ripplinger, Zhilin Qu, Jamie Vandenberg, Colleen Clancy, Isabelle Deschenes, Leighton Izu, Tamas Banyasz, Andras Varro, Heike Wulff, Eleonora Grandi, Michael Sanguinetti, Donald Bers, Jeanne Nerbonne and Nipavan Chiamvimonvat as speakers and panel discussants. This article summarizes state-of-the-art knowledge and controversies on the functional roles of cardiac K+ channels in normal and diseased heart. We endeavour to integrate current knowledge at multiple scales, from the single cell to the whole organ levels, and from both experimental and computational studies.
Collapse
Affiliation(s)
- Nipavan Chiamvimonvat
- Department of Internal Medicine, University of California, Davis, Genome and Biomedical Science Facility, Rm 6315, Davis, CA, 95616, USA.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, 95655, USA
| | - Ye Chen-Izu
- Department of Internal Medicine, University of California, Davis, Genome and Biomedical Science Facility, Rm 6315, Davis, CA, 95616, USA.,Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA.,Department of Biomedical Engineering, University of California, Davis, Genome and Biomedical Science Facility, Rm 2303, Davis, CA, 95616, USA
| | - Colleen E Clancy
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Isabelle Deschenes
- Department of Physiology and Biophysics, and Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44109, USA.,Heart and Vascular Research Center, MetroHealth Medical Center, Cleveland, OH, 44109, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Leighton Izu
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Zhilin Qu
- Division of Cardiology, Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, 3645 MRL, Los Angeles, CA, 90095, USA
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Jamie I Vandenberg
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW, 2010, Australia
| | - James N Weiss
- Division of Cardiology, Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, 3645 MRL, Los Angeles, CA, 90095, USA
| | - Gideon Koren
- Cardiovascular Research Center, Rhode Island Hospital and the Cardiovascular Institute, The Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Tamas Banyasz
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eleonora Grandi
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Michael C Sanguinetti
- Department of Internal Medicine, University of Utah, Nora Eccles Harrison Cardiovascular Research & Training Institute, Salt Lake City, UT, 84112, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Jeanne M Nerbonne
- Departments of Developmental Biology and Internal Medicine, Cardiovascular Division, Washington University Medical School, St Louis, MO, 63110, USA
| |
Collapse
|
23
|
Bezzerides VJ, Zhang D, Pu WT. Modeling Inherited Arrhythmia Disorders Using Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Circ J 2016; 81:12-21. [PMID: 27916777 DOI: 10.1253/circj.cj-16-1113] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inherited arrhythmia disorders (IADs) are a group of potentially lethal diseases that remain diagnostic and management challenges. Although the genetic basis for many of these disorders is well known, the pathogenicity of individual mutations and the resulting clinical outcomes are difficult to predict. Treatment options remain imperfect, and optimizing therapy for individual patients can be difficult. Recent advances in the derivation of induced pluripotent stem cells (iPSCs) from patients and creation of genetically engineered human models using CRISPR/Cas9 has the potential to dramatically advance translational arrhythmia research. In this review, we discuss the current state of modeling IADs using human iPSC-derived cardiomyocytes. We also discuss current limitations and areas for further study.
Collapse
|
24
|
Carro J, Rodríguez-Matas JF, Monasterio V, Pueyo E. Limitations in electrophysiological model development and validation caused by differences between simulations and experimental protocols. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 129:53-64. [PMID: 27899270 DOI: 10.1016/j.pbiomolbio.2016.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 09/20/2016] [Indexed: 10/20/2022]
Abstract
Models of ion channel dynamics are usually built by fitting isolated cell experimental values of individual parameters while neglecting the interaction between them. Another shortcoming regards the estimation of ionic current conductances, which is often based on quantification of Action Potential (AP)-derived markers. Although this procedure reduces the uncertainty in the calculation of conductances, many studies evaluate electrophysiological AP-derived markers from single cell simulations, whereas experimental measurements are obtained from tissue preparations. In this work, we explore the limitations of these approaches to estimate ion channel dynamics and maximum current conductances and how they could be overcome by using multiscale simulations of experimental protocols. Four human ventricular cell models, namely ten Tusscher and Panfilov (2006), Grandi et al. (2010), O'Hara et al. (2011), and Carro et al. (2011), were used. Two problems involving scales from ion channels to tissue were investigated: 1) characterization of L-type calcium voltage-dependent inactivation ICa,L; 2) identification of major ionic conductance contributors to steady-state AP markers, including APD90, APD75, APD50, APD25, Triangulation and maximal and minimal values of V and dV/dt during the AP (Vmax, Vmin, dV/dtmax, dV/dtmin). Our results show that: 1) ICa,L inactivation characteristics differed significantly when calculated from model equations and from simulations reproducing the experimental protocols. 2) Large differences were found in the ionic currents contributors to APD25, Triangulation, Vmax, dV/dtmax and dV/dtmin between single cells and 1D-tissue. When proposing any new model formulation, or evaluating an existing model, consistency between simulated and experimental data should be verified considering all involved effects and scales.
Collapse
Affiliation(s)
- Jesús Carro
- Universidad San Jorge, Campus Universitario, Autov A23 Km 299, 50830, Villanueva de Gállego, Zaragoza, Spain; Aragon Institute for Engineering Research, Universidad de Zaragoza, Spain; CIBER in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Spain.
| | - José F Rodríguez-Matas
- Aragon Institute for Engineering Research, Universidad de Zaragoza, Spain; LaBS, Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Italy.
| | - Violeta Monasterio
- Universidad San Jorge, Campus Universitario, Autov A23 Km 299, 50830, Villanueva de Gállego, Zaragoza, Spain.
| | - Esther Pueyo
- Aragon Institute for Engineering Research, Universidad de Zaragoza, Spain; CIBER in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Spain.
| |
Collapse
|
25
|
Barbuti A, Benzoni P, Campostrini G, Dell'Era P. Human derived cardiomyocytes: A decade of knowledge after the discovery of induced pluripotent stem cells. Dev Dyn 2016; 245:1145-1158. [DOI: 10.1002/dvdy.24455] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/05/2016] [Accepted: 05/05/2016] [Indexed: 12/27/2022] Open
Affiliation(s)
- Andrea Barbuti
- Department of Biosciences; Università degli Studi di Milano; Milan Italy
| | - Patrizia Benzoni
- Department of Biosciences; Università degli Studi di Milano; Milan Italy
| | - Giulia Campostrini
- Department of Biosciences; Università degli Studi di Milano; Milan Italy
| | - Patrizia Dell'Era
- Cellular Fate Reprogramming Unit, Department of Molecular and Translational Medicine; Università degli Studi di Brescia; Brescia Italy
| |
Collapse
|
26
|
Kirsch GE, Obejero-Paz CA, Bruening-Wright A. Functional Characterization of Human Stem Cell-Derived Cardiomyocytes. ACTA ACUST UNITED AC 2016; 64:11.12.1-26. [PMID: 25152802 DOI: 10.1002/0471141755.ph1112s64] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cardiac toxicity is a leading contributor to late-stage attrition in the drug discovery process and to withdrawal of approved from the market. In vitro assays that enable earlier and more accurate testing for cardiac risk provide early stage predictive indicators that aid in mitigating risk. Human cardiomyocytes, the most relevant subjects for early stage testing, are severely limited in supply. But human stem cell-derived cardiomyocytes (SC-hCM) are readily available from commercial sources and are increasingly used in academic research, drug discovery and safety pharmacology. As a result, SC-hCM electrophysiology has become a valuable tool to assess cardiac risk associated with drugs. This unit describes techniques for recording individual currents carried by sodium, calcium and potassium ions, as well as single cell action potentials, and impedance recordings from contracting syncytia of thousands of interconnected cells.
Collapse
|
27
|
Holzem KM, Gomez JF, Glukhov AV, Madden EJ, Koppel AC, Ewald GA, Trenor B, Efimov IR. Reduced response to IKr blockade and altered hERG1a/1b stoichiometry in human heart failure. J Mol Cell Cardiol 2016; 96:82-92. [PMID: 26093152 PMCID: PMC4683114 DOI: 10.1016/j.yjmcc.2015.06.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 05/27/2015] [Accepted: 06/01/2015] [Indexed: 01/27/2023]
Abstract
Heart failure (HF) claims 250,000 lives per year in the US, and nearly half of these deaths are sudden and presumably due to ventricular tachyarrhythmias. QT interval and action potential (AP) prolongation are hallmark proarrhythmic changes in the failing myocardium, which potentially result from alterations in repolarizing potassium currents. Thus, we aimed to examine whether decreased expression of the rapid delayed rectifier potassium current, IKr, contributes to repolarization abnormalities in human HF. To map functional IKr expression across the left ventricle (LV), we optically imaged coronary-perfused LV free wall from donor and end-stage failing human hearts. The LV wedge preparation was used to examine transmural AP durations at 80% repolarization (APD80), and treatment with the IKr-blocking drug, E-4031, was utilized to interrogate functional expression. We assessed the percent change in APD80 post-IKr blockade relative to baseline APD80 (∆APD80) and found that ∆APD80s are reduced in failing versus donor hearts in each transmural region, with 0.35-, 0.43-, and 0.41-fold reductions in endo-, mid-, and epicardium, respectively (p=0.008, 0.037, and 0.022). We then assessed hERG1 isoform gene and protein expression levels using qPCR and Western blot. While we did not observe differences in hERG1a or hERG1b gene expression between donor and failing hearts, we found a shift in the hERG1a:hERG1b isoform stoichiometry at the protein level. Computer simulations were then conducted to assess IKr block under E-4031 influence in failing and nonfailing conditions. Our results confirmed the experimental observations and E-4031-induced relative APD80 prolongation was greater in normal conditions than in failing conditions, provided that the cellular model of HF included a significant downregulation of IKr. In human HF, the response to IKr blockade is reduced, suggesting decreased functional IKr expression. This attenuated functional response is associated with altered hERG1a:hERG1b protein stoichiometry in the failing human LV, and failing cardiomyoctye simulations support the experimental findings. Thus, of IKr protein and functional expression may be important determinants of repolarization remodeling in the failing human LV.
Collapse
Affiliation(s)
- Katherine M Holzem
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Juan F Gomez
- Polytechnic University of Valencia, Valencia, Spain
| | - Alexey V Glukhov
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Eli J Madden
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Aaron C Koppel
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Gregory A Ewald
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | | | - Igor R Efimov
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA; Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia.
| |
Collapse
|
28
|
van Meer BJ, Tertoolen LGJ, Mummery CL. Concise Review: Measuring Physiological Responses of Human Pluripotent Stem Cell Derived Cardiomyocytes to Drugs and Disease. Stem Cells 2016; 34:2008-15. [PMID: 27250776 PMCID: PMC5113667 DOI: 10.1002/stem.2403] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/25/2016] [Accepted: 05/14/2016] [Indexed: 02/06/2023]
Abstract
Cardiomyocytes from human pluripotent stem cells (hPSC) are of growing interest as models to understand mechanisms underlying genetic disease, identify potential drug targets and for safety pharmacology as they may predict human relevant effects more accurately and inexpensively than animals or other cell models. Crucial to their optimal use are accurate methods to quantify cardiomyocyte phenotypes accurately and reproducibly. Here, we review current methods for determining biophysical parameters of hPSC‐derived cardiomyocytes (hPSC‐CMs) that recapitulate disease and drug responses. Even though hPSC‐CMs as currently available are immature, various biophysical methods are nevertheless already providing useful insights into the biology of the human heart and its maladies. Advantages and limitations of assays currently available looking toward applications of hPSC‐CMs are described with examples of how they have been used to date. This will help guide the choice of biophysical method to characterize healthy cardiomyocytes and their pathologies in vitro. Stem Cells2016;34:2008–2015
Collapse
Affiliation(s)
- Berend J van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Leon G J Tertoolen
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
29
|
Himeno Y, Asakura K, Cha CY, Memida H, Powell T, Amano A, Noma A. A human ventricular myocyte model with a refined representation of excitation-contraction coupling. Biophys J 2016. [PMID: 26200878 DOI: 10.1016/j.bpj.2015.06.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cardiac Ca(2+)-induced Ca(2+) release (CICR) occurs by a regenerative activation of ryanodine receptors (RyRs) within each Ca(2+)-releasing unit, triggered by the activation of L-type Ca(2+) channels (LCCs). CICR is then terminated, most probably by depletion of Ca(2+) in the junctional sarcoplasmic reticulum (SR). Hinch et al. previously developed a tightly coupled LCC-RyR mathematical model, known as the Hinch model, that enables simulations to deal with a variety of functional states of whole-cell populations of a Ca(2+)-releasing unit using a personal computer. In this study, we developed a membrane excitation-contraction model of the human ventricular myocyte, which we call the human ventricular cell (HuVEC) model. This model is a hybrid of the most recent HuVEC models and the Hinch model. We modified the Hinch model to reproduce the regenerative activation and termination of CICR. In particular, we removed the inactivated RyR state and separated the single step of RyR activation by LCCs into triggering and regenerative steps. More importantly, we included the experimental measurement of a transient rise in Ca(2+) concentrations ([Ca(2+)], 10-15 μM) during CICR in the vicinity of Ca(2+)-releasing sites, and thereby calculated the effects of the local Ca(2+) gradient on CICR as well as membrane excitation. This HuVEC model successfully reconstructed both membrane excitation and key properties of CICR. The time course of CICR evoked by an action potential was accounted for by autonomous changes in an instantaneous equilibrium open probability of couplons. This autonomous time course was driven by a core feedback loop including the pivotal local [Ca(2+)], influenced by a time-dependent decay in the SR Ca(2+) content during CICR.
Collapse
Affiliation(s)
- Yukiko Himeno
- Biosimulation Research Center, College of Life Sciences, Ritsumeikan University, Shiga, Japan
| | - Keiichi Asakura
- Biosimulation Research Center, College of Life Sciences, Ritsumeikan University, Shiga, Japan; Nippon Shinyaku Co., Ltd., Kyoto, Japan
| | - Chae Young Cha
- Biosimulation Research Center, College of Life Sciences, Ritsumeikan University, Shiga, Japan; Oxford Centre for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Hiraku Memida
- Biosimulation Research Center, College of Life Sciences, Ritsumeikan University, Shiga, Japan
| | - Trevor Powell
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Akira Amano
- Biosimulation Research Center, College of Life Sciences, Ritsumeikan University, Shiga, Japan
| | - Akinori Noma
- Biosimulation Research Center, College of Life Sciences, Ritsumeikan University, Shiga, Japan.
| |
Collapse
|
30
|
Evaluation of nefazodone-induced cardiotoxicity in human induced pluripotent stem cell-derived cardiomyocytes. Toxicol Appl Pharmacol 2016; 296:42-53. [DOI: 10.1016/j.taap.2016.01.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 01/12/2016] [Accepted: 01/21/2016] [Indexed: 01/10/2023]
|
31
|
Liu XP, Wooltorton JRA, Gaboyard-Niay S, Yang FC, Lysakowski A, Eatock RA. Sodium channel diversity in the vestibular ganglion: NaV1.5, NaV1.8, and tetrodotoxin-sensitive currents. J Neurophysiol 2016; 115:2536-55. [PMID: 26936982 DOI: 10.1152/jn.00902.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 02/02/2016] [Indexed: 01/02/2023] Open
Abstract
Firing patterns differ between subpopulations of vestibular primary afferent neurons. The role of sodium (NaV) channels in this diversity has not been investigated because NaV currents in rodent vestibular ganglion neurons (VGNs) were reported to be homogeneous, with the voltage dependence and tetrodotoxin (TTX) sensitivity of most neuronal NaV channels. RT-PCR experiments, however, indicated expression of diverse NaV channel subunits in the vestibular ganglion, motivating a closer look. Whole cell recordings from acutely dissociated postnatal VGNs confirmed that nearly all neurons expressed NaV currents that are TTX-sensitive and have activation midpoints between -30 and -40 mV. In addition, however, many VGNs expressed one of two other NaV currents. Some VGNs had a small current with properties consistent with NaV1.5 channels: low TTX sensitivity, sensitivity to divalent cation block, and a relatively negative voltage range, and some VGNs showed NaV1.5-like immunoreactivity. Other VGNs had a current with the properties of NaV1.8 channels: high TTX resistance, slow time course, and a relatively depolarized voltage range. In two NaV1.8 reporter lines, subsets of VGNs were labeled. VGNs with NaV1.8-like TTX-resistant current also differed from other VGNs in the voltage dependence of their TTX-sensitive currents and in the voltage threshold for spiking and action potential shape. Regulated expression of NaV channels in primary afferent neurons is likely to selectively affect firing properties that contribute to the encoding of vestibular stimuli.
Collapse
Affiliation(s)
- Xiao-Ping Liu
- Speech and Hearing Bioscience and Technology Program, Harvard-Massachusetts Institute of Technology Health Sciences and Technology Program, Cambridge, Massachusetts; Eaton-Peabody Laboratories, Massachusetts Eye and Ear, and Department of Otology and Laryngology, Harvard Medical School, Boston, Massachusetts
| | | | - Sophie Gaboyard-Niay
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois
| | - Fu-Chia Yang
- Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Neurobiology, Harvard Medical School, Boston, Massachusetts; and
| | - Anna Lysakowski
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois; Department of Otolaryngology-Head and Neck Surgery, University of Illinois at Chicago, Chicago, Illinois
| | - Ruth Anne Eatock
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, and Department of Otology and Laryngology, Harvard Medical School, Boston, Massachusetts; Department of Neurobiology, Harvard Medical School, Boston, Massachusetts; and Department of Otolaryngology-Head and Neck Surgery, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
32
|
Abstract
Gap junctions are channels which allow electrical signals to propagate through the heart from the sinoatrial node and through the atria, conduction system and onwards to the ventricles, and hence are essential for co-ordinated cardiac contraction. Twelve connexin (Cx) proteins make up one gap junction channel, of which there are three main subtypes in the heart; Cx40, Cx43 and Cx45. In the cardiac myocyte, gap junctions are present mainly at the intercalated discs between neighbouring myocytes, and assist in rapid electrical conduction throughout the ventricular myocardium. Fibroblasts provide the structural skeleton of the myocardium and fibroblast numbers significantly increase in heart disease. Fibroblasts also express connexins and this may facilitate heterocellular electrical coupling between myocytes and fibroblasts in the setting of cardiac disease. Interestingly, cardiac fibroblasts have been demonstrated to increase Cx43 expression in experimental models of myocardial infarction and functional gap junctions between myocytes and fibroblasts have been reported. Therefore, in the setting of heart disease enhanced cardiac myocyte: fibroblast coupling may influence the electrical activity of the myocyte and contribute to arrhythmias.
Collapse
|
33
|
Liu J, Laksman Z, Backx PH. The electrophysiological development of cardiomyocytes. Adv Drug Deliv Rev 2016; 96:253-73. [PMID: 26788696 DOI: 10.1016/j.addr.2015.12.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/23/2015] [Accepted: 12/31/2015] [Indexed: 02/07/2023]
Abstract
The generation of human cardiomyocytes (CMs) from human pluripotent stem cells (hPSCs) has become an important resource for modeling human cardiac disease and for drug screening, and also holds significant potential for cardiac regeneration. Many challenges remain to be overcome however, before innovation in this field can translate into a change in the morbidity and mortality associated with heart disease. Of particular importance for the future application of this technology is an improved understanding of the electrophysiologic characteristics of CMs, so that better protocols can be developed and optimized for generating hPSC-CMs. Many different cell culture protocols are currently utilized to generate CMs from hPSCs and all appear to yield relatively “developmentally” immature CMs with highly heterogeneous electrical properties. These hPSC-CMs are characterized by spontaneous beating at highly variable rates with a broad range of depolarization-repolarization patterns, suggestive of mixed populations containing atrial, ventricular and nodal cells. Many recent studies have attempted to introduce approaches to promote maturation and to create cells with specific functional properties. In this review, we summarize the studies in which the electrical properties of CMs derived from stem cells have been examined. In order to place this information in a useful context, we also review the electrical properties of CMs as they transition from the developing embryo to the adult human heart. The signal pathways involved in the regulation of ion channel expression during development are also briefly considered.
Collapse
|
34
|
Dubois VFS, Yu H, Danhof M, Della Pasqua O. Model-based evaluation of drug-induced QTc prolongation for compounds in early development. Br J Clin Pharmacol 2015; 79:148-61. [PMID: 25099645 DOI: 10.1111/bcp.12482] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 08/04/2014] [Indexed: 12/19/2022] Open
Abstract
AIMS Significant differences between dogs and humans have been observed in the concentration-QTc effect relationship of compounds with known pro-arrhythmic properties. These findings suggest that interspecies differences must be considered when evaluating drug effects. The aim of this study was to evaluate the performance of a model-based approach to assess the risk of QTc prolongation for three investigational compounds (NCE01, NCE02 and NCE03). METHODS Pharmacokinetic and pharmacodynamic data from experiments in conscious dogs and healthy subjects were included in this analysis. Pharmacokinetic modelling and deconvolution methods were applied to derive drug concentrations at the time of each QT measurement. An integrated pharmacokinetic-pharmacodynamic (PKPD) model was then used to describe QT prolongation. A threshold of ≥10 ms was used to characterize the probability of QTc prolongation. RESULTS The PKPD relationships of all three compounds were successfully described in both species. A strong effect was observed after administration of NCE01 to dogs and humans, with a slope of 0.0061 and 0.0662 ms nm(-1), respectively, and maximal probability of QTc prolongation ≥10 ms at peak concentration. For NCE02 and NCE03, QTc-shortening and borderline QT effects were observed both in dogs and humans, as described by negative or very shallow slopes (NCE02, -0.00098 and -0.01 ms nm(-1); NCE03, 0.00064 and -0.0002 ms nm(-1)). CONCLUSIONS Whilst NEC01 shows clear pro-arrhythmic effects, the liability for QT/QTc prolongation for NCE02 and NCE03 can be deemed low at the expected therapeutic exposure. Moreover, our results show the advantages of an integrated PKPD approach as the basis for translating pro-arrhythmic effects from dogs to humans.
Collapse
Affiliation(s)
- Vincent F S Dubois
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
35
|
Dubois VFS, de Witte WEA, Visser SAG, Danhof M, Della Pasqua O. Assessment of Interspecies Differences in Drug-Induced QTc Interval Prolongation in Cynomolgus Monkeys, Dogs and Humans. Pharm Res 2015; 33:40-51. [PMID: 26553352 PMCID: PMC4689776 DOI: 10.1007/s11095-015-1760-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 07/16/2015] [Indexed: 11/30/2022]
Abstract
Background and Purpose The selection of the most suitable animal species and subsequent translation of the concentration-effect relationship to humans are critical steps for accurate assessment of the pro-arrhythmic risk of candidate molecules. The objective of this investigation was to assess quantitatively the differences in the QTc prolonging effects of moxifloxacin between cynomolgus monkeys, dogs and humans. The impact of interspecies differences is also illustrated for a new candidate molecule. Experimental Approach Pharmacokinetic data and ECG recordings from pre-clinical protocols in monkeys and dogs and from a phase I trial in healthy subjects were identified for the purpose of this analysis. A previously established Bayesian model describing the combined effect of heart rate, circadian variation and drug effect on the QT interval was used to describe the pharmacokinetic-pharmacodynamic relationships. The probability of a ≥10 ms increase in QT was derived as measure of the pro-arrhythmic effect. Key Results For moxifloxacin, the concentrations associated with a 50% probability of QT prolongation ≥10 ms (Cp50) varied from 20.3 to 6.4 and 2.6 μM in dogs, monkeys and humans, respectively. For NCE05, these values were 0.4 μM vs 2.0 μM for monkeys and humans, respectively. Conclusions and Implications Our findings reveal significant interspecies differences in the QT-prolonging effect of moxifloxacin. In addition to the dissimilarity in pharmacokinetics across species, it is likely that differences in pharmacodynamics also play an important role. It appears that, regardless of the animal model used, a translation function is needed to predict concentration-effect relationships in humans.
Collapse
Affiliation(s)
- V F S Dubois
- Leiden Academic Centre for Drug Research, Division of Pharmacology, Leiden University, Leiden, The Netherlands
| | - W E A de Witte
- Leiden Academic Centre for Drug Research, Division of Pharmacology, Leiden University, Leiden, The Netherlands
| | - S A G Visser
- Global DMPK, AstraZeneca R&D, Sodertalje, Sweden
| | - M Danhof
- Leiden Academic Centre for Drug Research, Division of Pharmacology, Leiden University, Leiden, The Netherlands
| | - O Della Pasqua
- Leiden Academic Centre for Drug Research, Division of Pharmacology, Leiden University, Leiden, The Netherlands. .,Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, Stockley Park, Uxbridge, UK. .,Clinical Pharmacology & Therapeutics, University College London, London, UK.
| | | | | |
Collapse
|
36
|
Birket MJ, Ribeiro MC, Kosmidis G, Ward D, Leitoguinho AR, van de Pol V, Dambrot C, Devalla HD, Davis RP, Mastroberardino PG, Atsma DE, Passier R, Mummery CL. Contractile Defect Caused by Mutation in MYBPC3 Revealed under Conditions Optimized for Human PSC-Cardiomyocyte Function. Cell Rep 2015; 13:733-745. [PMID: 26489474 PMCID: PMC4644234 DOI: 10.1016/j.celrep.2015.09.025] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 07/31/2015] [Accepted: 09/05/2015] [Indexed: 12/23/2022] Open
Abstract
Maximizing baseline function of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is essential for their effective application in models of cardiac toxicity and disease. Here, we aimed to identify factors that would promote an adequate level of function to permit robust single-cell contractility measurements in a human induced pluripotent stem cell (hiPSC) model of hypertrophic cardiomyopathy (HCM). A simple screen revealed the collaborative effects of thyroid hormone, IGF-1 and the glucocorticoid analog dexamethasone on the electrophysiology, bioenergetics, and contractile force generation of hPSC-CMs. In this optimized condition, hiPSC-CMs with mutations in MYBPC3, a gene encoding myosin-binding protein C, which, when mutated, causes HCM, showed significantly lower contractile force generation than controls. This was recapitulated by direct knockdown of MYBPC3 in control hPSC-CMs, supporting a mechanism of haploinsufficiency. Modeling this disease in vitro using human cells is an important step toward identifying therapeutic interventions for HCM.
Collapse
Affiliation(s)
- Matthew J Birket
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Marcelo C Ribeiro
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Georgios Kosmidis
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Dorien Ward
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Ana Rita Leitoguinho
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Vera van de Pol
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Cheryl Dambrot
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Harsha D Devalla
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | | | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Robert Passier
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.
| |
Collapse
|
37
|
Nagy N, Szél T, Jost N, Tóth A, Gy. Papp J, Varró A. Novel experimental results in human cardiac electrophysiology: measurement of the Purkinje fibre action potential from the undiseased human heart. Can J Physiol Pharmacol 2015; 93:803-10. [DOI: 10.1139/cjpp-2014-0532] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Data obtained from canine cardiac electrophysiology studies are often extrapolated to the human heart. However, it has been previously demonstrated that because of the lower density of its K+ currents, the human ventricular action potential has a less extensive repolarization reserve. Since the relevance of canine data to the human heart has not yet been fully clarified, the aim of the present study was to determine for the first time the action potentials of undiseased human Purkinje fibres (PFs) and to compare them directly with those of dog PFs. All measurements were performed at 37 °C using the conventional microelectrode technique. At a stimulation rate of 1 Hz, the plateau potential of human PFs is more positive (8.0 ± 1.8 vs 8.6 ± 3.4 mV, n = 7), while the amplitude of the spike is less pronounced. The maximal rate of depolarization is significantly lower in human PKs than in canine PFs (406.7 ± 62 vs 643 ± 36 V/s, respectively, n = 7). We assume that the appreciable difference in the protein expression profiles of the 2 species may underlie these important disparities. Therefore, caution is advised when canine PF data are extrapolated to humans, and further experiments are required to investigate the characteristics of human PF repolarization and its possible role in arrhythmogenesis.
Collapse
Affiliation(s)
- Norbert Nagy
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - Tamás Szél
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Norbert Jost
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - András Tóth
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Julius Gy. Papp
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - András Varró
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| |
Collapse
|
38
|
Veerman CC, Kosmidis G, Mummery CL, Casini S, Verkerk AO, Bellin M. Immaturity of Human Stem-Cell-Derived Cardiomyocytes in Culture: Fatal Flaw or Soluble Problem? Stem Cells Dev 2015; 24:1035-52. [DOI: 10.1089/scd.2014.0533] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Christiaan C. Veerman
- Department of Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Georgios Kosmidis
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Simona Casini
- Department of Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Arie O. Verkerk
- Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
39
|
Horváth B, Hegyi B, Kistamás K, Váczi K, Bányász T, Magyar J, Szentandrássy N, Nánási PP. Cytosolic calcium changes affect the incidence of early afterdepolarizations in canine ventricular myocytes. Can J Physiol Pharmacol 2015; 93:527-34. [PMID: 25928391 DOI: 10.1139/cjpp-2014-0511] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This study was designed to investigate the influence of cytosolic Ca(2+) levels ([Ca(2+)]i) on action potential duration (APD) and on the incidence of early afterdepolarizations (EADs) in canine ventricular cardiomyocytes. Action potentials (AP) of isolated cells were recorded using conventional sharp microelectrodes, and the concomitant [Ca(2+)]i was monitored with the fluorescent dye Fura-2. EADs were evoked at a 0.2 Hz pacing rate by inhibiting the rapid delayed rectifier K(+) current with dofetilide, by activating the late sodium current with veratridine, or by activating the L-type calcium current with BAY K8644. These interventions progressively prolonged the AP and resulted in initiation of EADs. Reducing [Ca(2+)]i by application of the cell-permeant Ca(2+) chelator BAPTA-AM lengthened the AP at 1.0 Hz if it was applied alone, in the presence of veratridine, or in the presence of BAY K8644. However, BAPTA-AM shortened the AP if the cells were pretreated with dofetilide. The incidence of the evoked EADs was strongly reduced by BAPTA-AM in dofetilide, moderately reduced in veratridine, whereas EAD incidence was increased by BAPTA-AM in the presence of BAY K8644. Based on these experimental data, changes in [Ca(2+)]i have marked effects on APD as well as on the incidence of EADs; however, the underlying mechanisms may be different, depending on the mechanism of EAD generation. As a consequence, reduction of [Ca(2+)]i may eliminate EADs under some, but not all, experimental conditions.
Collapse
Affiliation(s)
- Balázs Horváth
- a Department of Physiology, Faculty of Medicine, University of Debrecen, H-4012 Debrecen, Nagyerdei krt 98, Hungary.,b Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Bence Hegyi
- a Department of Physiology, Faculty of Medicine, University of Debrecen, H-4012 Debrecen, Nagyerdei krt 98, Hungary
| | - Kornél Kistamás
- a Department of Physiology, Faculty of Medicine, University of Debrecen, H-4012 Debrecen, Nagyerdei krt 98, Hungary
| | - Krisztina Váczi
- a Department of Physiology, Faculty of Medicine, University of Debrecen, H-4012 Debrecen, Nagyerdei krt 98, Hungary
| | - Tamás Bányász
- a Department of Physiology, Faculty of Medicine, University of Debrecen, H-4012 Debrecen, Nagyerdei krt 98, Hungary
| | - János Magyar
- a Department of Physiology, Faculty of Medicine, University of Debrecen, H-4012 Debrecen, Nagyerdei krt 98, Hungary.,c Division of Sport Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Norbert Szentandrássy
- a Department of Physiology, Faculty of Medicine, University of Debrecen, H-4012 Debrecen, Nagyerdei krt 98, Hungary.,d Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, Hungary
| | - Péter P Nánási
- a Department of Physiology, Faculty of Medicine, University of Debrecen, H-4012 Debrecen, Nagyerdei krt 98, Hungary.,d Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, Hungary
| |
Collapse
|
40
|
Meijer van Putten RME, Mengarelli I, Guan K, Zegers JG, van Ginneken ACG, Verkerk AO, Wilders R. Ion channelopathies in human induced pluripotent stem cell derived cardiomyocytes: a dynamic clamp study with virtual IK1. Front Physiol 2015; 6:7. [PMID: 25691870 PMCID: PMC4315032 DOI: 10.3389/fphys.2015.00007] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/07/2015] [Indexed: 12/11/2022] Open
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) are widely used in studying basic mechanisms of cardiac arrhythmias that are caused by ion channelopathies. Unfortunately, the action potential profile of hiPSC-CMs-and consequently the profile of individual membrane currents active during that action potential-differs substantially from that of native human cardiomyocytes, largely due to almost negligible expression of the inward rectifier potassium current (IK1). In the present study, we attempted to "normalize" the action potential profile of our hiPSC-CMs by inserting a voltage dependent in silico IK1 into our hiPSC-CMs, using the dynamic clamp configuration of the patch clamp technique. Recordings were made from single hiPSC-CMs, using the perforated patch clamp technique at physiological temperature. We assessed three different models of IK1, with different degrees of inward rectification, and systematically varied the magnitude of the inserted IK1. Also, we modified the inserted IK1 in order to assess the effects of loss- and gain-of-function mutations in the KCNJ2 gene, which encodes the Kir2.1 protein that is primarily responsible for the IK1 channel in human ventricle. For our experiments, we selected spontaneously beating hiPSC-CMs, with negligible IK1 as demonstrated in separate voltage clamp experiments, which were paced at 1 Hz. Upon addition of in silico IK1 with a peak outward density of 4-6 pA/pF, these hiPSC-CMs showed a ventricular-like action potential morphology with a stable resting membrane potential near -80 mV and a maximum upstroke velocity >150 V/s (n = 9). Proarrhythmic action potential changes were observed upon injection of both loss-of-function and gain-of-function IK1, as associated with Andersen-Tawil syndrome type 1 and short QT syndrome type 3, respectively (n = 6). We conclude that injection of in silico IK1 makes the hiPSC-CM a more reliable model for investigating mechanisms underlying cardiac arrhythmias.
Collapse
Affiliation(s)
- Rosalie M E Meijer van Putten
- Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| | - Isabella Mengarelli
- Department of Experimental Cardiology, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| | - Kaomei Guan
- Department of Cardiology and Pneumology, Georg-August-University of Göttingen Göttingen, Germany
| | - Jan G Zegers
- Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| | - Antoni C G van Ginneken
- Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| | - Arie O Verkerk
- Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| | - Ronald Wilders
- Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| |
Collapse
|
41
|
Carro J, Rodriguez JF, Pueyo E. In silico simulations of experimental protocols for cardiac modeling. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2015; 2014:5695-8. [PMID: 25571288 DOI: 10.1109/embc.2014.6944920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A mathematical model of the AP involves the sum of different transmembrane ionic currents and the balance of intracellular ionic concentrations. To each ionic current corresponds an equation involving several effects. There are a number of model parameters that must be identified using specific experimental protocols in which the effects are considered as independent. However, when the model complexity grows, the interaction between effects becomes increasingly important. Therefore, model parameters identified considering the different effects as independent might be misleading. In this work, a novel methodology consisting in performing in silico simulations of the experimental protocol and then comparing experimental and simulated outcomes is proposed for parameter model identification and validation. The potential of the methodology is demonstrated by validating voltage-dependent L-type calcium current (ICaL) inactivation in recently proposed human ventricular AP models with different formulations. Our results show large differences between ICaL inactivation as calculated from the model equation and ICaL inactivation from the in silico simulations due to the interaction between effects and/or to the experimental protocol. Our results suggest that, when proposing any new model formulation, consistency between such formulation and the corresponding experimental data that is aimed at being reproduced needs to be first verified considering all involved factors.
Collapse
|
42
|
EAD and DAD mechanisms analyzed by developing a new human ventricular cell model. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 116:11-24. [PMID: 25192800 DOI: 10.1016/j.pbiomolbio.2014.08.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 08/09/2014] [Indexed: 12/31/2022]
Abstract
It has long been suggested that the Ca(2+)-mechanisms are largely involved in generating the early afterdepolarization (EAD) as well as the delayed afterdepolarization (DAD). This view was examined in a quantitative manner by applying the lead potential analysis to a new human ventricular cell model. In this ventricular cell model, the tight coupled LCC-RyR model (CaRU) based on local control theory (Hinch et al. 2004) and ion channel models mostly based on human electrophysiological data were included to reproduce realistic Ca(2+) dynamics as well as the membrane excitation. Simultaneously, the Ca(2+) accumulation near the Ca(2+) releasing site was incorporated as observed in real cardiac myocytes. The maximum rate of ventricular repolarization (-1.02 mV/ms) is due to IK1 (-0.55 mV/ms) and the rest is provided nearly equally by INCX (-0.20 mV/ms), INaL (-0.16 mV/ms) and INaT (-0.13 mV/ms). These INaL and INaT components are due to closure of the voltage gate, which remains partially open during the plateau potential. DADs could be evoked by applying high-frequency stimulations supplemented by a partial Na(+)/K(+) pump inhibition, or by a microinjection of Ca(2+). EADs was evoked by retarding the inactivation of INaL. The lead potential (VL) analysis revealed that IK1 and IKr played the primary role to reverse the AP repolarization to depolarizing limb of EAD. ICaL and INCX amplified EAD, while the remaining currents partially antagonized dVL/dt. The maximum rate of rise of EAD was attributable to the rapid activation of both ICaL (45.5%) and INCX (54.5%).
Collapse
|
43
|
Contribution of ion currents to beat-to-beat variability of action potential duration in canine ventricular myocytes. Pflugers Arch 2014; 467:1431-1443. [DOI: 10.1007/s00424-014-1581-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/11/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022]
|
44
|
Leyton-Mange JS, Milan DJ. Pluripotent stem cells as a platform for cardiac arrhythmia drug screening. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2014; 16:334. [PMID: 25074263 DOI: 10.1007/s11936-014-0334-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OPINION STATEMENT Since the first demonstrations of the differentiation of pluripotent stem cells to produce functional human cellular models such as cardiomyocytes, the scientific community has been captivated [1, 2••, 3]. In the time since that seminal work, the field has been catapulted forward by the demonstration that adult somatic cells can be reprogrammed to an induced state of pluripotency [4••], and more recently by the development of efficient and sophisticated genome editing tools [5••, 6••, 7], which together afford a theoretically unlimited supply of relevant genetic disease models. In particular, many of the early successes with induced pluripotent stem cell technology have been realized with cardiac arrhythmia syndromes [8••, 9-15]. There is interest in applying stem cell models in large-scale screens to discover novel therapeutics or drug toxicities. This manuscript aims to discuss the potential role of hPSC-derived cardiomyocyte models in therapeutic arrhythmia screens and review recent advances in the field that bring us closer to this reality.
Collapse
|
45
|
Iyer V, Sampson KJ, Kass RS. Modeling tissue- and mutation- specific electrophysiological effects in the long QT syndrome: role of the Purkinje fiber. PLoS One 2014; 9:e97720. [PMID: 24892747 PMCID: PMC4043730 DOI: 10.1371/journal.pone.0097720] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 03/28/2014] [Indexed: 12/05/2022] Open
Abstract
Congenital long QT syndrome is a heritable family of arrhythmias caused by mutations in 13 genes encoding ion channel complex proteins. Mounting evidence has implicated the Purkinje fiber network in the genesis of ventricular arrhythmias. In this study, we explore the hypothesis that long QT mutations can demonstrate different phenotypes depending on the tissue type of expression. Using computational models of the human ventricular myocyte and the Purkinje fiber cell, the biophysical alteration in channel function in LQT1, LQT2, LQT3, and LQT7 are modeled. We identified that the plateau potential was important in LQT1 and LQT2, in which mutation led to minimal action potential prolongation in Purkinje fiber cells. The phenotype of LQT3 mutation was dependent on the biophysical alteration induced as well as tissue type. The canonical ΔKPQ mutation causes severe action potential prolongation in both tissue types. For LQT3 mutation F1473C, characterized by shifted channel availability, a more severe phenotype was seen in Purkinje fiber cells with action potential prolongation and early afterdepolarizations. The LQT3 mutation S1904L demonstrated striking effects on action potential duration restitution and more severe action potential prolongation in Purkinje fiber cells at higher heart rates. Voltage clamp simulations highlight the mechanism of effect of these mutations in different tissue types, and impact of drug therapy is explored. We conclude that arrhythmia formation in long QT syndrome may depend not only on the basis of mutation and biophysical alteration, but also upon tissue of expression. The Purkinje fiber network may represent an important therapeutic target in the management of patients with heritable channelopathies.
Collapse
Affiliation(s)
- Vivek Iyer
- Department of Pharmacology, College of Physicians and Surgeons of Columbia University, New York, New York, United States of America
- * E-mail:
| | - Kevin J. Sampson
- Department of Pharmacology, College of Physicians and Surgeons of Columbia University, New York, New York, United States of America
| | - Robert S. Kass
- Department of Pharmacology, College of Physicians and Surgeons of Columbia University, New York, New York, United States of America
| |
Collapse
|
46
|
Kolettis TM. Ventricular tachyarrhythmias during acute myocardial infarction: the role of endothelin-1. Life Sci 2014; 118:136-40. [PMID: 24486303 DOI: 10.1016/j.lfs.2014.01.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/23/2013] [Accepted: 01/13/2014] [Indexed: 11/16/2022]
Abstract
Ventricular arrhythmogenesis during acute coronary syndromes is a common cause of sudden cardiac death, but the underlying mechanisms remain incompletely understood. Recent evidence indicates an emerging pathophysiologic role of endothelin-1 during myocardial ischaemia and evolving infarction. At the early stages post-coronary occlusion, endothelin-1 enhances sympathetic activation, an effect mediated via the ETA receptor, whereas the ETB receptor exerts protective actions. The importance of this interaction is clearly decreased during subsequent stages, during which endothelin-1 may participate in the genesis of ventricular tachycardia or fibrillation via other mechanisms; of these, the effects of endothelin-1 on repolarizing potassium currents and electrical conduction via gap junctions merit further research. The relative roles of ETA and ETB receptors during this phase are unclear. Evaluation of the arrhythmogenic effects of endothelin-1 during acute coronary syndromes may provide the tools towards lowering sudden cardiac death rates.
Collapse
Affiliation(s)
- Theofilos M Kolettis
- Department of Cardiology, University of Ioannina, Cardiovascular Research Institute, Ioannina and Athens, Greece.
| |
Collapse
|
47
|
Lessons from the heart: mirroring electrophysiological characteristics during cardiac development to in vitro differentiation of stem cell derived cardiomyocytes. J Mol Cell Cardiol 2013; 67:12-25. [PMID: 24370890 DOI: 10.1016/j.yjmcc.2013.12.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 11/14/2013] [Accepted: 12/13/2013] [Indexed: 01/12/2023]
Abstract
The ability of human pluripotent stem cells (hPSCs) to differentiate into any cell type of the three germ layers makes them a very promising cell source for multiple purposes, including regenerative medicine, drug discovery, and as a model to study disease mechanisms and progression. One of the first specialized cell types to be generated from hPSC was cardiomyocytes (CM), and differentiation protocols have evolved over the years and now allow for robust and large-scale production of hPSC-CM. Still, scientists are struggling to achieve the same, mainly ventricular, phenotype of the hPSC-CM in vitro as their adult counterpart in vivo. In vitro generated cardiomyocytes are generally described as fetal-like rather than adult. In this review, we compare the in vivo development of cardiomyocytes to the in vitro differentiation of hPSC into CM with focus on electrophysiology, structure and contractility. Furthermore, known epigenetic changes underlying the differences between adult human CM and CM differentiated from pluripotent stem cells are described. This should provide the reader with an extensive overview of the current status of human stem cell-derived cardiomyocyte phenotype and function. Additionally, the reader will gain insight into the underlying signaling pathways and mechanisms responsible for cardiomyocyte development.
Collapse
|
48
|
Effects of Endothelin-1 Chronic Stimulation on Electrical Restitution, Beat-to-beat Variability of Repolarization, and Ventricular Arrhythmogenesis. J Cardiovasc Pharmacol 2013; 62:549-58. [DOI: 10.1097/fjc.0000000000000015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
49
|
Blazeski A, Zhu R, Hunter DW, Weinberg SH, Zambidis ET, Tung L. Cardiomyocytes derived from human induced pluripotent stem cells as models for normal and diseased cardiac electrophysiology and contractility. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 110:166-77. [PMID: 22971665 PMCID: PMC3910285 DOI: 10.1016/j.pbiomolbio.2012.07.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 12/21/2022]
Abstract
Since the first description of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), these cells have garnered tremendous interest for their potential use in patient-specific analysis and therapy. Additionally, hiPSC-CMs can be derived from donor cells from patients with specific cardiac disorders, enabling in vitro human disease models for mechanistic study and therapeutic drug assessment. However, a full understanding of their electrophysiological and contractile function is necessary before this potential can be realized. Here, we review this emerging field from a functional perspective, with particular emphasis on beating rate, action potential, ionic currents, multicellular conduction, calcium handling and contraction. We further review extant hiPSC-CM disease models that recapitulate genetic myocardial disease.
Collapse
Affiliation(s)
- Adriana Blazeski
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - Renjun Zhu
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - David W. Hunter
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - Seth H. Weinberg
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - Elias T. Zambidis
- Institute for Cell Engineering and Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University, Baltimore, MD
| | - Leslie Tung
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| |
Collapse
|
50
|
Hoekstra M, Mummery CL, Wilde AAM, Bezzina CR, Verkerk AO. Induced pluripotent stem cell derived cardiomyocytes as models for cardiac arrhythmias. Front Physiol 2012; 3:346. [PMID: 23015789 PMCID: PMC3449331 DOI: 10.3389/fphys.2012.00346] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 08/09/2012] [Indexed: 12/20/2022] Open
Abstract
Cardiac arrhythmias are a major cause of morbidity and mortality. In younger patients, the majority of sudden cardiac deaths have an underlying Mendelian genetic cause. Over the last 15 years, enormous progress has been made in identifying the distinct clinical phenotypes and in studying the basic cellular and genetic mechanisms associated with the primary Mendelian (monogenic) arrhythmia syndromes. Investigation of the electrophysiological consequences of an ion channel mutation is ideally done in the native cardiomyocyte (CM) environment. However, the majority of such studies so far have relied on heterologous expression systems in which single ion channel genes are expressed in non-cardiac cells. In some cases, transgenic mouse models have been generated, but these also have significant shortcomings, primarily related to species differences. The discovery that somatic cells can be reprogrammed to pluripotency as induced pluripotent stem cells (iPSC) has generated much interest since it presents an opportunity to generate patient- and disease-specific cell lines from which normal and diseased human CMs can be obtained These genetically diverse human model systems can be studied in vitro and used to decipher mechanisms of disease and identify strategies and reagents for new therapies. Here, we review the present state of the art with respect to cardiac disease models already generated using IPSC technology and which have been (partially) characterized. Human iPSC (hiPSC) models have been described for the cardiac arrhythmia syndromes, including LQT1, LQT2, LQT3-Brugada Syndrome, LQT8/Timothy syndrome and catecholaminergic polymorphic ventricular tachycardia (CPVT). In most cases, the hiPSC-derived cardiomyoctes recapitulate the disease phenotype and have already provided opportunities for novel insight into cardiac pathophysiology. It is expected that the lines will be useful in the development of pharmacological agents for the management of these disorders.
Collapse
Affiliation(s)
- Maaike Hoekstra
- Department of Clinical and Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| | | | | | | | | |
Collapse
|