1
|
Joshi J, Albers C, Smole N, Guo S, Smith SA. Human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) for modeling cardiac arrhythmias: strengths, challenges and potential solutions. Front Physiol 2024; 15:1475152. [PMID: 39328831 PMCID: PMC11424716 DOI: 10.3389/fphys.2024.1475152] [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: 08/02/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024] Open
Abstract
Ion channels and cytoskeletal proteins in the cardiac dyad play a critical role in maintaining excitation-contraction (E-C) coupling and provide cardiac homeostasis. Functional changes in these dyad proteins, whether induced by genetic, epigenetic, metabolic, therapeutic, or environmental factors, can disrupt normal cardiac electrophysiology, leading to abnormal E-C coupling and arrhythmias. Animal models and heterologous cell cultures provide platforms to elucidate the pathogenesis of arrhythmias for basic cardiac research; however, these traditional systems do not truly reflect human cardiac electro-pathophysiology. Notably, patients with the same genetic variants of inherited channelopathies (ICC) often exhibit incomplete penetrance and variable expressivity which underscores the need to establish patient-specific disease models to comprehend the mechanistic pathways of arrhythmias and determine personalized therapies. Patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) inherit the genetic background of the patient and reflect the electrophysiological characteristics of the native cardiomyocytes. Thus, iPSC-CMs provide an innovative and translational pivotal platform in cardiac disease modeling and therapeutic screening. In this review, we will examine how patient-specific iPSC-CMs historically evolved to model arrhythmia syndromes in a dish, and their utility in understanding the role of specific ion channels and their functional characteristics in causing arrhythmias. We will also examine how CRISPR/Cas9 have enabled the establishment of patient-independent and variant-induced iPSC-CMs-based arrhythmia models. Next, we will examine the limitations of using human iPSC-CMs with respect to in vitro arrhythmia modeling that stems from variations in iPSCs or toxicity due to gene editing on iPSC or iPSC-CMs and explore how such hurdles are being addressed. Importantly, we will also discuss how novel 3D iPSC-CM models can better capture in vitro characteristics and how all-optical platforms provide non-invasive and high- throughput electrophysiological data that is useful for stratification of emerging arrhythmogenic variants and drug discovery. Finally, we will examine strategies to improve iPSC-CM maturity, including powerful gene editing and optogenetic tools that can introduce/modify specific ion channels in iPSC-CMs and tailor cellular and functional characteristics. We anticipate that an elegant synergy of iPSCs, novel gene editing, 3D- culture models, and all-optical platforms will offer a high-throughput template to faithfully recapitulate in vitro arrhythmogenic events necessary for personalized arrhythmia monitoring and drug screening process.
Collapse
Affiliation(s)
- Jyotsna Joshi
- Department of Internal Medicine, Division of Cardiology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Cora Albers
- Department of Internal Medicine, Division of Cardiology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Nathan Smole
- Department of Internal Medicine, Division of Cardiology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Shuliang Guo
- Department of Internal Medicine, Division of Cardiology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Sakima A Smith
- Department of Internal Medicine, Division of Cardiology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
2
|
Kistamás K, Lamberto F, Vaiciuleviciute R, Leal F, Muenthaisong S, Marte L, Subías-Beltrán P, Alaburda A, Arvanitis DN, Zana M, Costa PF, Bernotiene E, Bergaud C, Dinnyés A. The Current State of Realistic Heart Models for Disease Modelling and Cardiotoxicity. Int J Mol Sci 2024; 25:9186. [PMID: 39273136 PMCID: PMC11394806 DOI: 10.3390/ijms25179186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
One of the many unresolved obstacles in the field of cardiovascular research is an uncompromising in vitro cardiac model. While primary cell sources from animal models offer both advantages and disadvantages, efforts over the past half-century have aimed to reduce their use. Additionally, obtaining a sufficient quantity of human primary cardiomyocytes faces ethical and legal challenges. As the practically unlimited source of human cardiomyocytes from induced pluripotent stem cells (hiPSC-CM) is now mostly resolved, there are great efforts to improve their quality and applicability by overcoming their intrinsic limitations. The greatest bottleneck in the field is the in vitro ageing of hiPSC-CMs to reach a maturity status that closely resembles that of the adult heart, thereby allowing for more appropriate drug developmental procedures as there is a clear correlation between ageing and developing cardiovascular diseases. Here, we review the current state-of-the-art techniques in the most realistic heart models used in disease modelling and toxicity evaluations from hiPSC-CM maturation through heart-on-a-chip platforms and in silico models to the in vitro models of certain cardiovascular diseases.
Collapse
Affiliation(s)
- Kornél Kistamás
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
| | - Federica Lamberto
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str 1, H-2100 Gödöllő, Hungary
| | - Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
| | - Filipa Leal
- Biofabics Lda, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | | | - Luis Marte
- Digital Health Unit, Eurecat-Centre Tecnològic de Catalunya, 08005 Barcelona, Spain
| | - Paula Subías-Beltrán
- Digital Health Unit, Eurecat-Centre Tecnològic de Catalunya, 08005 Barcelona, Spain
| | - Aidas Alaburda
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
- Institute of Biosciences, Life Sciences Center, Vilnius University, Sauletekio al. 7, LT-10257 Vilnius, Lithuania
| | - Dina N Arvanitis
- Laboratory for Analysis and Architecture of Systems-French National Centre for Scientific Research (LAAS-CNRS), 7 Avenue du Colonel Roche, F-31400 Toulouse, France
| | - Melinda Zana
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
| | - Pedro F Costa
- Biofabics Lda, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
- Faculty of Fundamental Sciences, Vilnius Tech, Sauletekio al. 11, LT-10223 Vilnius, Lithuania
| | - Christian Bergaud
- Laboratory for Analysis and Architecture of Systems-French National Centre for Scientific Research (LAAS-CNRS), 7 Avenue du Colonel Roche, F-31400 Toulouse, France
| | - András Dinnyés
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str 1, H-2100 Gödöllő, Hungary
| |
Collapse
|
3
|
Bartolucci C, Sala L. The Dynamic Clamp Technique: A Robust Toolkit for Investigating Potassium Channel Function. Methods Mol Biol 2024; 2796:211-227. [PMID: 38856904 DOI: 10.1007/978-1-0716-3818-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The dynamic clamp technique has emerged as a powerful tool in the field of cardiac electrophysiology, enabling researchers to investigate the intricate dynamics of ion currents in cardiac cells. Potassium channels play a critical role in the functioning of cardiac cells and the overall electrical stability of the heart. This chapter provides a comprehensive overview of the methods and applications of dynamic clamp in the study of key potassium currents in cardiac cells. A step-by-step guide is presented, detailing the experimental setup and protocols required for implementing the dynamic clamp technique in cardiac cell studies. Special attention is given to the design and construction of a dynamic clamp setup with Real Time eXperimental Interface, configurations, and the incorporation of mathematical models to mimic ion channel behavior. The chapter's core focuses on applying dynamic clamp to elucidate the properties of various potassium channels in cardiac cells. It discusses how dynamic clamp can be used to investigate channel kinetics, voltage-dependent properties, and the impact of different potassium channel subtypes on cardiac electrophysiology. The chapter will also include examples of specific dynamic clamp experiments that studied potassium currents or their applications in cardiac cells.
Collapse
Affiliation(s)
- Chiara Bartolucci
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy.
| | - Luca Sala
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
| |
Collapse
|
4
|
Verkerk AO, Wilders R. Injection of I K1 through dynamic clamp can make all the difference in patch-clamp studies on hiPSC-derived cardiomyocytes. Front Physiol 2023; 14:1326160. [PMID: 38152247 PMCID: PMC10751953 DOI: 10.3389/fphys.2023.1326160] [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: 10/22/2023] [Accepted: 11/24/2023] [Indexed: 12/29/2023] Open
Abstract
Human-induced stem cell-derived cardiomyocytes (hiPSC-CMs) are a valuable tool for studying development, pharmacology, and (inherited) arrhythmias. Unfortunately, hiPSC-CMs are depolarized and spontaneously active, even the working cardiomyocyte subtypes such as atrial- and ventricular-like hiPSC-CMs, in contrast to the situation in the atria and ventricles of adult human hearts. Great efforts have been made, using many different strategies, to generate more mature, quiescent hiPSC-CMs with more close-to-physiological resting membrane potentials, but despite promising results, it is still difficult to obtain hiPSC-CMs with such properties. The dynamic clamp technique allows to inject a current with characteristics of the inward rectifier potassium current (IK1), computed in real time according to the actual membrane potential, into patch-clamped hiPSC-CMs during action potential measurements. This results in quiescent hiPSC-CMs with a close-to-physiological resting membrane potential. As a result, action potential measurements can be performed with normal ion channel availability, which is particularly important for the physiological functioning of the cardiac SCN5A-encoded fast sodium current (INa). We performed in vitro and in silico experiments to assess the beneficial effects of the dynamic clamp technique in dissecting the functional consequences of the SCN5A-1795insD+/- mutation. In two separate sets of patch-clamp experiments on control hiPSC-CMs and on hiPSC-CMs with mutations in ACADVL and GNB5, we assessed the value of dynamic clamp in detecting delayed afterdepolarizations and in investigating factors that modulate the resting membrane potential. We conclude that the dynamic clamp technique has highly beneficial effects in all of the aforementioned settings and should be widely used in patch-clamp studies on hiPSC-CMs while waiting for the ultimate fully mature hiPSC-CMs.
Collapse
Affiliation(s)
- Arie O. Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
5
|
Reisqs JB, Moreau A, Sleiman Y, Boutjdir M, Richard S, Chevalier P. Arrhythmogenic cardiomyopathy as a myogenic disease: highlights from cardiomyocytes derived from human induced pluripotent stem cells. Front Physiol 2023; 14:1191965. [PMID: 37250123 PMCID: PMC10210147 DOI: 10.3389/fphys.2023.1191965] [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: 03/22/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiomyopathy characterized by the replacement of myocardium by fibro-fatty infiltration and cardiomyocyte loss. ACM predisposes to a high risk for ventricular arrhythmias. ACM has initially been defined as a desmosomal disease because most of the known variants causing the disease concern genes encoding desmosomal proteins. Studying this pathology is complex, in particular because human samples are rare and, when available, reflect the most advanced stages of the disease. Usual cellular and animal models cannot reproduce all the hallmarks of human pathology. In the last decade, human-induced pluripotent stem cells (hiPSC) have been proposed as an innovative human cellular model. The differentiation of hiPSCs into cardiomyocytes (hiPSC-CM) is now well-controlled and widely used in many laboratories. This hiPSC-CM model recapitulates critical features of the pathology and enables a cardiomyocyte-centered comprehensive approach to the disease and the screening of anti-arrhythmic drugs (AAD) prescribed sometimes empirically to the patient. In this regard, this model provides unique opportunities to explore and develop new therapeutic approaches. The use of hiPSC-CMs will undoubtedly help the development of precision medicine to better cure patients suffering from ACM. This review aims to summarize the recent advances allowing the use of hiPSCs in the ACM context.
Collapse
Affiliation(s)
- J. B. Reisqs
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
| | - A. Moreau
- Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, PhyMedExp, Montpellier, France
| | - Y. Sleiman
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
| | - M. Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Sciences University, NY, United States
- Department of Medicine, New York University School of Medicine, NY, United States
| | - S. Richard
- Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, PhyMedExp, Montpellier, France
| | - P. Chevalier
- Neuromyogene Institute, Claude Bernard University, Lyon 1, Villeurbanne, France
- Service de Rythmologie, Hospices Civils de Lyon, Lyon, France
| |
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: 4.5] [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
|
Oeing CU, Pepin ME, Saul KB, Agircan AS, Assenov Y, Merkel TS, Sedaghat-Hamedani F, Weis T, Meder B, Guan K, Plass C, Weichenhan D, Siede D, Backs J. Indirect epigenetic testing identifies a diagnostic signature of cardiomyocyte DNA methylation in heart failure. Basic Res Cardiol 2023; 118:9. [PMID: 36939901 PMCID: PMC10027651 DOI: 10.1007/s00395-022-00954-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/06/2022] [Accepted: 09/15/2022] [Indexed: 03/21/2023]
Abstract
Precision-based molecular phenotyping of heart failure must overcome limited access to cardiac tissue. Although epigenetic alterations have been found to underlie pathological cardiac gene dysregulation, the clinical utility of myocardial epigenomics remains narrow owing to limited clinical access to tissue. Therefore, the current study determined whether patient plasma confers indirect phenotypic, transcriptional, and/or epigenetic alterations to ex vivo cardiomyocytes to mirror the failing human myocardium. Neonatal rat ventricular myocytes (NRVMs) and single-origin human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and were treated with blood plasma samples from patients with dilated cardiomyopathy (DCM) and donor subjects lacking history of cardiovascular disease. Following plasma treatments, NRVMs and hiPSC-CMs underwent significant hypertrophy relative to non-failing controls, as determined via automated high-content screening. Array-based DNA methylation analysis of plasma-treated hiPSC-CMs and cardiac biopsies uncovered robust, and conserved, alterations in cardiac DNA methylation, from which 100 sites were validated using an independent cohort. Among the CpG sites identified, hypo-methylation of the ATG promoter was identified as a diagnostic marker of HF, wherein cg03800765 methylation (AUC = 0.986, P < 0.0001) was found to out-perform circulating NT-proBNP levels in differentiating heart failure. Taken together, these findings support a novel approach of indirect epigenetic testing in human HF.
Collapse
Affiliation(s)
- Christian U Oeing
- Institute of Experimental Cardiology, University Hospital Heidelberg, University of Heidelberg and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
- Department of Internal Medicine and Cardiology, Charité University Medicine, DZHK (German Center for Cardiovascular Research), Partner site Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Mark E Pepin
- Institute of Experimental Cardiology, University Hospital Heidelberg, University of Heidelberg and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Kerstin B Saul
- Institute of Experimental Cardiology, University Hospital Heidelberg, University of Heidelberg and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Ayça Seyhan Agircan
- Institute of Experimental Cardiology, University Hospital Heidelberg, University of Heidelberg and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Yassen Assenov
- Cancer Epigenomics, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Tobias S Merkel
- Institute of Experimental Cardiology, University Hospital Heidelberg, University of Heidelberg and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Farbod Sedaghat-Hamedani
- Department of Cardiology, University of Heidelberg, DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Tanja Weis
- Department of Cardiology, University of Heidelberg, DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Benjamin Meder
- Department of Cardiology, University of Heidelberg, DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Medical Centre Dresden, Dresden, Germany
| | - Christoph Plass
- Cancer Epigenomics, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Dieter Weichenhan
- Cancer Epigenomics, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Dominik Siede
- Institute of Experimental Cardiology, University Hospital Heidelberg, University of Heidelberg and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Johannes Backs
- Institute of Experimental Cardiology, University Hospital Heidelberg, University of Heidelberg and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany.
| |
Collapse
|
8
|
Altomare C, Bartolucci C, Sala L, Balbi C, Burrello J, Pietrogiovanna N, Burrello A, Bolis S, Panella S, Arici M, Krause R, Rocchetti M, Severi S, Barile L. A dynamic clamping approach using in silico IK1 current for discrimination of chamber-specific hiPSC-derived cardiomyocytes. Commun Biol 2023; 6:291. [PMID: 36934210 PMCID: PMC10024709 DOI: 10.1038/s42003-023-04674-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 03/07/2023] [Indexed: 03/20/2023] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CM) constitute a mixed population of ventricular-, atrial-, nodal-like cells, limiting the reliability for studying chamber-specific disease mechanisms. Previous studies characterised CM phenotype based on action potential (AP) morphology, but the classification criteria were still undefined. Our aim was to use in silico models to develop an automated approach for discriminating the electrophysiological differences between hiPSC-CM. We propose the dynamic clamp (DC) technique with the injection of a specific IK1 current as a tool for deriving nine electrical biomarkers and blindly classifying differentiated CM. An unsupervised learning algorithm was applied to discriminate CM phenotypes and principal component analysis was used to visualise cell clustering. Pharmacological validation was performed by specific ion channel blocker and receptor agonist. The proposed approach improves the translational relevance of the hiPSC-CM model for studying mechanisms underlying inherited or acquired atrial arrhythmias in human CM, and for screening anti-arrhythmic agents.
Collapse
Affiliation(s)
- Claudia Altomare
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Euler institute, Università Svizzera italiana, Lugano, Switzerland
| | - Chiara Bartolucci
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | - Luca Sala
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Carolina Balbi
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Jacopo Burrello
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Division of Internal Medicine 4 and Hypertension Unit, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Nicole Pietrogiovanna
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Alessio Burrello
- Department of Electrical, Electronic and Information Engineering (DEI), University of Bologna, Bologna, Italy
| | - Sara Bolis
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Stefano Panella
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Martina Arici
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Rolf Krause
- Euler institute, Università Svizzera italiana, Lugano, Switzerland
| | - Marcella Rocchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Stefano Severi
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy.
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland.
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
- Euler institute, Università Svizzera italiana, Lugano, Switzerland.
- Faculty of Biomedical Sciences, Università Svizzera italiana, Lugano, Switzerland.
- Institute of Life Science, Scuola Superiore Sant'Anna, Pisa, Italy.
| |
Collapse
|
9
|
Cellular electrophysiological effects of botulinum toxin A on neonatal rat cardiomyocytes and on cardiomyocytes derived from human-induced pluripotent stem cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:513-524. [PMID: 36399184 DOI: 10.1007/s00210-022-02332-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022]
Abstract
Botulinum toxin A is a well-known neurotransmitter inhibitor with a wide range of applications in modern medicine. Recently, botulinum toxin A preparations have been used in clinical trials to suppress cardiac arrhythmias, especially in the postoperative period. Its antiarrhythmic action is associated with inhibition of the nervous system of the heart, but its direct effect on heart tissue remains unclear. Accordingly, we investigate the effect of botulinum toxin A on isolated cardiac cells and on layers of cardiac cells capable of conducting excitation. Cardiomyocytes of neonatal rat pups and human cardiomyocytes obtained through cell reprogramming were used. A patch-clamp study showed that botulinum toxin A inhibited fast sodium currents and L-type calcium currents in a dose-dependent manner, with no apparent effect on potassium currents. Optical mapping showed that in the presence of botulinum toxin A, the propagation of the excitation wave in the layer of cardiac cells slows down sharply, conduction at high concentrations becomes chaotic, but reentry waves do not form. The combination of botulinum toxin A with a preparation of chitosan showed a stronger inhibitory effect by an order of magnitude. Further, the inhibitory effect of botulinum toxin A is not permanent and disappears after 12 days of cell culture in a botulinum toxin A-free medium. The main conclusion of the work is that the antiarrhythmic effect of botulinum toxin A found in clinical studies is associated not only with depression of the nervous system but also with a direct effect on heart tissue. Moreover, the combination of botulinum toxin A and chitosan reduces the effective dose of botulinum toxin A.
Collapse
|
10
|
Verkerk AO, Doszpod IJ, Mengarelli I, Magyar T, Polyák A, Pászti B, Efimov IR, Wilders R, Koncz I. Acetylcholine Reduces L-Type Calcium Current without Major Changes in Repolarization of Canine and Human Purkinje and Ventricular Tissue. Biomedicines 2022; 10:biomedicines10112987. [PMID: 36428555 PMCID: PMC9687254 DOI: 10.3390/biomedicines10112987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Vagal nerve stimulation (VNS) holds a strong basis as a potentially effective treatment modality for chronic heart failure, which explains why a multicenter VNS study in heart failure with reduced ejection fraction is ongoing. However, more detailed information is required on the effect of acetylcholine (ACh) on repolarization in Purkinje and ventricular cardiac preparations to identify the advantages, risks, and underlying cellular mechanisms of VNS. Here, we studied the effect of ACh on the action potential (AP) of canine Purkinje fibers (PFs) and several human ventricular preparations. In addition, we characterized the effects of ACh on the L-type Ca2+ current (ICaL) and AP of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and performed computer simulations to explain the observed effects. Using microelectrode recordings, we found a small but significant AP prolongation in canine PFs. In the human myocardium, ACh slightly prolonged the AP in the midmyocardium but resulted in minor AP shortening in subepicardial tissue. Perforated patch-clamp experiments on hiPSC-CMs demonstrated that 5 µM ACh caused an ≈15% decrease in ICaL density without changes in gating properties. Using dynamic clamp, we found that under blocked K+ currents, 5 µM ACh resulted in an ≈23% decrease in AP duration at 90% of repolarization in hiPSC-CMs. Computer simulations using the O'Hara-Rudy human ventricular cell model revealed that the overall effect of ACh on AP duration is a tight interplay between the ACh-induced reduction in ICaL and ACh-induced changes in K+ currents. In conclusion, ACh results in minor changes in AP repolarization and duration of canine PFs and human ventricular myocardium due to the concomitant inhibition of inward ICaL and outward K+ currents, which limits changes in net repolarizing current and thus prevents major changes in AP repolarization.
Collapse
Affiliation(s)
- Arie O. Verkerk
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Illés J. Doszpod
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Isabella Mengarelli
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Tibor Magyar
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Alexandra Polyák
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Bence Pászti
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Igor R. Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Biomedical Engineering, Northwestern University, Chicago, IL 60611, USA
- Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Correspondence: (R.W.); (I.K.)
| | - István Koncz
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
- Correspondence: (R.W.); (I.K.)
| |
Collapse
|
11
|
Portero V, Nicol T, Podliesna S, Marchal GA, Baartscheer A, Casini S, Tadros R, Treur JL, Tanck MWT, Cox IJ, Probert F, Hough TA, Falcone S, Beekman L, Müller-Nurasyid M, Kastenmüller G, Gieger C, Peters A, Kääb S, Sinner MF, Blease A, Verkerk AO, Bezzina CR, Potter PK, Remme CA. Chronically elevated branched chain amino acid levels are pro-arrhythmic. Cardiovasc Res 2022; 118:1742-1757. [PMID: 34142125 PMCID: PMC9215196 DOI: 10.1093/cvr/cvab207] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/16/2021] [Indexed: 01/03/2023] Open
Abstract
AIMS Cardiac arrhythmias comprise a major health and economic burden and are associated with significant morbidity and mortality, including cardiac failure, stroke, and sudden cardiac death (SCD). Development of efficient preventive and therapeutic strategies is hampered by incomplete knowledge of disease mechanisms and pathways. Our aim is to identify novel mechanisms underlying cardiac arrhythmia and SCD using an unbiased approach. METHODS AND RESULTS We employed a phenotype-driven N-ethyl-N-nitrosourea mutagenesis screen and identified a mouse line with a high incidence of sudden death at young age (6-9 weeks) in the absence of prior symptoms. Affected mice were found to be homozygous for the nonsense mutation Bcat2p.Q300*/p.Q300* in the Bcat2 gene encoding branched chain amino acid transaminase 2. At the age of 4-5 weeks, Bcat2p.Q300*/p.Q300* mice displayed drastic increase of plasma levels of branch chain amino acids (BCAAs-leucine, isoleucine, valine) due to the incomplete catabolism of BCAAs, in addition to inducible arrhythmias ex vivo as well as cardiac conduction and repolarization disturbances. In line with these findings, plasma BCAA levels were positively correlated to electrocardiogram indices of conduction and repolarization in the German community-based KORA F4 Study. Isolated cardiomyocytes from Bcat2p.Q300*/p.Q300* mice revealed action potential (AP) prolongation, pro-arrhythmic events (early and late afterdepolarizations, triggered APs), and dysregulated calcium homeostasis. Incubation of human pluripotent stem cell-derived cardiomyocytes with elevated concentration of BCAAs induced similar calcium dysregulation and pro-arrhythmic events which were prevented by rapamycin, demonstrating the crucial involvement of mTOR pathway activation. CONCLUSIONS Our findings identify for the first time a causative link between elevated BCAAs and arrhythmia, which has implications for arrhythmogenesis in conditions associated with BCAA metabolism dysregulation such as diabetes, metabolic syndrome, and heart failure.
Collapse
Affiliation(s)
- Vincent Portero
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Thomas Nicol
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire, UK
| | - Svitlana Podliesna
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Gerard A Marchal
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Antonius Baartscheer
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Simona Casini
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Rafik Tadros
- Cardiovascular Genetics Center, Montreal Heart Institute and Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Jorien L Treur
- Department of Psychiatry, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Michael W T Tanck
- Amsterdam UMC, University of Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health (APH), The Netherlands
| | - I Jane Cox
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, Kings College, London, UK
| | - Fay Probert
- Department of Chemistry, University of Oxford, Oxford, UK
| | - Tertius A Hough
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire, UK
| | - Sara Falcone
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire, UK
| | - Leander Beekman
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Martina Müller-Nurasyid
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- IBE, Faculty of Medicine, Ludwig Maximilian’s University (LMU) Munich, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site: Munich Heart Alliance, Munich, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site: Munich Heart Alliance, Munich, Germany
| | - Stefan Kääb
- German Centre for Cardiovascular Research (DZHK), Partner Site: Munich Heart Alliance, Munich, Germany
- Department of Medicine I (Cardiology), University Hospital, LMU Munich, Munich, Germany
| | - Moritz F Sinner
- German Centre for Cardiovascular Research (DZHK), Partner Site: Munich Heart Alliance, Munich, Germany
- Department of Medicine I (Cardiology), University Hospital, LMU Munich, Munich, Germany
| | - Andrew Blease
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire, UK
| | - Arie O Verkerk
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Connie R Bezzina
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Paul K Potter
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK
| | - Carol Ann Remme
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, Location AMC, Room K2-104.2, Meibergdreef 9, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| |
Collapse
|
12
|
Song Y, Zheng Z, Lian J. Deciphering Common Long QT Syndrome Using CRISPR/Cas9 in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Front Cardiovasc Med 2022; 9:889519. [PMID: 35647048 PMCID: PMC9136094 DOI: 10.3389/fcvm.2022.889519] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
From carrying potentially pathogenic genes to severe clinical phenotypes, the basic research in the inherited cardiac ion channel disease such as long QT syndrome (LQTS) has been a significant challenge in explaining gene-phenotype heterogeneity. These have opened up new pathways following the parallel development and successful application of stem cell and genome editing technologies. Stem cell-derived cardiomyocytes and subsequent genome editing have allowed researchers to introduce desired genes into cells in a dish to replicate the disease features of LQTS or replace causative genes to normalize the cellular phenotype. Importantly, this has made it possible to elucidate potential genetic modifiers contributing to clinical heterogeneity and hierarchically manage newly identified variants of uncertain significance (VUS) and more therapeutic options to be tested in vitro. In this paper, we focus on and summarize the recent advanced application of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) combined with clustered regularly interspaced short palindromic repeats/CRISPR-associated system 9 (CRISPR/Cas9) in the interpretation for the gene-phenotype relationship of the common LQTS and presence challenges, increasing our understanding of the effects of mutations and the physiopathological mechanisms in the field of cardiac arrhythmias.
Collapse
Affiliation(s)
- Yongfei Song
- Department of Cardiovascular, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo, China
- Yongfei Song
| | - Zequn Zheng
- Department of Cardiovascular, Medical College, Ningbo University, Ningbo, China
- Department of Cardiovascular, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Jiangfang Lian
- Department of Cardiovascular, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo, China
- Department of Cardiovascular, Medical College, Ningbo University, Ningbo, China
- Department of Cardiovascular, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- *Correspondence: Jiangfang Lian
| |
Collapse
|
13
|
Horváth B, Szentandrássy N, Dienes C, Kovács ZM, Nánási PP, Chen-Izu Y, Izu LT, Banyasz T. Exploring the Coordination of Cardiac Ion Channels With Action Potential Clamp Technique. Front Physiol 2022; 13:864002. [PMID: 35370800 PMCID: PMC8966222 DOI: 10.3389/fphys.2022.864002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 02/15/2022] [Indexed: 11/30/2022] Open
Abstract
The patch clamp technique underwent continual advancement and developed numerous variants in cardiac electrophysiology since its introduction in the late 1970s. In the beginning, the capability of the technique was limited to recording one single current from one cell stimulated with a rectangular command pulse. Since that time, the technique has been extended to record multiple currents under various command pulses including action potential. The current review summarizes the development of the patch clamp technique in cardiac electrophysiology with special focus on the potential applications in integrative physiology.
Collapse
Affiliation(s)
- Balázs Horváth
- Department of Physiology, University of Debrecen, Debrecen, Hungary
| | - Norbert Szentandrássy
- Department of Physiology, University of Debrecen, Debrecen, Hungary
- Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Csaba Dienes
- Department of Physiology, University of Debrecen, Debrecen, Hungary
| | | | - Péter P. Nánási
- Department of Physiology, University of Debrecen, Debrecen, Hungary
- Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Ye Chen-Izu
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Leighton T. Izu
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Tamas Banyasz
- Department of Physiology, University of Debrecen, Debrecen, Hungary
- *Correspondence: Tamas Banyasz,
| |
Collapse
|
14
|
Nijak A, Simons E, Vandendriessche B, Van de Sande D, Fransen E, Sieliwończyk E, Van Gucht I, Van Craenenbroeck E, Saenen J, Heidbuchel H, Ponsaerts P, Labro AJ, Snyders D, De Vos W, Schepers D, Alaerts M, Loeys BL. Morpho-functional comparison of differentiation protocols to create iPSC-derived cardiomyocytes. Biol Open 2022; 11:274508. [PMID: 35195246 PMCID: PMC8890088 DOI: 10.1242/bio.059016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/15/2021] [Indexed: 11/30/2022] Open
Abstract
Cardiomyocytes derived from induced pluripotent stem cells (iPSC-CMs) offer an attractive platform for cardiovascular research. Patient-specific iPSC-CMs are very useful for studying disease development, and bear potential for disease diagnostics, prognosis evaluation and development of personalized treatment. Several monolayer-based serum-free protocols have been described for the differentiation of iPSCs into cardiomyocytes, but data on their performance are scarce. In this study, we evaluated two protocols that are based on temporal modulation of the Wnt/β-catenin pathway for iPSC-CM differentiation from four iPSC lines, including two control individuals and two patients carrying an SCN5A mutation. The SCN5A gene encodes the cardiac voltage-gated sodium channel (Nav1.5) and loss-of-function mutations can cause the cardiac arrhythmia Brugada syndrome. We performed molecular characterization of the obtained iPSC-CMs by immunostaining for cardiac specific markers and by expression analysis of selected cardiac structural and ionic channel protein-encoding genes with qPCR. We also investigated cell growth morphology, contractility and survival of the iPSC-CMs after dissociation. Finally, we performed electrophysiological characterization of the cells, focusing on the action potential (AP) and calcium transient (CT) characteristics using patch-clamping and optical imaging, respectively. Based on our comprehensive morpho-functional analysis, we concluded that both tested protocols result in a high percentage of contracting CMs. Moreover, they showed acceptable survival and cell quality after dissociation (>50% of cells with a smooth cell membrane, possible to seal during patch-clamping). Both protocols generated cells presenting with typical iPSC-CM AP and CT characteristics, although one protocol (that involves sequential addition of CHIR99021 and Wnt-C59) rendered iPSC-CMs, which were more accessible for patch-clamp and calcium transient experiments and showed an expression pattern of cardiac-specific markers more similar to this observed in human heart left ventricle samples. Summary: In this study, we evaluated two protocols that are based on temporal modulation of the Wnt/β -catenin pathway for iPSC-CM differentiation from four iPSC lines. We show that both protocols were successful in the generation of contracting iPSC-CMs. However, one of the tested protocols rendered cells that were more accessible for patch-clamp experiments and showed an expression pattern of cardiac-specific markers more similar to this of human heart left ventricle samples.
Collapse
Affiliation(s)
- Aleksandra Nijak
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium
| | - Eline Simons
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium
| | - Bert Vandendriessche
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium
| | - Dieter Van de Sande
- Laboratory of Molecular Biophysics, Cellular and Network Excitability, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Erik Fransen
- StatUa Center of Statistics, University of Antwerp 2650, Antwerp, Belgium
| | - Ewa Sieliwończyk
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium
| | - Ilse Van Gucht
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium
| | - Emeline Van Craenenbroeck
- Department of Cardiology, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp 2650, Belgium
| | - Johan Saenen
- Department of Cardiology, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp 2650, Belgium
| | - Hein Heidbuchel
- Department of Cardiology, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp 2650, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Alain J Labro
- Laboratory of Molecular Biophysics, Cellular and Network Excitability, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium.,Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium
| | - Dirk Snyders
- Laboratory of Molecular Biophysics, Cellular and Network Excitability, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Winnok De Vos
- Laboratory of Cell Biology and Histology, Faculty of Veterinary Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Dorien Schepers
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium.,Laboratory of Molecular Biophysics, Cellular and Network Excitability, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Maaike Alaerts
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium
| | - Bart L Loeys
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp & Antwerp University Hospital, Antwerp 2650, Belgium.,Department of Human Genetics, Radboud University Medical Centre, Nijmegen 6525, The Netherlands
| |
Collapse
|
15
|
Koncz I, Verkerk AO, Nicastro M, Wilders R, Árpádffy-Lovas T, Magyar T, Tóth N, Nagy N, Madrid M, Lin Z, Efimov IR. Acetylcholine Reduces IKr and Prolongs Action Potentials in Human Ventricular Cardiomyocytes. Biomedicines 2022; 10:biomedicines10020244. [PMID: 35203454 PMCID: PMC8869322 DOI: 10.3390/biomedicines10020244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023] Open
Abstract
Vagal nerve stimulation (VNS) has a meaningful basis as a potentially effective treatment for heart failure with reduced ejection fraction. There is an ongoing VNS randomized study, and four studies are completed. However, relatively little is known about the effect of acetylcholine (ACh) on repolarization in human ventricular cardiomyocytes, as well as the effect of ACh on the rapid component of the delayed rectifier K+ current (IKr). Here, we investigated the effect of ACh on the action potential parameters in human ventricular preparations and on IKr in human induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs). Using standard microelectrode technique, we demonstrated that ACh (5 µM) significantly increased the action potential duration in human left ventricular myocardial slices. ACh (5 µM) also prolonged repolarization in a human Purkinje fiber and a papillary muscle. Optical mapping revealed that ACh increased the action potential duration in human left ventricular myocardial slices and that the effect was dose-dependent. Perforated patch clamp experiments demonstrated action potential prolongation and a significant decrease in IKr by ACh (5 µM) in hiPSC-CMs. Computer simulations of the electrical activity of a human ventricular cardiomyocyte showed an increase in action potential duration upon implementation of the experimentally observed ACh-induced changes in the fully activated conductance and steady-state activation of IKr. Our findings support the hypothesis that ACh can influence the repolarization in human ventricular cardiomyocytes by at least changes in IKr.
Collapse
Affiliation(s)
- István Koncz
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; (I.K.); (M.M.); (Z.L.)
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
| | - Arie O. Verkerk
- Heart Center, Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (A.O.V.); (M.N.)
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Michele Nicastro
- Heart Center, Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (A.O.V.); (M.N.)
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Tamás Árpádffy-Lovas
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
| | - Tibor Magyar
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
| | - Noémi Tóth
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
- ELKH-SZTE Research Group of Cardiovascular Pharmacology, 6721 Szeged, Hungary
| | - Micah Madrid
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; (I.K.); (M.M.); (Z.L.)
| | - Zexu Lin
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; (I.K.); (M.M.); (Z.L.)
| | - Igor R. Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; (I.K.); (M.M.); (Z.L.)
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Chicago, IL 60611, USA
- Correspondence: ; Tel.: +1-202-294-8182
| |
Collapse
|
16
|
Du C, Rasmusson RL, Bett GC, Franks B, Zhang H, Hancox JC. Investigation of the Effects of the Short QT Syndrome D172N Kir2.1 Mutation on Ventricular Action Potential Profile Using Dynamic Clamp. Front Pharmacol 2022; 12:794620. [PMID: 35115940 PMCID: PMC8806151 DOI: 10.3389/fphar.2021.794620] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/14/2021] [Indexed: 12/22/2022] Open
Abstract
The congenital short QT syndrome (SQTS) is a cardiac condition that leads to abbreviated ventricular repolarization and an increased susceptibility to arrhythmia and sudden death. The SQT3 form of the syndrome is due to mutations to the KCNJ2 gene that encodes Kir2.1, a critical component of channels underlying cardiac inwardly rectifying K+ current, IK1. The first reported SQT3 KCNJ2 mutation gives rise to the D172N Kir2.1 mutation, the consequences of which have been studied on recombinant channels in vitro and in ventricular cell and tissue simulations. The aim of this study was to establish the effects of the D172N mutation on ventricular repolarization through real-time replacement of IK1 using the dynamic clamp technique. Whole-cell patch-clamp recordings were made from adult guinea-pig left ventricular myocytes at physiological temperature. Action potentials (APs) were elicited at 1 Hz. Intrinsic IK1 was inhibited with a low concentration (50 µM) of Ba2+ ions, which led to AP prolongation and triangulation, accompanied by a ∼6 mV depolarization of resting membrane potential. Application of synthetic IK1 through dynamic clamp restored AP duration, shape and resting potential. Replacement of wild-type (WT) IK1 with heterozygotic (WT-D172N) or homozygotic (D172N) mutant formulations under dynamic clamp significantly abbreviated AP duration (APD90) and accelerated maximal AP repolarization velocity, with no significant hyperpolarization of resting potential. Across stimulation frequencies from 0.5 to 3 Hz, the relationship between APD90 and cycle length was downward shifted, reflecting AP abbreviation at all stimulation frequencies tested. In further AP measurements at 1 Hz from hiPSC cardiomyocytes, the D172N mutation produced similar effects on APD and repolarization velocity; however, resting potential was moderately hyperpolarized by application of mutant IK1 to these cells. Overall, the results of this study support the major changes in ventricular cell AP repolarization with the D172N predicted from prior AP modelling and highlight the potential utility of using adult ventricular cardiomyocytes for dynamic clamp exploration of functional consequences of Kir2.1 mutations.
Collapse
Affiliation(s)
- Chunyun Du
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Randall L. Rasmusson
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University of New York, University at Buffalo, Buffalo, NY, United States
- Cytocybernetics Inc, North Tonawanda, NY, United States
| | - Glenna C. Bett
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University of New York, University at Buffalo, Buffalo, NY, United States
- Cytocybernetics Inc, North Tonawanda, NY, United States
- Department of Obstetrics and Gynecology, Center for Cellular and Systems Electrophysiology, State University of New York, University at Buffalo, Buffalo, NY, United States
| | | | - Henggui Zhang
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
| | - Jules C. Hancox
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
- *Correspondence: Jules C. Hancox,
| |
Collapse
|
17
|
Approaches to Optimize Stem Cell-Derived Cardiomyocyte Maturation and Function. CURRENT STEM CELL REPORTS 2021. [DOI: 10.1007/s40778-021-00197-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
18
|
Marchal GA, Jouni M, Chiang DY, Pérez-Hernández M, Podliesna S, Yu N, Casini S, Potet F, Veerman CC, Klerk M, Lodder EM, Mengarelli I, Guan K, Vanoye CG, Rothenberg E, Charpentier F, Redon R, George AL, Verkerk AO, Bezzina CR, MacRae CA, Burridge PW, Delmar M, Galjart N, Portero V, Remme CA. Targeting the Microtubule EB1-CLASP2 Complex Modulates Na V1.5 at Intercalated Discs. Circ Res 2021; 129:349-365. [PMID: 34092082 PMCID: PMC8298292 DOI: 10.1161/circresaha.120.318643] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Gerard A Marchal
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Mariam Jouni
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - David Y Chiang
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA (D.Y.C., C.A.M.)
| | | | - Svitlana Podliesna
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Nuo Yu
- Department of Cell Biology, Erasmus Medical Centre Rotterdam, The Netherlands (N.Y., N.G.)
| | - Simona Casini
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Franck Potet
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Christiaan C Veerman
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Mischa Klerk
- Department of Medical Biology, Amsterdam UMC - location AMC, The Netherlands (M.K., A.O.V.)
| | - Elisabeth M Lodder
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Isabella Mengarelli
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Germany (K.G.)
| | - Carlos G Vanoye
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Eli Rothenberg
- Department of Biochemistry and Pharmacology (E.R.), NYU School of Medicine
| | - Flavien Charpentier
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Richard Redon
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Alfred L George
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Arie O Verkerk
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
- Department of Medical Biology, Amsterdam UMC - location AMC, The Netherlands (M.K., A.O.V.)
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Calum A MacRae
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA (D.Y.C., C.A.M.)
| | - Paul W Burridge
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Mario Delmar
- Division of Cardiology (M.P.-H., M.D.), NYU School of Medicine
| | - Niels Galjart
- Department of Cell Biology, Erasmus Medical Centre Rotterdam, The Netherlands (N.Y., N.G.)
| | - Vincent Portero
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| |
Collapse
|
19
|
Maturation strategies and limitations of induced pluripotent stem cell-derived cardiomyocytes. Biosci Rep 2021; 41:226678. [PMID: 33057659 PMCID: PMC8209171 DOI: 10.1042/bsr20200833] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) have the ability to differentiate into cardiomyocytes (CMs). They are not only widely used in cardiac pharmacology screening, human heart disease modeling, and cell transplantation-based treatments, but also the most promising source of CMs for experimental and clinical applications. However, their use is largely restricted by the immature phenotype of structure and function, which is similar to embryonic or fetal CMs and has certain differences from adult CMs. In order to overcome this critical issue, many studies have explored and revealed new strategies to induce the maturity of iPSC-CMs. Therefore, this article aims to review recent induction methods of mature iPSC-CMs, related mechanisms, and limitations.
Collapse
|
20
|
Gnecchi M, Sala L, Schwartz PJ. Precision Medicine and cardiac channelopathies: when dreams meet reality. Eur Heart J 2021; 42:1661-1675. [PMID: 33686390 PMCID: PMC8088342 DOI: 10.1093/eurheartj/ehab007] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/10/2020] [Accepted: 01/12/2021] [Indexed: 12/17/2022] Open
Abstract
Precision Medicine (PM) is an innovative approach that, by relying on large populations’ datasets, patients’ genetics and characteristics, and advanced technologies, aims at improving risk stratification and at identifying patient-specific management through targeted diagnostic and therapeutic strategies. Cardiac channelopathies are being progressively involved in the evolution brought by PM and some of them are benefiting from these novel approaches, especially the long QT syndrome. Here, we have explored the main layers that should be considered when developing a PM approach for cardiac channelopathies, with a focus on modern in vitro strategies based on patient-specific human-induced pluripotent stem cells and on in silico models. PM is where scientists and clinicians must meet and integrate their expertise to improve medical care in an innovative way but without losing common sense. We have indeed tried to provide the cardiologist’s point of view by comparing state-of-the-art techniques and approaches, including revolutionary discoveries, to current practice. This point matters because the new approaches may, or may not, exceed the efficacy and safety of established therapies. Thus, our own eagerness to implement the most recent translational strategies for cardiac channelopathies must be tempered by an objective assessment to verify whether the PM approaches are indeed making a difference for the patients. We believe that PM may shape the diagnosis and treatment of cardiac channelopathies for years to come. Nonetheless, its potential superiority over standard therapies should be constantly monitored and assessed before translating intellectually rewarding new discoveries into clinical practice.
Collapse
Affiliation(s)
- Massimiliano Gnecchi
- Department of Cardiothoracic and Vascular Sciences-Coronary Care Unit and Laboratory of Clinical and Experimental Cardiology, Fondazione IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy.,Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Viale Golgi 19, 27100 Pavia, Italy.,Department of Medicine, University of Cape Town, J-Floor, Old Main Building, Groote Schuur Hospital, Observatory, 7925 Cape Town, South Africa
| | - Luca Sala
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Via Pier Lombardo 22 - 20135 Milan, Italy
| | - Peter J Schwartz
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Via Pier Lombardo 22 - 20135 Milan, Italy
| |
Collapse
|
21
|
Feyen DAM, McKeithan WL, Bruyneel AAN, Spiering S, Hörmann L, Ulmer B, Zhang H, Briganti F, Schweizer M, Hegyi B, Liao Z, Pölönen RP, Ginsburg KS, Lam CK, Serrano R, Wahlquist C, Kreymerman A, Vu M, Amatya PL, Behrens CS, Ranjbarvaziri S, Maas RGC, Greenhaw M, Bernstein D, Wu JC, Bers DM, Eschenhagen T, Metallo CM, Mercola M. Metabolic Maturation Media Improve Physiological Function of Human iPSC-Derived Cardiomyocytes. Cell Rep 2021; 32:107925. [PMID: 32697997 PMCID: PMC7437654 DOI: 10.1016/j.celrep.2020.107925] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/15/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have enormous potential for the study of human cardiac disorders. However, their physiological immaturity severely limits their utility as a model system and their adoption for drug discovery. Here, we describe maturation media designed to provide oxidative substrates adapted to the metabolic needs of human iPSC (hiPSC)-CMs. Compared with conventionally cultured hiPSC-CMs, metabolically matured hiPSC-CMs contract with greater force and show an increased reliance on cardiac sodium (Na+) channels and sarcoplasmic reticulum calcium (Ca2+) cycling. The media enhance the function, long-term survival, and sarcomere structures in engineered heart tissues. Use of the maturation media made it possible to reliably model two genetic cardiac diseases: long QT syndrome type 3 due to a mutation in the cardiac Na+ channel SCN5A and dilated cardiomyopathy due to a mutation in the RNA splicing factor RBM20. The maturation media should increase the fidelity of hiPSC-CMs as disease models. Physiological immaturity of iPSC-derived cardiomyocytes limits their fidelity as disease models. Feyen et al. developed a low glucose, high oxidative substrate media that increase maturation of ventricular-like hiPSC-CMs in 2D and 3D cultures relative to standard protocols. Improved characteristics include a low resting Vm, rapid depolarization, and increased Ca2+ dependence and force generation.
Collapse
Affiliation(s)
- Dries A M Feyen
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wesley L McKeithan
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Arne A N Bruyneel
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sean Spiering
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Larissa Hörmann
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bärbel Ulmer
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hui Zhang
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Francesca Briganti
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michaela Schweizer
- Electron Microscopy Unit, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bence Hegyi
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Zhandi Liao
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | | | - Kenneth S Ginsburg
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Chi Keung Lam
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ricardo Serrano
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Christine Wahlquist
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Alexander Kreymerman
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michelle Vu
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Prashila L Amatya
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Charlotta S Behrens
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sara Ranjbarvaziri
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Renee G C Maas
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Matthew Greenhaw
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Daniel Bernstein
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Joseph C Wu
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Mark Mercola
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
22
|
Verkerk AO, Marchal GA, Zegers JG, Kawasaki M, Driessen AHG, Remme CA, de Groot JR, Wilders R. Patch-Clamp Recordings of Action Potentials From Human Atrial Myocytes: Optimization Through Dynamic Clamp. Front Pharmacol 2021; 12:649414. [PMID: 33912059 PMCID: PMC8072333 DOI: 10.3389/fphar.2021.649414] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/18/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction: Atrial fibrillation (AF) is the most common cardiac arrhythmia. Consequently, novel therapies are being developed. Ultimately, the impact of compounds on the action potential (AP) needs to be tested in freshly isolated human atrial myocytes. However, the frequent depolarized state of these cells upon isolation seriously hampers reliable AP recordings. Purpose: We assessed whether AP recordings from single human atrial myocytes could be improved by providing these cells with a proper inward rectifier K+ current (IK1), and consequently with a regular, non-depolarized resting membrane potential (RMP), through “dynamic clamp”. Methods: Single myocytes were enzymatically isolated from left atrial appendage tissue obtained from patients with paroxysmal AF undergoing minimally invasive surgical ablation. APs were elicited at 1 Hz and measured using perforated patch-clamp methodology, injecting a synthetic IK1 to generate a regular RMP. The injected IK1 had strong or moderate rectification. For comparison, a regular RMP was forced through injection of a constant outward current. A wide variety of ion channel blockers was tested to assess their modulatory effects on AP characteristics. Results: Without any current injection, RMPs ranged from −9.6 to −86.2 mV in 58 cells. In depolarized cells (RMP positive to −60 mV), RMP could be set at −80 mV using IK1 or constant current injection and APs could be evoked upon stimulation. AP duration differed significantly between current injection methods (p < 0.05) and was shortest with constant current injection and longest with injection of IK1 with strong rectification. With moderate rectification, AP duration at 90% repolarization (APD90) was similar to myocytes with regular non-depolarized RMP, suggesting that a synthetic IK1 with moderate rectification is the most appropriate for human atrial myocytes. Importantly, APs evoked using each injection method were still sensitive to all drugs tested (lidocaine, nifedipine, E-4031, low dose 4-aminopyridine, barium, and apamin), suggesting that the major ionic currents of the atrial cells remained functional. However, certain drug effects were quantitatively dependent on the current injection approach used. Conclusion: Injection of a synthetic IK1 with moderate rectification facilitates detailed AP measurements in human atrial myocytes. Therefore, dynamic clamp represents a promising tool for testing novel antiarrhythmic drugs.
Collapse
Affiliation(s)
- Arie O Verkerk
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Gerard A Marchal
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jan G Zegers
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Makiri Kawasaki
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Antoine H G Driessen
- Department of Cardiothoracic Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Joris R de Groot
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
23
|
Verkerk AO, Wilders R. Dynamic Clamp in Electrophysiological Studies on Stem Cell-Derived Cardiomyocytes-Why and How? J Cardiovasc Pharmacol 2021; 77:267-279. [PMID: 33229908 DOI: 10.1097/fjc.0000000000000955] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/31/2020] [Indexed: 12/15/2022]
Abstract
ABSTRACT Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are supposed to be a good human-based model, with virtually unlimited cell source, for studies on mechanisms underlying cardiac development and cardiac diseases, and for identification of drug targets. However, a major drawback of hPSC-CMs as a model system, especially for electrophysiological studies, is their depolarized state and associated spontaneous electrical activity. Various approaches are used to overcome this drawback, including the injection of "synthetic" inward rectifier potassium current (IK1), which is computed in real time, based on the recorded membrane potential ("dynamic clamp"). Such injection of an IK1-like current results in quiescent hPSC-CMs with a nondepolarized resting potential that show "adult-like" action potentials on stimulation, with functional availability of the most important ion channels involved in cardiac electrophysiology. These days, dynamic clamp has become a widely appreciated electrophysiological tool. However, setting up a dynamic clamp system can still be laborious and difficult, both because of the required hardware and the implementation of the dedicated software. In the present review, we first summarize the potential mechanisms underlying the depolarized state of hPSC-CMs and the functional consequences of this depolarized state. Next, we explain how an existing manual patch clamp setup can be extended with dynamic clamp. Finally, we shortly validate the extended setup with atrial-like and ventricular-like hPSC-CMs. We feel that dynamic clamp is a highly valuable tool in the field of cellular electrophysiological studies on hPSC-CMs and hope that our directions for setting up such dynamic clamp system may prove helpful.
Collapse
Affiliation(s)
- Arie O Verkerk
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands ; and
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands ; and
| |
Collapse
|
24
|
Verkerk AO, Knottnerus SJG, Portero V, Bleeker JC, Ferdinandusse S, Guan K, IJlst L, Visser G, Wanders RJA, Wijburg FA, Bezzina CR, Mengarelli I, Houtkooper RH. Electrophysiological Abnormalities in VLCAD Deficient hiPSC-Cardiomyocytes Do not Improve with Carnitine Supplementation. Front Pharmacol 2021; 11:616834. [PMID: 33597881 PMCID: PMC7883678 DOI: 10.3389/fphar.2020.616834] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Patients with a deficiency in very long-chain acyl-CoA dehydrogenase (VLCAD), an enzyme that is involved in the mitochondrial beta-oxidation of long-chain fatty acids, are at risk for developing cardiac arrhythmias. In human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs), VLCAD deficiency (VLCADD) results in a series of abnormalities, including: 1) accumulation of long-chain acylcarnitines, 2) action potential shortening, 3) higher systolic and diastolic intracellular Ca2+ concentrations, and 4) development of delayed afterdepolarizations. In the fatty acid oxidation process, carnitine is required for bidirectional transport of acyl groups across the mitochondrial membrane. Supplementation has been suggested as potential therapeutic approach in VLCADD, but its benefits are debated. Here, we studied the effects of carnitine supplementation on the long-chain acylcarnitine levels and performed electrophysiological analyses in VLCADD patient-derived hiPSC-CMs with a ACADVL gene mutation (p.Val283Ala/p.Glu381del). Under standard culture conditions, VLCADD hiPSC-CMs showed high concentrations of long-chain acylcarnitines, short action potentials, and high delayed afterdepolarizations occurrence. Incubation of the hiPSC-CMs with 400 µM L-carnitine for 48 h led to increased long-chain acylcarnitine levels both in medium and cells. In addition, carnitine supplementation neither restored abnormal action potential parameters nor the increased occurrence of delayed afterdepolarizations in VLCADD hiPSC-CMs. We conclude that long-chain acylcarnitine accumulation and electrophysiological abnormalities in VLCADD hiPSC-CMs are not normalized by carnitine supplementation, indicating that this treatment is unlikely to be beneficial against cardiac arrhythmias in VLCADD patients.
Collapse
Affiliation(s)
- Arie O Verkerk
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Suzan J G Knottnerus
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Vincent Portero
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jeannette C Bleeker
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Lodewijk IJlst
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Gepke Visser
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Frits A Wijburg
- Department of Pediatric Metabolic Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Isabella Mengarelli
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| |
Collapse
|
25
|
Microelectrode Arrays: A Valuable Tool to Analyze Stem Cell-Derived Cardiomyocytes. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
26
|
De Waard S, Montnach J, Ribeiro B, Nicolas S, Forest V, Charpentier F, Mangoni ME, Gaborit N, Ronjat M, Loussouarn G, Lemarchand P, De Waard M. Functional Impact of BeKm-1, a High-Affinity hERG Blocker, on Cardiomyocytes Derived from Human-Induced Pluripotent Stem Cells. Int J Mol Sci 2020; 21:ijms21197167. [PMID: 32998413 PMCID: PMC7582727 DOI: 10.3390/ijms21197167] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 12/22/2022] Open
Abstract
IKr current, a major component of cardiac repolarization, is mediated by human Ether-à-go-go-Related Gene (hERG, Kv11.1) potassium channels. The blockage of these channels by pharmacological compounds is associated to drug-induced long QT syndrome (LQTS), which is a life-threatening disorder characterized by ventricular arrhythmias and defects in cardiac repolarization that can be illustrated using cardiomyocytes derived from human-induced pluripotent stem cells (hiPS-CMs). This study was meant to assess the modification in hiPS-CMs excitability and contractile properties by BeKm-1, a natural scorpion venom peptide that selectively interacts with the extracellular face of hERG, by opposition to reference compounds that act onto the intracellular face. Using an automated patch-clamp system, we compared the affinity of BeKm-1 for hERG channels with some reference compounds. We fully assessed its effects on the electrophysiological, calcium handling, and beating properties of hiPS-CMs. By delaying cardiomyocyte repolarization, the peptide induces early afterdepolarizations and reduces spontaneous action potentials, calcium transients, and contraction frequencies, therefore recapitulating several of the critical phenotype features associated with arrhythmic risk in drug-induced LQTS. BeKm-1 exemplifies an interesting reference compound in the integrated hiPS-CMs cell model for all drugs that may block the hERG channel from the outer face. Being a peptide that is easily modifiable, it will serve as an ideal molecular platform for the design of new hERG modulators displaying additional functionalities.
Collapse
Affiliation(s)
- Stephan De Waard
- L’institut du thorax, INSERM, CNRS, Université de Nantes, F-44007 Nantes, France; (S.D.W.); (J.M.); (B.R.); (S.N.); (V.F.); (F.C.); (N.G.); (M.R.); (G.L.); (P.L.)
- LabEx Ion Channels, Science & Therapeutics, F-06560 Valbonne, France;
| | - Jérôme Montnach
- L’institut du thorax, INSERM, CNRS, Université de Nantes, F-44007 Nantes, France; (S.D.W.); (J.M.); (B.R.); (S.N.); (V.F.); (F.C.); (N.G.); (M.R.); (G.L.); (P.L.)
| | - Barbara Ribeiro
- L’institut du thorax, INSERM, CNRS, Université de Nantes, F-44007 Nantes, France; (S.D.W.); (J.M.); (B.R.); (S.N.); (V.F.); (F.C.); (N.G.); (M.R.); (G.L.); (P.L.)
| | - Sébastien Nicolas
- L’institut du thorax, INSERM, CNRS, Université de Nantes, F-44007 Nantes, France; (S.D.W.); (J.M.); (B.R.); (S.N.); (V.F.); (F.C.); (N.G.); (M.R.); (G.L.); (P.L.)
| | - Virginie Forest
- L’institut du thorax, INSERM, CNRS, Université de Nantes, F-44007 Nantes, France; (S.D.W.); (J.M.); (B.R.); (S.N.); (V.F.); (F.C.); (N.G.); (M.R.); (G.L.); (P.L.)
| | - Flavien Charpentier
- L’institut du thorax, INSERM, CNRS, Université de Nantes, F-44007 Nantes, France; (S.D.W.); (J.M.); (B.R.); (S.N.); (V.F.); (F.C.); (N.G.); (M.R.); (G.L.); (P.L.)
| | - Matteo Elia Mangoni
- LabEx Ion Channels, Science & Therapeutics, F-06560 Valbonne, France;
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, F34094 Montpellier, France
| | - Nathalie Gaborit
- L’institut du thorax, INSERM, CNRS, Université de Nantes, F-44007 Nantes, France; (S.D.W.); (J.M.); (B.R.); (S.N.); (V.F.); (F.C.); (N.G.); (M.R.); (G.L.); (P.L.)
| | - Michel Ronjat
- L’institut du thorax, INSERM, CNRS, Université de Nantes, F-44007 Nantes, France; (S.D.W.); (J.M.); (B.R.); (S.N.); (V.F.); (F.C.); (N.G.); (M.R.); (G.L.); (P.L.)
- LabEx Ion Channels, Science & Therapeutics, F-06560 Valbonne, France;
| | - Gildas Loussouarn
- L’institut du thorax, INSERM, CNRS, Université de Nantes, F-44007 Nantes, France; (S.D.W.); (J.M.); (B.R.); (S.N.); (V.F.); (F.C.); (N.G.); (M.R.); (G.L.); (P.L.)
| | - Patricia Lemarchand
- L’institut du thorax, INSERM, CNRS, Université de Nantes, F-44007 Nantes, France; (S.D.W.); (J.M.); (B.R.); (S.N.); (V.F.); (F.C.); (N.G.); (M.R.); (G.L.); (P.L.)
| | - Michel De Waard
- L’institut du thorax, INSERM, CNRS, Université de Nantes, F-44007 Nantes, France; (S.D.W.); (J.M.); (B.R.); (S.N.); (V.F.); (F.C.); (N.G.); (M.R.); (G.L.); (P.L.)
- LabEx Ion Channels, Science & Therapeutics, F-06560 Valbonne, France;
- Smartox Biotechnology, 6 rue des Platanes, F-38120 Saint-Egrève, France
- Correspondence: ; Tel.: +33-228-080-076
| |
Collapse
|
27
|
Li W, Han JL, Entcheva E. Syncytium cell growth increases Kir2.1 contribution in human iPSC-cardiomyocytes. Am J Physiol Heart Circ Physiol 2020; 319:H1112-H1122. [PMID: 32986966 PMCID: PMC7789971 DOI: 10.1152/ajpheart.00148.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) enable cardiotoxicity testing and personalized medicine. However, their maturity is of concern, including relatively depolarized resting membrane potential and more spontaneous activity compared with adult cardiomyocytes, implicating low or lacking inward rectifier potassium current (Ik1). Here, protein quantification confirms Kir2.1 expression in hiPSC-CM syncytia, albeit several times lower than in adult heart tissue. We find that hiPSC-CM culture density influences Kir2.1 expression at the mRNA level (potassium inwardly rectifying channel subfamily J member 2) and at the protein level and its associated electrophysiology phenotype. Namely, all-optical cardiac electrophysiology and pharmacological treatments reveal reduction of spontaneous and irregular activity and increase in action potential upstroke in denser cultures. Blocking Ik1-like currents with BaCl2 increased spontaneous frequency and blunted action potential upstrokes during pacing in a dose-dependent manner only in the highest-density cultures, in line with Ik1’s role in regulating the resting membrane potential. Our results emphasize the importance of syncytial growth of hiPSC-CMs for more physiologically relevant phenotype and the power of all-optical electrophysiology to study cardiomyocytes in their multicellular setting. NEW & NOTEWORTHY We identify cell culture density and cell-cell contact as an important factor in determining the expression of a key ion channel at the transcriptional and the protein levels, KCNJ2/Kir2.1, and its contribution to the electrophysiology of human induced pluripotent stem cell-derived cardiomyocytes. Our results indicate that studies on isolated cells, out of tissue context, may underestimate the cellular ion channel properties being characterized.
Collapse
Affiliation(s)
- Weizhen Li
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - Julie L Han
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| |
Collapse
|
28
|
Hilderink S, Devalla HD, Bosch L, Wilders R, Verkerk AO. Ultrarapid Delayed Rectifier K + Channelopathies in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Front Cell Dev Biol 2020; 8:536. [PMID: 32850774 PMCID: PMC7399090 DOI: 10.3389/fcell.2020.00536] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia. About 5-15% of AF patients have a mutation in a cardiac gene, including mutations in KCNA5, encoding the Kv1.5 α-subunit of the ion channel carrying the atrial-specific ultrarapid delayed rectifier K+ current (IKur). Both loss-of-function and gain-of-function AF-related mutations in KCNA5 are known, but their effects on action potentials (APs) of human cardiomyocytes have been poorly studied. Here, we assessed the effects of wild-type and mutant IKur on APs of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). We found that atrial-like hiPSC-CMs, generated by a retinoic acid-based differentiation protocol, have APs with faster repolarization compared to ventricular-like hiPSC-CMs, resulting in shorter APs with a lower AP plateau. Native IKur, measured as current sensitive to 50 μM 4-aminopyridine, was 1.88 ± 0.49 (mean ± SEM, n = 17) and 0.26 ± 0.26 pA/pF (n = 17) in atrial- and ventricular-like hiPSC-CMs, respectively. In both atrial- and ventricular-like hiPSC-CMs, IKur blockade had minimal effects on AP parameters. Next, we used dynamic clamp to inject various amounts of a virtual IKur, with characteristics as in freshly isolated human atrial myocytes, into 11 atrial-like and 10 ventricular-like hiPSC-CMs, in which native IKur was blocked. Injection of IKur with 100% density shortened the APs, with its effect being strongest on the AP duration at 20% repolarization (APD20) of atrial-like hiPSC-CMs. At IKur densities < 100% (compared to 100%), simulating loss-of-function mutations, significant AP prolongation and raise of plateau were observed. At IKur densities > 100%, simulating gain-of-function mutations, APD20 was decreased in both atrial- and ventricular-like hiPSC-CMs, but only upon a strong increase in IKur. In ventricular-like hiPSC-CMs, lowering of the plateau resulted in AP shortening. We conclude that a decrease in IKur, mimicking loss-of-function mutations, has a stronger effect on the AP of hiPSC-CMs than an increase, mimicking gain-of-function mutations, whereas in ventricular-like hiPSC-CMs such increase results in AP shortening, causing their AP morphology to become more atrial-like. Effects of native IKur modulation on atrial-like hiPSC-CMs are less pronounced than effects of virtual IKur injection because IKur density of atrial-like hiPSC-CMs is substantially smaller than that of freshly isolated human atrial myocytes.
Collapse
Affiliation(s)
- Sarah Hilderink
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Harsha D Devalla
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Leontien Bosch
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
29
|
Gaur N, Ortega F, Verkerk AO, Mengarelli I, Krogh-Madsen T, Christini DJ, Coronel R, Vigmond EJ. Validation of quantitative measure of repolarization reserve as a novel marker of drug induced proarrhythmia. J Mol Cell Cardiol 2020; 145:122-132. [PMID: 32325153 DOI: 10.1016/j.yjmcc.2020.04.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/01/2020] [Accepted: 04/14/2020] [Indexed: 11/25/2022]
Abstract
Repolarization reserve, the robustness of a cell to repolarize even when one of the repolarization mechanisms is failing, has been described qualitatively in terms of ionic currents, but has not been quantified by a generic metric that is applicable to drug screening. Prolonged repolarization leading to repolarization failure is highly arrhythmogenic. It may lead to ventricular tachycardia caused by triggered activity from early afterdepolarizations (EADs), or it may promote the occurrence of unidirectional conduction block and reentry. Both types of arrhythmia may deteriorate into ventricular fibrillation (VF) and death. We define the Repolarization Reserve Current (RRC) as the minimum constant current necessary to prevent normal repolarization of a cell. After developing and testing RRC for nine computational ionic models of various species, we applied it experimentally to atrial and ventricular human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM), and isolated guinea-pig ventricular cardiomyocytes. In simulations, repolarization was all-or-none with a precise, model-dependent critical RRC, resulting in a discrete shift in the Action Potential Duration (APD) - RRC relation, in the occurrence of EADs and repolarization failure. These data were faithfully reproduced in cellular experiments. RRC allows simple, fast, unambiguous quantification of the arrhythmogenic propensity in cardiac cells of various origins and species without the need of prior knowledge of underlying currents and is suitable for high throughput applications, and personalized medicine applications.
Collapse
Affiliation(s)
- Namit Gaur
- IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, F-33600 Pessac-Bordeaux, France; Univ. Bordeaux, IMB, UMR 5251, F-33400 Talence, France
| | | | - Arie O Verkerk
- Dept. of Medical Biology, Academic Medical Center, Amsterdam, the Netherlands; Dept. of Experimental Cardiology, Academic Medical Center, Amsterdam, the Netherlands
| | - Isabella Mengarelli
- Dept. of Experimental Cardiology, Academic Medical Center, Amsterdam, the Netherlands
| | | | | | - Ruben Coronel
- IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, F-33600 Pessac-Bordeaux, France; Dept. of Experimental Cardiology, Academic Medical Center, Amsterdam, the Netherlands
| | - Edward J Vigmond
- IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, F-33600 Pessac-Bordeaux, France; Univ. Bordeaux, IMB, UMR 5251, F-33400 Talence, France.
| |
Collapse
|
30
|
Knottnerus SJG, Mengarelli I, Wüst RCI, Baartscheer A, Bleeker JC, Coronel R, Ferdinandusse S, Guan K, IJlst L, Li W, Luo X, Portero VM, Ulbricht Y, Visser G, Wanders RJA, Wijburg FA, Verkerk AO, Houtkooper RH, Bezzina CR. Electrophysiological Abnormalities in VLCAD Deficient hiPSC-Cardiomyocytes Can Be Improved by Lowering Accumulation of Fatty Acid Oxidation Intermediates. Int J Mol Sci 2020; 21:ijms21072589. [PMID: 32276429 PMCID: PMC7177397 DOI: 10.3390/ijms21072589] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/03/2020] [Accepted: 04/05/2020] [Indexed: 12/14/2022] Open
Abstract
Patients with very long-chain acyl-CoA dehydrogenase deficiency (VLCADD) can present with life-threatening cardiac arrhythmias. The pathophysiological mechanism is unknown. We reprogrammed fibroblasts from one mildly and one severely affected VLCADD patient, into human induced pluripotent stem cells (hiPSCs) and differentiated these into cardiomyocytes (VLCADD-CMs). VLCADD-CMs displayed shorter action potentials (APs), more delayed afterdepolarizations (DADs) and higher systolic and diastolic intracellular Ca2+ concentration ([Ca2+]i) than control CMs. The mitochondrial booster resveratrol mitigated the biochemical, electrophysiological and [Ca2+]i changes in the mild but not in the severe VLCADD-CMs. Accumulation of potentially toxic intermediates of fatty acid oxidation was blocked by substrate reduction with etomoxir. Incubation with etomoxir led to marked prolongation of AP duration and reduced DADs and [Ca2+]i in both VLCADD-CMs. These results provide compelling evidence that reduced accumulation of fatty acid oxidation intermediates, either by enhanced fatty acid oxidation flux through increased mitochondria biogenesis (resveratrol) or by inhibition of fatty acid transport into the mitochondria (etomoxir), rescues pro-arrhythmia defects in VLCADD-CMs and open doors for new treatments.
Collapse
Affiliation(s)
- Suzan J. G. Knottnerus
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands; (S.J.G.K.); (R.C.I.W.); (J.C.B.); (S.F.); (L.I.); (G.V.); (R.J.A.W.)
- Department of Paediatric Metabolic Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht, 3584 EA Utrecht, The Netherlands
| | - Isabella Mengarelli
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.M.); (A.B.); (R.C.); (V.M.P.); (A.O.V.)
| | - Rob C. I. Wüst
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands; (S.J.G.K.); (R.C.I.W.); (J.C.B.); (S.F.); (L.I.); (G.V.); (R.J.A.W.)
| | - Antonius Baartscheer
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.M.); (A.B.); (R.C.); (V.M.P.); (A.O.V.)
| | - Jeannette C. Bleeker
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands; (S.J.G.K.); (R.C.I.W.); (J.C.B.); (S.F.); (L.I.); (G.V.); (R.J.A.W.)
- Department of Paediatric Metabolic Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht, 3584 EA Utrecht, The Netherlands
| | - Ruben Coronel
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.M.); (A.B.); (R.C.); (V.M.P.); (A.O.V.)
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands; (S.J.G.K.); (R.C.I.W.); (J.C.B.); (S.F.); (L.I.); (G.V.); (R.J.A.W.)
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, 01069 Dresden, Germany; (K.G.); (W.L.); (X.L.); (Y.U.)
| | - Lodewijk IJlst
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands; (S.J.G.K.); (R.C.I.W.); (J.C.B.); (S.F.); (L.I.); (G.V.); (R.J.A.W.)
| | - Wener Li
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, 01069 Dresden, Germany; (K.G.); (W.L.); (X.L.); (Y.U.)
| | - Xiaojing Luo
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, 01069 Dresden, Germany; (K.G.); (W.L.); (X.L.); (Y.U.)
| | - Vincent M. Portero
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.M.); (A.B.); (R.C.); (V.M.P.); (A.O.V.)
| | - Ying Ulbricht
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, 01069 Dresden, Germany; (K.G.); (W.L.); (X.L.); (Y.U.)
| | - Gepke Visser
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands; (S.J.G.K.); (R.C.I.W.); (J.C.B.); (S.F.); (L.I.); (G.V.); (R.J.A.W.)
- Department of Paediatric Metabolic Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht, 3584 EA Utrecht, The Netherlands
| | - Ronald J. A. Wanders
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands; (S.J.G.K.); (R.C.I.W.); (J.C.B.); (S.F.); (L.I.); (G.V.); (R.J.A.W.)
| | - Frits A. Wijburg
- Department of Paediatric Metabolic Diseases, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Arie O. Verkerk
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.M.); (A.B.); (R.C.); (V.M.P.); (A.O.V.)
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, 1105 AZ Amsterdam, The Netherlands; (S.J.G.K.); (R.C.I.W.); (J.C.B.); (S.F.); (L.I.); (G.V.); (R.J.A.W.)
- Correspondence: (R.H.H.); (C.R.B.)
| | - Connie R. Bezzina
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.M.); (A.B.); (R.C.); (V.M.P.); (A.O.V.)
- Correspondence: (R.H.H.); (C.R.B.)
| |
Collapse
|
31
|
Lee YK, Sala L, Mura M, Rocchetti M, Pedrazzini M, Ran X, Mak TSH, Crotti L, Sham PC, Torre E, Zaza A, Schwartz PJ, Tse HF, Gnecchi M. MTMR4 SNVs modulate ion channel degradation and clinical severity in congenital long QT syndrome: insights in the mechanism of action of protective modifier genes. Cardiovasc Res 2020; 117:767-779. [PMID: 32173736 PMCID: PMC7898949 DOI: 10.1093/cvr/cvaa019] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/23/2019] [Accepted: 01/22/2020] [Indexed: 01/26/2023] Open
Abstract
Aims In long QT syndrome (LQTS) patients, modifier genes modulate the arrhythmic risk associated with a disease-causing mutation. Their recognition can improve risk stratification and clinical management, but their discovery represents a challenge. We tested whether a cellular-driven approach could help to identify new modifier genes and especially their mechanism of action. Methods and results We generated human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) from two patients carrying the same KCNQ1-Y111C mutation, but presenting opposite clinical phenotypes. We showed that the phenotype of the iPSC-CMs derived from the symptomatic patient is due to impaired trafficking and increased degradation of the mutant KCNQ1 and wild-type human ether-a-go-go-related gene. In the iPSC-CMs of the asymptomatic (AS) patient, the activity of an E3 ubiquitin-protein ligase (Nedd4L) involved in channel protein degradation was reduced and resulted in a decreased arrhythmogenic substrate. Two single-nucleotide variants (SNVs) on the Myotubularin-related protein 4 (MTMR4) gene, an interactor of Nedd4L, were identified by whole-exome sequencing as potential contributors to decreased Nedd4L activity. Correction of these SNVs by CRISPR/Cas9 unmasked the LQTS phenotype in AS cells. Importantly, the same MTMR4 variants were present in 77% of AS Y111C mutation carriers of a separate cohort. Thus, genetically mediated interference with Nedd4L activation seems associated with protective effects. Conclusion Our finding represents the first demonstration of the cellular mechanism of action of a protective modifier gene in LQTS. It provides new clues for advanced risk stratification and paves the way for the design of new therapies targeting this specific molecular pathway.
Collapse
Affiliation(s)
- Yee-Ki Lee
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Luca Sala
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milano, Italy.,Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
| | - Manuela Mura
- Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Department of Cardiothoracic and Vascular Sciences, Coronary Care Unit and Laboratory of Clinical and Experimental Cardiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marcella Rocchetti
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Matteo Pedrazzini
- Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
| | - Xinru Ran
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong, Hong Kong SAR, China.,Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | - Timothy S H Mak
- Department of Psychiatry, The University of Hong Kong, Hong Kong SAR, China
| | - Lia Crotti
- Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy.,Department of Cardiovascular, Neural and Metabolic Sciences, Istituto Auxologico Italiano, IRCCS, San Luca Hospital, Milan, Italy.,Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Pak C Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong SAR, China.,Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.,State Key Laboratory for Cognitive and Brain Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Eleonora Torre
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Antonio Zaza
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Peter J Schwartz
- Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong, Hong Kong SAR, China.,Guangzhou Institutes of Biomedicine and Health, Guangzhou, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Hong Kong SAR, China
| | - Massimiliano Gnecchi
- Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Department of Cardiothoracic and Vascular Sciences, Coronary Care Unit and Laboratory of Clinical and Experimental Cardiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Pavia, Italy.,Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
32
|
Ashwood C, Waas M, Weerasekera R, Gundry RL. Reference glycan structure libraries of primary human cardiomyocytes and pluripotent stem cell-derived cardiomyocytes reveal cell-type and culture stage-specific glycan phenotypes. J Mol Cell Cardiol 2020; 139:33-46. [PMID: 31972267 DOI: 10.1016/j.yjmcc.2019.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/23/2019] [Accepted: 12/27/2019] [Indexed: 12/16/2022]
Abstract
Cell surface glycoproteins play critical roles in maintaining cardiac structure and function in health and disease and the glycan-moiety attached to the protein is critical for proper protein folding, stability and signaling [1]. However, despite mounting evidence that glycan structures are key modulators of heart function and must be considered when developing cardiac biomarkers, we currently do not have a comprehensive view of the glycans present in the normal human heart. In the current study, we used porous graphitized carbon liquid chromatography interfaced with mass spectrometry (PGC-LC-MS) to generate glycan structure libraries for primary human heart tissue homogenate, cardiomyocytes (CM) enriched from human heart tissue, and human induced pluripotent stem cell derived CM (hiPSC-CM). Altogether, we established the first reference structure libraries of the cardiac glycome containing 265 N- and O-glycans. Comparing the N-glycome of CM enriched from primary heart tissue to that of heart tissue homogenate, the same pool of N-glycan structures was detected in each sample type but the relative signal of 21 structures significantly differed between samples, with the high mannose class increased in enriched CM. Moreover, by comparing primary CM to hiPSC-CM collected during 20-100 days of differentiation, dynamic changes in the glycan profile throughout in vitro differentiation were observed and differences between primary and hiPSC-CM were revealed. Namely, >30% of the N-glycome significantly changed across these time-points of differentiation and only 23% of the N-glycan structures were shared between hiPSC-CM and primary CM. These observations are an important complement to current genomic, transcriptomic, and proteomic profiling and reveal new considerations for the use and interpretation of hiPSC-CM models for studies of human development, disease, and drug testing. Finally, these data are expected to support future regenerative medicine efforts by informing targets for evaluating the immunogenic potential of hiPSC-CM and harnessing differences between immature, proliferative hiPSC-CM and adult primary CM.
Collapse
Affiliation(s)
- Christopher Ashwood
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew Waas
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ranjuna Weerasekera
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Rebekah L Gundry
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Center for Biomedical Mass Spectrometry Research, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
33
|
Lodrini AM, Barile L, Rocchetti M, Altomare C. Human Induced Pluripotent Stem Cells Derived from a Cardiac Somatic Source: Insights for an In-Vitro Cardiomyocyte Platform. Int J Mol Sci 2020; 21:ijms21020507. [PMID: 31941149 PMCID: PMC7013592 DOI: 10.3390/ijms21020507] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/10/2020] [Accepted: 01/10/2020] [Indexed: 12/24/2022] Open
Abstract
Reprogramming of adult somatic cells into induced pluripotent stem cells (iPSCs) has revolutionized the complex scientific field of disease modelling and personalized therapy. Cardiac differentiation of human iPSCs into cardiomyocytes (hiPSC-CMs) has been used in a wide range of healthy and disease models by deriving CMs from different somatic cells. Unfortunately, hiPSC-CMs have to be improved because existing protocols are not completely able to obtain mature CMs recapitulating physiological properties of human adult cardiac cells. Therefore, improvements and advances able to standardize differentiation conditions are needed. Lately, evidences of an epigenetic memory retained by the somatic cells used for deriving hiPSC-CMs has led to evaluation of different somatic sources in order to obtain more mature hiPSC-derived CMs.
Collapse
Affiliation(s)
- Alessandra Maria Lodrini
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milano 20126, Italy; (A.M.L.); (M.R.)
| | - Lucio Barile
- Fondazione Cardiocentro Ticino, Lugano 6900, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6900, Switzerland
| | - Marcella Rocchetti
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milano 20126, Italy; (A.M.L.); (M.R.)
| | - Claudia Altomare
- Fondazione Cardiocentro Ticino, Lugano 6900, Switzerland;
- Correspondence:
| |
Collapse
|
34
|
van den Brink L, Brandão KO, Yiangou L, Mol MPH, Grandela C, Mummery CL, Verkerk AO, Davis RP. Cryopreservation of human pluripotent stem cell-derived cardiomyocytes is not detrimental to their molecular and functional properties. Stem Cell Res 2020; 43:101698. [PMID: 31945612 PMCID: PMC7611364 DOI: 10.1016/j.scr.2019.101698] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 12/06/2019] [Accepted: 12/30/2019] [Indexed: 12/15/2022] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have emerged as a powerful platform for in vitro modelling of cardiac diseases, safety pharmacology and drug screening. All these applications require large quantities of well-characterised and standardised batches of hiPSC-CMs. Cryopreservation of hiPSC-CMs without affecting their biochemical or biophysical phenotype is essential for facilitating this, but ideally requires the cells being unchanged by the freeze-thaw procedure. We therefore compared the in vitro functional and molecular characteristics of fresh and cryopreserved hiPSC-CMs generated from multiple independent hiPSC lines. While the frozen hiPSC-CMs exhibited poorer replating than their freshly-derived counterparts, there was no difference in the proportion of cardiomyocytes retrieved from the mixed population when this was factored in, although for several lines a higher percentage of ventricular-like hiPSC-CMs were recovered following cryopreservation. Furthermore, cryopreserved hiPSC-CMs from one line exhibited longer action potential durations. These results provide evidence that cryopreservation does not compromise the in vitro molecular, physiological and mechanical properties of hiPSC-CMs, though can lead to an enrichment in ventricular myocytes. It also validates this procedure for storing hiPSC-CMs, thereby allowing the same batch of hiPSC-CMs to be used for multiple applications and evaluations.
Collapse
Affiliation(s)
- Lettine van den Brink
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands
| | - Karina O Brandão
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands
| | - Mervyn P H Mol
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands
| | - Catarina Grandela
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam UMC, 1105 AZ Amsterdam, the Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands.
| |
Collapse
|
35
|
Becker N, Horváth A, De Boer T, Fabbri A, Grad C, Fertig N, George M, Obergrussberger A. Automated Dynamic Clamp for Simulation of I
K1
in Human Induced Pluripotent Stem Cell–Derived Cardiomyocytes in Real Time Using Patchliner Dynamite
8. ACTA ACUST UNITED AC 2019; 88:e70. [DOI: 10.1002/cpph.70] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | | | - Teun De Boer
- Department of Medical PhysiologyUniversity Medical Center Utrecht Utrecht The Netherlands
| | - Alan Fabbri
- Department of Medical PhysiologyUniversity Medical Center Utrecht Utrecht The Netherlands
| | | | | | | | | |
Collapse
|
36
|
Establishment of an automated patch-clamp platform for electrophysiological and pharmacological evaluation of hiPSC-CMs. Stem Cell Res 2019; 41:101662. [DOI: 10.1016/j.scr.2019.101662] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 11/05/2019] [Accepted: 11/17/2019] [Indexed: 02/04/2023] Open
|
37
|
Casini S, Marchal GA, Kawasaki M, Nariswari FA, Portero V, van den Berg NWE, Guan K, Driessen AHG, Veldkamp MW, Mengarelli I, de Groot JR, Verkerk AO, Remme CA. Absence of Functional Na v1.8 Channels in Non-diseased Atrial and Ventricular Cardiomyocytes. Cardiovasc Drugs Ther 2019; 33:649-660. [PMID: 31916131 PMCID: PMC6994555 DOI: 10.1007/s10557-019-06925-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Several studies have indicated a potential role for SCN10A/NaV1.8 in modulating cardiac electrophysiology and arrhythmia susceptibility. However, by which mechanism SCN10A/NaV1.8 impacts on cardiac electrical function is still a matter of debate. To address this, we here investigated the functional relevance of NaV1.8 in atrial and ventricular cardiomyocytes (CMs), focusing on the contribution of NaV1.8 to the peak and late sodium current (INa) under normal conditions in different species. METHODS The effects of the NaV1.8 blocker A-803467 were investigated through patch-clamp analysis in freshly isolated rabbit left ventricular CMs, human left atrial CMs and human-induced pluripotent stem cell-derived CMs (hiPSC-CMs). RESULTS A-803467 treatment caused a slight shortening of the action potential duration (APD) in rabbit CMs and hiPSC-CMs, while it had no effect on APD in human atrial cells. Resting membrane potential, action potential (AP) amplitude, and AP upstroke velocity were unaffected by A-803467 application. Similarly, INa density was unchanged after exposure to A-803467 and NaV1.8-based late INa was undetectable in all cell types analysed. Finally, low to absent expression levels of SCN10A were observed in human atrial tissue, rabbit ventricular tissue and hiPSC-CMs. CONCLUSION We here demonstrate the absence of functional NaV1.8 channels in non-diseased atrial and ventricular CMs. Hence, the association of SCN10A variants with cardiac electrophysiology observed in, e.g. genome wide association studies, is likely the result of indirect effects on SCN5A expression and/or NaV1.8 activity in cell types other than CMs.
Collapse
Affiliation(s)
- Simona Casini
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands.
| | - Gerard A Marchal
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Makiri Kawasaki
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Fransisca A Nariswari
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Vincent Portero
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | | | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Antoine H G Driessen
- Department of Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Marieke W Veldkamp
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Isabella Mengarelli
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Joris R de Groot
- Department of Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Arie O Verkerk
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
- Department of Medical Biology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC, Meibergdreef 15, Amsterdam, 1105 AZ, The Netherlands
| |
Collapse
|
38
|
Garg P, Garg V, Shrestha R, Sanguinetti MC, Kamp TJ, Wu JC. Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes as Models for Cardiac Channelopathies: A Primer for Non-Electrophysiologists. Circ Res 2019; 123:224-243. [PMID: 29976690 DOI: 10.1161/circresaha.118.311209] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Life threatening ventricular arrhythmias leading to sudden cardiac death are a major cause of morbidity and mortality. In the absence of structural heart disease, these arrhythmias, especially in the younger population, are often an outcome of genetic defects in specialized membrane proteins called ion channels. In the heart, exceptionally well-orchestrated activity of a diversity of ion channels mediates the cardiac action potential. Alterations in either the function or expression of these channels can disrupt the configuration of the action potential, leading to abnormal electrical activity of the heart that can sometimes initiate an arrhythmia. Understanding the pathophysiology of inherited arrhythmias can be challenging because of the complexity of the disorder and lack of appropriate cellular and in vivo models. Recent advances in human induced pluripotent stem cell technology have provided remarkable progress in comprehending the underlying mechanisms of ion channel disorders or channelopathies by modeling these complex arrhythmia syndromes in vitro in a dish. To fully realize the potential of induced pluripotent stem cells in elucidating the mechanistic basis and complex pathophysiology of channelopathies, it is crucial to have a basic knowledge of cardiac myocyte electrophysiology. In this review, we will discuss the role of the various ion channels in cardiac electrophysiology and the molecular and cellular mechanisms of arrhythmias, highlighting the promise of human induced pluripotent stem cell-cardiomyocytes as a model for investigating inherited arrhythmia syndromes and testing antiarrhythmic strategies. Overall, this review aims to provide a basic understanding of the electrical activity of the heart and related channelopathies, especially to clinicians or research scientists in the cardiovascular field with limited electrophysiology background.
Collapse
Affiliation(s)
- Priyanka Garg
- From the Stanford Cardiovascular Institute (P.G., R.S., J.C.W.).,Department of Medicine, Division of Cardiology (P.G., R.S., J.C.W.).,Institute for Stem Cell Biology and Regenerative Medicine (P.G., R.S., J.C.W.)
| | - Vivek Garg
- Stanford University School of Medicine, CA; Department of Physiology, University of California San Francisco (V.G.)
| | - Rajani Shrestha
- From the Stanford Cardiovascular Institute (P.G., R.S., J.C.W.).,Department of Medicine, Division of Cardiology (P.G., R.S., J.C.W.).,Institute for Stem Cell Biology and Regenerative Medicine (P.G., R.S., J.C.W.)
| | | | - Timothy J Kamp
- Department of Medicine, University of Wisconsin-Madison (T.J.K.)
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute (P.G., R.S., J.C.W.) .,Department of Medicine, Division of Cardiology (P.G., R.S., J.C.W.).,Institute for Stem Cell Biology and Regenerative Medicine (P.G., R.S., J.C.W.)
| |
Collapse
|
39
|
Fabbri A, Goversen B, Vos MA, van Veen TAB, de Boer TP. Required G K1 to Suppress Automaticity of iPSC-CMs Depends Strongly on I K1 Model Structure. Biophys J 2019; 117:2303-2315. [PMID: 31623886 PMCID: PMC6990378 DOI: 10.1016/j.bpj.2019.08.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/24/2019] [Accepted: 08/26/2019] [Indexed: 01/06/2023] Open
Abstract
Human-induced pluripotent stem cells derived cardiomyocytes (hiPSC-CMs) are a virtually endless source of human cardiomyocytes that may become a great tool for safety pharmacology; however, their electrical phenotype is immature: they show spontaneous action potentials (APs) and an unstable and depolarized resting membrane potential (RMP) because of lack of IK1. Such immaturity hampers their application in assessing drug safety. The electronic overexpression of IK1 (e.g., through the dynamic clamp (DC) technique) is an option to overcome this deficit. In this computational study, we aim to estimate how much IK1 is needed to bring hiPSC-CMs to a stable and hyperpolarized RMP and which mathematical description of IK1 is most suitable for DC experiments. We compared five mature IK1 formulations (Bett, Dhamoon, Ishihara, O’Hara-Rudy, and ten Tusscher) with the native one (Paci), evaluating the main properties (outward peak, degree of rectification), and we quantified their effects on AP features (RMP, V˙max, APD50, APD90 (AP duration at 50 and 90% of repolarization), and APD50/APD90) after including them in the hiPSC-CM mathematical model by Paci. Then, we automatically identified the critical conductance for IK1 ( GK1, critical), the minimally required amount of IK1 suppressing spontaneous activity. Preconditioning the cell model with depolarizing/hyperpolarizing prepulses allowed us to highlight time dependency of the IK1 formulations. Simulations showed that inclusion of mature IK1 formulations resulted in hyperpolarized RMP and higher V˙max, and observed GK1, critical and the effect on AP duration strongly depended on IK1 formulation. Finally, the Ishihara IK1 led to shorter (−16.3%) and prolonged (+6.5%) APD90 in response to hyperpolarizing and depolarizing prepulses, respectively, whereas other models showed negligible effects. Fine-tuning of GK1 is an important step in DC experiments. Our computational work proposes a procedure to automatically identify how much IK1 current is required to inject to stop the spontaneous activity and suggests the use of the Ishihara IK1 model to perform DC experiments in hiPSC-CMs.
Collapse
Affiliation(s)
- Alan Fabbri
- University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Marc A Vos
- University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Teun P de Boer
- University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
40
|
Phenotype-Based High-Throughput Classification of Long QT Syndrome Subtypes Using Human Induced Pluripotent Stem Cells. Stem Cell Reports 2019; 13:394-404. [PMID: 31378668 PMCID: PMC6700479 DOI: 10.1016/j.stemcr.2019.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 06/29/2019] [Accepted: 06/30/2019] [Indexed: 11/23/2022] Open
Abstract
For long QT syndrome (LQTS), recent progress in genome-sequencing technologies enabled the identification of rare genomic variants with diagnostic, prognostic, and therapeutic implications. However, pathogenic stratification of the identified variants remains challenging, especially in variants of uncertain significance. This study aimed to propose a phenotypic cell-based diagnostic assay for identifying LQTS to recognize pathogenic variants in a high-throughput manner suitable for screening. We investigated the response of LQT2-induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iPSC-CMs) following IKr blockade using a multi-electrode array, finding that the response to IKr blockade was significantly smaller than in Control-iPSC-CMs. Furthermore, we found that LQT1-iPSC-CMs and LQT3-iPSC-CMs could be distinguished from Control-iPSC-CMs by IKs blockade and INa blockade, respectively. This strategy might be helpful in compensating for the shortcomings of genetic testing of LQTS patients.
Collapse
|
41
|
Kernik DC, Morotti S, Wu H, Garg P, Duff HJ, Kurokawa J, Jalife J, Wu JC, Grandi E, Clancy CE. A computational model of induced pluripotent stem-cell derived cardiomyocytes incorporating experimental variability from multiple data sources. J Physiol 2019; 597:4533-4564. [PMID: 31278749 PMCID: PMC6767694 DOI: 10.1113/jp277724] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/05/2019] [Indexed: 12/22/2022] Open
Abstract
Key points Induced pluripotent stem cell‐derived cardiomyocytes (iPSC‐CMs) capture patient‐specific genotype–phenotype relationships, as well as cell‐to‐cell variability of cardiac electrical activity Computational modelling and simulation provide a high throughput approach to reconcile multiple datasets describing physiological variability, and also identify vulnerable parameter regimes We have developed a whole‐cell model of iPSC‐CMs, composed of single exponential voltage‐dependent gating variable rate constants, parameterized to fit experimental iPSC‐CM outputs We have utilized experimental data across multiple laboratories to model experimental variability and investigate subcellular phenotypic mechanisms in iPSC‐CMs This framework links molecular mechanisms to cellular‐level outputs by revealing unique subsets of model parameters linked to known iPSC‐CM phenotypes
Abstract There is a profound need to develop a strategy for predicting patient‐to‐patient vulnerability in the emergence of cardiac arrhythmia. A promising in vitro method to address patient‐specific proclivity to cardiac disease utilizes induced pluripotent stem cell‐derived cardiomyocytes (iPSC‐CMs). A major strength of this approach is that iPSC‐CMs contain donor genetic information and therefore capture patient‐specific genotype–phenotype relationships. A cited detriment of iPSC‐CMs is the cell‐to‐cell variability observed in electrical activity. We postulated, however, that cell‐to‐cell variability may constitute a strength when appropriately utilized in a computational framework to build cell populations that can be employed to identify phenotypic mechanisms and pinpoint key sensitive parameters. Thus, we have exploited variation in experimental data across multiple laboratories to develop a computational framework for investigating subcellular phenotypic mechanisms. We have developed a whole‐cell model of iPSC‐CMs composed of simple model components comprising ion channel models with single exponential voltage‐dependent gating variable rate constants, parameterized to fit experimental iPSC‐CM data for all major ionic currents. By optimizing ionic current model parameters to multiple experimental datasets, we incorporate experimentally‐observed variability in the ionic currents. The resulting population of cellular models predicts robust inter‐subject variability in iPSC‐CMs. This approach links molecular mechanisms to known cellular‐level iPSC‐CM phenotypes, as shown by comparing immature and mature subpopulations of models to analyse the contributing factors underlying each phenotype. In the future, the presented models can be readily expanded to include genetic mutations and pharmacological interventions for studying the mechanisms of rare events, such as arrhythmia triggers. Induced pluripotent stem cell‐derived cardiomyocytes (iPSC‐CMs) capture patient‐specific genotype–phenotype relationships, as well as cell‐to‐cell variability of cardiac electrical activity Computational modelling and simulation provide a high throughput approach to reconcile multiple datasets describing physiological variability, and also identify vulnerable parameter regimes We have developed a whole‐cell model of iPSC‐CMs, composed of single exponential voltage‐dependent gating variable rate constants, parameterized to fit experimental iPSC‐CM outputs We have utilized experimental data across multiple laboratories to model experimental variability and investigate subcellular phenotypic mechanisms in iPSC‐CMs This framework links molecular mechanisms to cellular‐level outputs by revealing unique subsets of model parameters linked to known iPSC‐CM phenotypes
Collapse
Affiliation(s)
- Divya C Kernik
- Department of Physiology and Membrane Biology, Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, USA
| | - Stefano Morotti
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, USA
| | - HaoDi Wu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Priyanka Garg
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Henry J Duff
- Libin Cardiovascular Institute of Alberta, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Junko Kurokawa
- Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - José Jalife
- Department of Internal Medicine, Center for Arrhythmia Research, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI, USA.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), and CIBERV, Madrid, Spain
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Eleonora Grandi
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, USA
| | - Colleen E Clancy
- Department of Physiology and Membrane Biology, Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, USA
| |
Collapse
|
42
|
Veerman CC, Mengarelli I, Koopman CD, Wilders R, van Amersfoorth SC, Bakker D, Wolswinkel R, Hababa M, de Boer TP, Guan K, Milnes J, Lodder EM, Bakkers J, Verkerk AO, Bezzina CR. Genetic variation in GNB5 causes bradycardia by augmenting the cholinergic response via increased acetylcholine-activated potassium current ( I K,ACh). Dis Model Mech 2019; 12:dmm.037994. [PMID: 31208990 PMCID: PMC6679373 DOI: 10.1242/dmm.037994] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 06/06/2019] [Indexed: 12/19/2022] Open
Abstract
Mutations in GNB5, encoding the G-protein β5 subunit (Gβ5), have recently been linked to a multisystem disorder that includes severe bradycardia. Here, we investigated the mechanism underlying bradycardia caused by the recessive p.S81L Gβ5 variant. Using CRISPR/Cas9-based targeting, we generated an isogenic series of human induced pluripotent stem cell (hiPSC) lines that were either wild type, heterozygous or homozygous for the GNB5 p.S81L variant. These were differentiated into cardiomyocytes (hiPSC-CMs) that robustly expressed the acetylcholine-activated potassium channel [I(KACh); also known as IK,ACh]. Baseline electrophysiological properties of the lines did not differ. Upon application of carbachol (CCh), homozygous p.S81L hiPSC-CMs displayed an increased acetylcholine-activated potassium current (I K,ACh) density and a more pronounced decrease of spontaneous activity as compared to wild-type and heterozygous p.S81L hiPSC-CMs, explaining the bradycardia in homozygous carriers. Application of the specific I(KACh) blocker XEN-R0703 resulted in near-complete reversal of the phenotype. Our results provide mechanistic insights and proof of principle for potential therapy in patients carrying GNB5 mutations.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Christiaan C Veerman
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Isabella Mengarelli
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Charlotte D Koopman
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Hubrecht Institute, 3584 CT Utrecht, The Netherlands
| | - Ronald Wilders
- Amsterdam UMC, University of Amsterdam, Department of Medical Biology, Heart Failure Research Center, 1105 AZ Amsterdam, The Netherlands
| | - Shirley C van Amersfoorth
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Diane Bakker
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Rianne Wolswinkel
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Mariam Hababa
- Hubrecht Institute, 3584 CT Utrecht, The Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Kaomei Guan
- Department of Pharmacology and Toxicology, Technische Universität Dresden, 01062 Dresden, Germany
| | | | - Elisabeth M Lodder
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Jeroen Bakkers
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Hubrecht Institute, 3584 CT Utrecht, The Netherlands
| | - Arie O Verkerk
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands.,Amsterdam UMC, University of Amsterdam, Department of Medical Biology, Heart Failure Research Center, 1105 AZ Amsterdam, The Netherlands
| | - Connie R Bezzina
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
43
|
Sala L, Gnecchi M, Schwartz PJ. Long QT Syndrome Modelling with Cardiomyocytes Derived from Human-induced Pluripotent Stem Cells. Arrhythm Electrophysiol Rev 2019; 8:105-110. [PMID: 31114684 PMCID: PMC6528025 DOI: 10.15420/aer.2019.1.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Long QT syndrome (LQTS) is a potentially severe arrhythmogenic disorder, associated with a prolonged QT interval and sudden death, caused by mutations in key genes regulating cardiac electrophysiology. Current strategies to study LQTS in vitro include heterologous systems or animal models. Despite their value, the overwhelming power of genetic tools has exposed the many limitations of these technologies. In 2010, human-induced pluripotent stem cells (hiPSCs) revolutionised the field and allowed scientists to study in vitro some of the disease traits of LQTS on hiPSC-derived cardiomyocytes (hiPSC-CMs) from LQTS patients. In this concise review we present how the hiPSC technology has been used to model three main forms of LQTS and the severe form of LQTS associated with mutations in calmodulin. We also introduce some of the most recent challenges that must be tackled in the upcoming years to successfully shift hiPSC-CMs from powerful in vitro disease modelling tools into assets to improve risk stratification and clinical decision-making.
Collapse
Affiliation(s)
- Luca Sala
- Istituto Auxologico Italiano, IRCCS, Laboratory of Cardiovascular Genetics Milan, Italy
| | - Massimiliano Gnecchi
- Coronary Care Unit and Laboratory of Experimental Cardiology for Cell and Molecular Therapy, IRCCS Policlinico San Matteo Foundation Pavia, Italy.,Department of Medicine, University of Cape Town Cape Town, South Africa
| | - Peter J Schwartz
- Istituto Auxologico Italiano, IRCCS, Laboratory of Cardiovascular Genetics Milan, Italy.,Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin Milan, Italy.,Cardiovascular Genetics Laboratory, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town Cape Town, South Africa
| |
Collapse
|
44
|
Quach B, Krogh-Madsen T, Entcheva E, Christini DJ. Light-Activated Dynamic Clamp Using iPSC-Derived Cardiomyocytes. Biophys J 2018; 115:2206-2217. [PMID: 30447994 PMCID: PMC6289097 DOI: 10.1016/j.bpj.2018.10.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/23/2018] [Accepted: 10/02/2018] [Indexed: 01/31/2023] Open
Abstract
iPSC-derived cardiomyocytes (iPSC-CMs) are a potentially advantageous platform for drug screening because they provide a renewable source of human cardiomyocytes. One obstacle to their implementation is their immature electrophysiology, which reduces relevance to adult arrhythmogenesis. To address this, dynamic clamp is used to inject current representing the insufficient potassium current, IK1, thereby producing more adult-like electrophysiology. However, dynamic clamp requires patch clamp and is therefore low throughput and ill-suited for large-scale drug screening. Here, we use optogenetics to generate such a dynamic-clamp current. The optical dynamic clamp (ODC) uses outward-current-generating opsin, ArchT, to mimic IK1, resulting in more adult-like action potential morphology, similar to IK1 injection via classic dynamic clamp. Furthermore, in the presence of an IKr blocker, ODC revealed expected action potential prolongation and reduced spontaneous excitation. The ODC presented here still requires an electrode to measure Vm but provides a first step toward contactless dynamic clamp, which will not only enable high-throughput screening but may also allow control within multicellular iPSC-CM formats to better recapitulate adult in vivo physiology.
Collapse
Affiliation(s)
- Bonnie Quach
- Cardiovascular Research Institute, New York, New York; Weill Cornell Medicine, New York, New York
| | - Trine Krogh-Madsen
- Cardiovascular Research Institute, New York, New York; Weill Cornell Medicine, New York, New York
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - David J Christini
- Cardiovascular Research Institute, New York, New York; Weill Cornell Medicine, New York, New York.
| |
Collapse
|
45
|
de Boer TP. Using Light to Endow Stem-Cell-Derived Cardiomyocytes With Virtual I K1 Conductances. Biophys J 2018; 115:2079-2080. [PMID: 30442328 DOI: 10.1016/j.bpj.2018.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 10/19/2018] [Accepted: 10/24/2018] [Indexed: 11/17/2022] Open
Affiliation(s)
- Teun P de Boer
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
46
|
Ma D, Liu Z, Loh LJ, Zhao Y, Li G, Liew R, Islam O, Wu J, Chung YY, Teo WS, Ching CK, Tan BY, Chong D, Ho KL, Lim P, Yong RYY, Panama BK, Kaplan AD, Bett GCL, Ware J, Bezzina CR, Verkerk AO, Cook SA, Rasmusson RL, Wei H. Identification of an I Na-dependent and I to-mediated proarrhythmic mechanism in cardiomyocytes derived from pluripotent stem cells of a Brugada syndrome patient. Sci Rep 2018; 8:11246. [PMID: 30050137 PMCID: PMC6062539 DOI: 10.1038/s41598-018-29574-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 07/16/2018] [Indexed: 02/08/2023] Open
Abstract
Brugada syndrome (BrS) is an inherited cardiac arrhythmia commonly associated with SCN5A mutations, yet its ionic mechanisms remain unclear due to a lack of cellular models. Here, we used human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from a BrS patient (BrS1) to evaluate the roles of Na+ currents (INa) and transient outward K+ currents (Ito) in BrS induced action potential (AP) changes. To understand the role of these current changes in repolarization we employed dynamic clamp to “electronically express” IK1 and restore normal resting membrane potentials and allow normal recovery of the inactivating currents, INa, ICa and Ito. HiPSC-CMs were generated from BrS1 with a compound SCN5A mutation (p. A226V & p. R1629X) and a healthy sibling control (CON1). Genome edited hiPSC-CMs (BrS2) with a milder p. T1620M mutation and a commercial control (CON2) were also studied. CON1, CON2 and BrS2, had unaltered peak INa amplitudes, and normal APs whereas BrS1, with over 75% loss of INa, displayed a loss-of-INa basal AP morphology (at 1.0 Hz) manifested by a reduced maximum upstroke velocity (by ~80%, p < 0.001) and AP amplitude (p < 0.001), and an increased phase-1 repolarization pro-arrhythmic AP morphology (at 0.1 Hz) in ~25% of cells characterized by marked APD shortening (~65% shortening, p < 0.001). Moreover, Ito densities of BrS1 and CON1 were comparable and increased from 1.0 Hz to 0.1 Hz by ~ 100%. These data indicate that a repolarization deficit could be a mechanism underlying BrS.
Collapse
Affiliation(s)
- Dongrui Ma
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Zhenfeng Liu
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Li Jun Loh
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Yongxing Zhao
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Guang Li
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Reginald Liew
- Cardiovascular & Metabolic Disorders Program, Duke-NUS Medical School Singapore, Singapore, 169857, Republic of Singapore
| | - Omedul Islam
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Jianjun Wu
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Ying Ying Chung
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Wee Siong Teo
- Department of Cardiology, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Chi Keong Ching
- Department of Cardiology, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Boon Yew Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Daniel Chong
- Department of Cardiology, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Kah Leng Ho
- Department of Cardiology, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Paul Lim
- Department of Cardiology, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore
| | - Rita Yu Yin Yong
- Defense Medical and Environmental Research Institute, DSO National Laboratories, Singapore, 117510, Republic of Singapore
| | - Brian K Panama
- University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
| | - Aaron D Kaplan
- University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
| | - Glenna C L Bett
- University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
| | - James Ware
- Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Connie R Bezzina
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Arie O Verkerk
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Stuart A Cook
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore.,Cardiovascular & Metabolic Disorders Program, Duke-NUS Medical School Singapore, Singapore, 169857, Republic of Singapore.,Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Randall L Rasmusson
- University at Buffalo, State University of New York, Buffalo, NY, 14214, USA.
| | - Heming Wei
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Republic of Singapore. .,Cardiovascular & Metabolic Disorders Program, Duke-NUS Medical School Singapore, Singapore, 169857, Republic of Singapore.
| |
Collapse
|
47
|
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: 7.9] [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
|
48
|
Gélinas R, El Khoury N, Chaix MA, Beauchamp C, Alikashani A, Ethier N, Boucher G, Villeneuve L, Robb L, Latour F, Mondesert B, Rivard L, Goyette P, Talajic M, Fiset C, Rioux JD. Characterization of a Human Induced Pluripotent Stem Cell-Derived Cardiomyocyte Model for the Study of Variant Pathogenicity: Validation of a KCNJ2 Mutation. ACTA ACUST UNITED AC 2018; 10:CIRCGENETICS.117.001755. [PMID: 29021306 DOI: 10.1161/circgenetics.117.001755] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 07/10/2017] [Indexed: 01/11/2023]
Abstract
BACKGROUND Long-QT syndrome is a potentially fatal condition for which 30% of patients are without a genetically confirmed diagnosis. Rapid identification of causal mutations is thus a priority to avoid at-risk situations that can lead to fatal cardiac events. Massively parallel sequencing technologies are useful for the identification of sequence variants; however, electrophysiological testing of newly identified variants is crucial to demonstrate causality. Long-QT syndrome could, therefore, benefit from having a standardized platform for functional characterization of candidate variants in the physiological context of human cardiomyocytes. METHODS AND RESULTS Using a variant in Kir2.1 (Gly52Val) revealed by whole-exome sequencing in a patient presenting with symptoms of long-QT syndrome as a proof of principle, we demonstrated that commercially available human induced pluripotent stem cell-derived cardiomyocytes are a powerful model for screening variants involved in genetic cardiac diseases. Immunohistochemistry experiments and whole-cell current recordings in human embryonic kidney cells expressing the wild-type or the mutant Kir2.1 demonstrated that Kir2.1-52V alters channel cellular trafficking and fails to form a functional channel. Using human induced pluripotent stem cell-derived cardiomyocytes, we not only confirmed these results but also further demonstrated that Kir2.1-52V is associated with a dramatic prolongation of action potential duration with evidence of arrhythmic activity, parameters which could not have been studied using human embryonic kidney cells. CONCLUSIONS Our study confirms the pathogenicity of Kir2.1-52V in 1 patient with long-QT syndrome and also supports the use of isogenic human induced pluripotent stem cell-derived cardiomyocytes as a physiologically relevant model for the screening of variants of unknown function.
Collapse
Affiliation(s)
- Roselle Gélinas
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Nabil El Khoury
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Marie-A Chaix
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Claudine Beauchamp
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Azadeh Alikashani
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Nathalie Ethier
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Gabrielle Boucher
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Louis Villeneuve
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Laura Robb
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Frédéric Latour
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Blandine Mondesert
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Lena Rivard
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Philippe Goyette
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Mario Talajic
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada
| | - Céline Fiset
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada.
| | - John David Rioux
- From the Research Center (R.G., N.E.K., M.-A.C., C.B., A.A., N.E., G.B., L.V., L.R., F.L., B.M., L.R., P.G., M.T., C.F., J.D.R.) and Cardiovascular Genetics Center (L. Robb, L. Rivard, M.T.), Montreal Heart Institute, Quebec, Canada; and Department of Medicine (R.G., M.-A.C., B.M., L.R., M.T., J.D.R.) and Faculty of Pharmacy (N.E.K., C.F.), Université de Montréal, Quebec, Canada.
| |
Collapse
|
49
|
Brandão KO, Tabel VA, Atsma DE, Mummery CL, Davis RP. Human pluripotent stem cell models of cardiac disease: from mechanisms to therapies. Dis Model Mech 2018; 10:1039-1059. [PMID: 28883014 PMCID: PMC5611968 DOI: 10.1242/dmm.030320] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
It is now a decade since human induced pluripotent stem cells (hiPSCs) were first described. The reprogramming of adult somatic cells to a pluripotent state has become a robust technology that has revolutionised our ability to study human diseases. Crucially, these cells capture all the genetic aspects of the patient from which they were derived. Combined with advances in generating the different cell types present in the human heart, this has opened up new avenues to study cardiac disease in humans and investigate novel therapeutic approaches to treat these pathologies. Here, we provide an overview of the current state of the field regarding the generation of cardiomyocytes from human pluripotent stem cells and methods to assess them functionally, an essential requirement when investigating disease and therapeutic outcomes. We critically evaluate whether treatments suggested by these in vitro models could be translated to clinical practice. Finally, we consider current shortcomings of these models and propose methods by which they could be further improved.
Collapse
Affiliation(s)
- Karina O Brandão
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Viola A Tabel
- 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
| | - Christine L Mummery
- 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
| |
Collapse
|
50
|
Horváth A, Lemoine MD, Löser A, Mannhardt I, Flenner F, Uzun AU, Neuber C, Breckwoldt K, Hansen A, Girdauskas E, Reichenspurner H, Willems S, Jost N, Wettwer E, Eschenhagen T, Christ T. Low Resting Membrane Potential and Low Inward Rectifier Potassium Currents Are Not Inherent Features of hiPSC-Derived Cardiomyocytes. Stem Cell Reports 2018; 10:822-833. [PMID: 29429959 PMCID: PMC5918194 DOI: 10.1016/j.stemcr.2018.01.012] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 01/12/2018] [Accepted: 01/15/2018] [Indexed: 11/18/2022] Open
Abstract
Human induced pluripotent stem cell (hiPSC) cardiomyocytes (CMs) show less negative resting membrane potential (RMP), which is attributed to small inward rectifier currents (IK1). Here, IK1 was measured in hiPSC-CMs (proprietary and commercial cell line) cultured as monolayer (ML) or 3D engineered heart tissue (EHT) and, for direct comparison, in CMs from human right atrial (RA) and left ventricular (LV) tissue. RMP was measured in isolated cells and intact tissues. IK1 density in ML- and EHT-CMs from the proprietary line was similar to LV and RA, respectively. IK1 density in EHT-CMs from the commercial line was 2-fold smaller than in the proprietary line. RMP in EHT of both lines was similar to RA and LV. Repolarization fraction and IK,ACh response discriminated best between RA and LV and indicated predominantly ventricular phenotype in hiPSC-CMs/EHT. The data indicate that IK1 is not necessarily low in hiPSC-CMs, and technical issues may underlie low RMP in hiPSC-CMs.
Collapse
Affiliation(s)
- András Horváth
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany; Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, 6721 Szeged, Hungary
| | - Marc D Lemoine
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; Department of Cardiology-Electrophysiology, University Heart Center Hamburg, 20246 Hamburg, Germany
| | - Alexandra Löser
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Ingra Mannhardt
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Frederik Flenner
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Ahmet Umur Uzun
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Christiane Neuber
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Kaja Breckwoldt
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Arne Hansen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Evaldas Girdauskas
- Department of Cardiovascular Surgery, University Heart Center Hamburg, 20246 Hamburg, Germany
| | - Hermann Reichenspurner
- Department of Cardiovascular Surgery, University Heart Center Hamburg, 20246 Hamburg, Germany
| | - Stephan Willems
- Department of Cardiology-Electrophysiology, University Heart Center Hamburg, 20246 Hamburg, Germany
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, 6721 Szeged, Hungary
| | - Erich Wettwer
- Institute of Pharmacology, University Duisburg-Essen, 45122 Essen, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Torsten Christ
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany.
| |
Collapse
|