1
|
Begovic M, Schneider L, Zhou X, Hamdani N, Akin I, El-Battrawy I. The Role of Human-Induced Pluripotent Stem Cells in Studying Cardiac Channelopathies. Int J Mol Sci 2024; 25:12034. [PMID: 39596103 PMCID: PMC11593457 DOI: 10.3390/ijms252212034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/22/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
Cardiac channelopathies are inherited diseases that increase the risk of sudden cardiac death. While different genes have been associated with inherited channelopathies, there are still subtypes, e.g., catecholaminergic polymorphic ventricular tachycardia and Brugada syndrome, where the genetic cause remains unknown. Various models, including animal models, heterologous expression systems, and the human-induced pluripotent stem-cell-derived cardiomyocytes (hiPSCs-CMs) model, have been used to study the pathophysiological mechanisms of channelopathies. Recently, researchers have focused on using hiPSCs-CMs to understand the genotype-phenotype correlation and screen drugs. By combining innovative techniques such as Clustered Regularly Interspaced Short Palindromic Repeats/Clustered Regularly Interspaced Short Palindromic Repeats associated protein 9 (CRISPR/Cas9)-mediated genome editing, and three-dimensional (3D) engineered heart tissues, we can gain new insights into the pathophysiological mechanisms of channelopathies. This approach holds promise for improving personalized drug treatment. This review highlights the role of hiPSCs-CMs in understanding the pathomechanism of Brugada syndrome and catecholaminergic polymorphic ventricular tachycardia and how these models can be utilized for drug screening.
Collapse
Affiliation(s)
- Merima Begovic
- Institute of Physiology, Department of Cellular and Translational Physiology, Ruhr-University Bochum, 44801 Bochum, Germany; (M.B.); (L.S.); (N.H.)
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Luca Schneider
- Institute of Physiology, Department of Cellular and Translational Physiology, Ruhr-University Bochum, 44801 Bochum, Germany; (M.B.); (L.S.); (N.H.)
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Xiaobo Zhou
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Nazha Hamdani
- Institute of Physiology, Department of Cellular and Translational Physiology, Ruhr-University Bochum, 44801 Bochum, Germany; (M.B.); (L.S.); (N.H.)
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Physiology, Cardiovascular Research Institute, University Maastricht, 6229HX Maastricht, The Netherlands
- HCEMM-SU Cardiovascular Comorbidities Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Intézet címe Semmelweis University, 1089 Budapest, Hungary
- Department of Cardiology and Rhythmology, St. Josef Hospital, Ruhr University, 44791 Bochum, Germany
| | - Ibrahim Akin
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Ibrahim El-Battrawy
- Institute of Physiology, Department of Cellular and Translational Physiology, Ruhr-University Bochum, 44801 Bochum, Germany; (M.B.); (L.S.); (N.H.)
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Cardiology and Rhythmology, St. Josef Hospital, Ruhr University, 44791 Bochum, Germany
| |
Collapse
|
2
|
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
|
3
|
Martínez-Moreno R, Carreras D, Sarquella-Brugada G, Pérez GJ, Selga E, Scornik FS, Brugada R. Loss of sodium current caused by a Brugada syndrome-associated variant is determined by patient-specific genetic background. Heart Rhythm 2024; 21:331-339. [PMID: 38008367 DOI: 10.1016/j.hrthm.2023.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND Brugada syndrome (BrS) is an inherited cardiac arrhythmogenic disease that predisposes patients to sudden cardiac death. It is associated with mutations in SCN5A, which encodes the cardiac sodium channel alpha subunit (NaV1.5). BrS-related mutations have incomplete penetrance and variable expressivity within families. OBJECTIVE The purpose of this study was to determine the role of patient-specific genetic background on the cellular and clinical phenotype among carriers of NaV1.5_p.V1525M. METHODS We studied sodium currents from patient-specific human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and heterologously transfected human embryonic kidney (HEK) tsA201 cells using the whole-cell patch-clamp technique. We determined gene and protein expression by quantitative polymerase chain reaction, RNA sequencing, and western blot and performed a genetic panel for arrhythmogenic diseases. RESULTS Our results showed a large reduction in INa density in hiPSC-CM derived from 2 V1525M single nucleotide variant (SNV) carriers compared with hiPSC-CM derived from a noncarrier, suggesting a dominant-negative effect of the NaV1.5_p.V1525M channel. INa was not affected in hiPSC-CMs derived from a V1525M SNV carrier who also carries the NaV1.5_p.H558R polymorphism. Heterozygous expression of V1525M in HEK-293T cells produced a loss of INa function, not observed when this variant was expressed together with H558R. In addition, the antiarrhythmic drug mexiletine rescued INa function in hiPSC-CM. SCN5A expression was increased in the V1525M carrier who also expresses NaV1.5_p.H558R. CONCLUSION Our results in patient-specific hiPSC-CM point to a dominant-negative effect of NaV1.5_p.V1525M, which can be reverted by the presence of NaV1.5_p.H558R. Overall, our data points to a role of patient-specific genetic background as a determinant for incomplete penetrance in BrS.
Collapse
Affiliation(s)
- Rebecca Martínez-Moreno
- Departament de Ciències Mèdiques, Facultat de Medicina, Universitat de Girona, Girona, Spain; Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta, Girona, Spain
| | - David Carreras
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta, Girona, Spain
| | - Georgia Sarquella-Brugada
- Departament de Ciències Mèdiques, Facultat de Medicina, Universitat de Girona, Girona, Spain; Arrhythmia, Inherited Cardiac Diseases and Sudden Death Unit, Hospital Sant Joan de Déu, Esplugues, Barcelona, Spain; Arrítmies pediàtriques, cardiologia genètica i mort sobtada. Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Guillermo J Pérez
- Departament de Ciències Mèdiques, Facultat de Medicina, Universitat de Girona, Girona, Spain; Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta, Girona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| | - Elisabet Selga
- Departament de Ciències Mèdiques, Facultat de Medicina, Universitat de Girona, Girona, Spain; Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta, Girona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain.
| | - Fabiana S Scornik
- Departament de Ciències Mèdiques, Facultat de Medicina, Universitat de Girona, Girona, Spain; Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta, Girona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| | - Ramon Brugada
- Departament de Ciències Mèdiques, Facultat de Medicina, Universitat de Girona, Girona, Spain; Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta, Girona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Hospital Josep Trueta, Girona, Spain
| |
Collapse
|
4
|
Abrasheva VO, Kovalenko SG, Slotvitsky M, Romanova SА, Aitova AA, Frolova S, Tsvelaya V, Syunyaev RA. Human sodium current voltage-dependence at physiological temperature measured by coupling a patch-clamp experiment to a mathematical model. J Physiol 2024; 602:633-661. [PMID: 38345560 DOI: 10.1113/jp285162] [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: 06/16/2023] [Accepted: 01/02/2024] [Indexed: 02/20/2024] Open
Abstract
Voltage-gated Na+ channels are crucial to action potential propagation in excitable tissues. Because of the high amplitude and rapid activation of the Na+ current, voltage-clamp measurements are very challenging and are usually performed at room temperature. In this study, we measured Na+ current voltage-dependence in stem cell-derived cardiomyocytes at physiological temperature. While the apparent activation and inactivation curves, measured as the dependence of current amplitude on voltage, fall within the range reported in previous studies, we identified a systematic error in our measurements. This error is caused by the deviation of the membrane potential from the command potential of the amplifier. We demonstrate that it is possible to account for this artifact using computer simulation of the patch-clamp experiment. We obtained surprising results through patch-clamp model optimization: a half-activation of -11.5 mV and a half-inactivation of -87 mV. Although the half-activation deviates from previous research, we demonstrate that this estimate reproduces the conduction velocity dependence on extracellular potassium concentration. KEY POINTS: Voltage-gated Na+ currents play a crucial role in excitable tissues including neurons, cardiac and skeletal muscle. Measurement of Na+ current is challenging because of its high amplitude and rapid kinetics, especially at physiological temperature. We have used the patch-clamp technique to measure human Na+ current voltage-dependence in human induced pluripotent stem cell-derived cardiomyocytes. The patch-clamp data were processed by optimization of the model accounting for voltage-clamp experiment artifacts, revealing a large difference between apparent parameters of Na+ current and the results of the optimization. We conclude that actual Na+ current activation is extremely depolarized in comparison to previous studies. The new Na+ current model provides a better understanding of action potential propagation; we demonstrate that it explains propagation in hyperkalaemic conditions.
Collapse
Affiliation(s)
| | - Sandaara G Kovalenko
- Moscow Institute of Physics and Technology, Moscow, Russia
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
- ITMO University, St Petersburg, Russia
| | - Mihail Slotvitsky
- Moscow Institute of Physics and Technology, Moscow, Russia
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
- ITMO University, St Petersburg, Russia
| | - Serafima А Romanova
- Moscow Institute of Physics and Technology, Moscow, Russia
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
| | - Aleria A Aitova
- Moscow Institute of Physics and Technology, Moscow, Russia
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
- ITMO University, St Petersburg, Russia
| | - Sheida Frolova
- Moscow Institute of Physics and Technology, Moscow, Russia
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
| | - Valeria Tsvelaya
- Moscow Institute of Physics and Technology, Moscow, Russia
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
- ITMO University, St Petersburg, Russia
| | | |
Collapse
|
5
|
Theisen B, Holtz A, Rajagopalan V. Noncoding RNAs and Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes in Cardiac Arrhythmic Brugada Syndrome. Cells 2023; 12:2398. [PMID: 37830612 PMCID: PMC10571919 DOI: 10.3390/cells12192398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/14/2023] Open
Abstract
Hundreds of thousands of people die each year as a result of sudden cardiac death, and many are due to heart rhythm disorders. One of the major causes of these arrhythmic events is Brugada syndrome, a cardiac channelopathy that results in abnormal cardiac conduction, severe life-threatening arrhythmias, and, on many occasions, death. This disorder has been associated with mutations and dysfunction of about two dozen genes; however, the majority of the patients do not have a definite cause for the diagnosis of Brugada Syndrome. The protein-coding genes represent only a very small fraction of the mammalian genome, and the majority of the noncoding regions of the genome are actively transcribed. Studies have shown that most of the loci associated with electrophysiological traits are located in noncoding regulatory regions and are expected to affect gene expression dosage and cardiac ion channel function. Noncoding RNAs serve an expanding number of regulatory and other functional roles within the cells, including but not limited to transcriptional, post-transcriptional, and epigenetic regulation. The major noncoding RNAs found in Brugada Syndrome include microRNAs; however, others such as long noncoding RNAs are also identified. They contribute to pathogenesis by interacting with ion channels and/or are detectable as clinical biomarkers. Stem cells have received significant attention in the recent past, and can be differentiated into many different cell types including those in the heart. In addition to contractile and relaxational properties, BrS-relevant electrophysiological phenotypes are also demonstrated in cardiomyocytes differentiated from stem cells induced from adult human cells. In this review, we discuss the current understanding of noncoding regions of the genome and their RNA biology in Brugada Syndrome. We also delve into the role of stem cells, especially human induced pluripotent stem cell-derived cardiac differentiated cells, in the investigation of Brugada syndrome in preclinical and clinical studies.
Collapse
Affiliation(s)
- Benjamin Theisen
- Department of Biomedical and Anatomical Sciences, New York Institute of Technology College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR 72401, USA
| | - Austin Holtz
- Department of Biomedical and Anatomical Sciences, New York Institute of Technology College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR 72401, USA
| | - Viswanathan Rajagopalan
- Department of Biomedical and Anatomical Sciences, New York Institute of Technology College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR 72401, USA
- Arkansas Biosciences Institute, Jonesboro, AR 72401, USA
| |
Collapse
|
6
|
Cai D, Wang X, Sun Y, Fan H, Zhou J, Yang Z, Qiu H, Wang J, Su J, Gong T, Jiang C, Liang P. Patient-specific iPSC-derived cardiomyocytes reveal aberrant activation of Wnt/β-catenin signaling in SCN5A-related Brugada syndrome. Stem Cell Res Ther 2023; 14:241. [PMID: 37679791 PMCID: PMC10486057 DOI: 10.1186/s13287-023-03477-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Mutations in the cardiac sodium channel gene SCN5A cause Brugada syndrome (BrS), an arrhythmic disorder that is a leading cause of sudden death and lacks effective treatment. An association between SCN5A and Wnt/β-catenin signaling has been recently established. However, the role of Wnt/β-catenin signaling in BrS and underlying mechanisms remains unknown. METHODS Three healthy control subjects and one BrS patient carrying a novel frameshift mutation (T1788fs) in the SCN5A gene were recruited in this study. Control and BrS patient-specific induced pluripotent stem cells (iPSCs) were generated from skin fibroblasts using nonintegrated Sendai virus. All iPSCs were differentiated into cardiomyocytes using monolayer-based differentiation protocol. Action potentials and sodium currents were recorded from control and BrS iPSC-derived cardiomyocytes (iPSC-CMs) by single-cell patch clamp. RESULTS BrS iPSC-CMs exhibited increased burden of arrhythmias and abnormal action potential profile featured by slower depolarization, decreased action potential amplitude, and increased beating interval variation. Moreover, BrS iPSC-CMs showed cardiac sodium channel (Nav1.5) loss-of-function as compared to control iPSC-CMs. Interestingly, the electrophysiological abnormalities and Nav1.5 loss-of-function observed in BrS iPSC-CMs were accompanied by aberrant activation of Wnt/β-catenin signaling. Notably, inhibition of Wnt/β-catenin significantly rescued Nav1.5 defects and arrhythmic phenotype in BrS iPSC-CMs. Mechanistically, SCN5A-encoded Nav1.5 interacts with β-catenin, and reduced expression of Nav1.5 leads to re-localization of β-catenin in BrS iPSC-CMs, which aberrantly activates Wnt/β-catenin signaling to suppress SCN5A transcription. CONCLUSIONS Our findings suggest that aberrant activation of Wnt/β-catenin signaling contributes to the pathogenesis of SCN5A-related BrS and point to Wnt/β-catenin as a potential therapeutic target.
Collapse
Affiliation(s)
- Dongsheng Cai
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, China
| | - Xiaochen Wang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Yaxun Sun
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, China
| | - Hangping Fan
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Jingjun Zhou
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Zongkuai Yang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Hangyuan Qiu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, China
| | - Jue Wang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Jun Su
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Tingyu Gong
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Chenyang Jiang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, China.
| | - Ping Liang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China.
| |
Collapse
|
7
|
Sun Y, Su J, Wang X, Wang J, Guo F, Qiu H, Fan H, Cai D, Wang H, Lin M, Wang W, Feng Y, Fu G, Gong T, Liang P, Jiang C. Patient-specific iPSC-derived cardiomyocytes reveal variable phenotypic severity of Brugada syndrome. EBioMedicine 2023; 95:104741. [PMID: 37544203 PMCID: PMC10427992 DOI: 10.1016/j.ebiom.2023.104741] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Brugada syndrome (BrS) is a cardiac channelopathy that can result in sudden cardiac death (SCD). SCN5A is the most frequent gene linked to BrS, but the genotype-phenotype correlations are not completely matched. Clinical phenotypes of a particular SCN5A variant may range from asymptomatic to SCD. Here, we used comparison of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) derived from a SCN5A mutation-positive (D356Y) BrS family with severely affected proband, asymptomatic mutation carriers (AMCs) and healthy controls to investigate this variation. METHODS 26 iPSC lines were generated from skin fibroblasts using nonintegrated Sendai virus. The generated iPSCs were differentiated into cardiomyocytes using a monolayer-based differentiation protocol. FINDINGS D356Y iPSC-CMs exhibited increased beat interval variability, slower depolarization, cardiac arrhythmias, defects of Na+ channel function and irregular Ca2+ signaling, when compared to controls. Importantly, the phenotype severity observed in AMC iPSC-CMs was milder than that of proband iPSC-CMs, an observation exacerbated by flecainide. Interestingly, the iPSC-CMs of the proband exhibited markedly decreased Ca2+ currents in comparison with control and AMC iPSC-CMs. CRISPR/Cas9-mediated genome editing to correct D356Y in proband iPSC-CMs effectively rescued the arrhythmic phenotype and restored Na+ and Ca2+ currents. Moreover, drug screening using established BrS iPSC-CM models demonstrated that quinidine and sotalol possessed antiarrhythmic effects in an individual-dependent manner. Clinically, venous and oral administration of calcium partially reduced the malignant arrhythmic events of the proband in mid-term follow-up. INTERPRETATION Patient-specific and genome-edited iPSC-CMs can recapitulate the varying phenotypic severity of BrS. Our findings suggest that preservation of the Ca2+ currents might be a compensatory mechanism to resist arrhythmogenesis in BrS AMCs. FUNDING National Key R&D Program of China (2017YFA0103700), National Natural Science Foundation of China (81922006, 81870175), Natural Science Foundation of Zhejiang Province (LD21H020001, LR15H020001), National Natural Science Foundation of China (81970269), Key Research and Development Program of Zhejiang Province (2019C03022) and Natural Science Foundation of Zhejiang Province (LY16H020002).
Collapse
Affiliation(s)
- Yaxun Sun
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China
| | - Jun Su
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China; Institute of Translational Medicine, Zhejiang University, 310029, Hangzhou, China
| | - Xiaochen Wang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China; Institute of Translational Medicine, Zhejiang University, 310029, Hangzhou, China
| | - Jue Wang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China; Institute of Translational Medicine, Zhejiang University, 310029, Hangzhou, China
| | - Fengfeng Guo
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China; Institute of Translational Medicine, Zhejiang University, 310029, Hangzhou, China
| | - Hangyuan Qiu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China
| | - Hangping Fan
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China; Institute of Translational Medicine, Zhejiang University, 310029, Hangzhou, China
| | - Dongsheng Cai
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China
| | - Hao Wang
- Prenatal Diagnosis Center, Hangzhou Women's Hospital, Hangzhou, 310008, China
| | - Miao Lin
- Department of Cardiology, Wenzhou Central Hospital, 325000, Wenzhou, China
| | - Wei Wang
- Jiangxi Provincial Cardiovascular Disease Research Institute, Jiangxi Provincial People's Hospital, Nanchang, 330006, China
| | - Ye Feng
- Institute of Translational Medicine, Zhejiang University, 310029, Hangzhou, China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China
| | - Tingyu Gong
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China; Institute of Translational Medicine, Zhejiang University, 310029, Hangzhou, China; Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Ping Liang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China; Institute of Translational Medicine, Zhejiang University, 310029, Hangzhou, China.
| | - Chenyang Jiang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China.
| |
Collapse
|
8
|
Nasilli G, Yiangou L, Palandri C, Cerbai E, Davis RP, Verkerk AO, Casini S, Remme CA. Beneficial effects of chronic mexiletine treatment in a human model of SCN5A overlap syndrome. Europace 2023; 25:euad154. [PMID: 37369559 PMCID: PMC10299896 DOI: 10.1093/europace/euad154] [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] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
AIMS SCN5A mutations are associated with various cardiac phenotypes, including long QT syndrome type 3 (LQT3), Brugada syndrome (BrS), and cardiac conduction disease (CCD). Certain mutations, such as SCN5A-1795insD, lead to an overlap syndrome, with patients exhibiting both features of BrS/CCD [decreased sodium current (INa)] and LQT3 (increased late INa). The sodium channel blocker mexiletine may acutely decrease LQT3-associated late INa and chronically increase peak INa associated with SCN5A loss-of-function mutations. However, most studies have so far employed heterologous expression systems and high mexiletine concentrations. We here investigated the effects of a therapeutic dose of mexiletine on the mixed phenotype associated with the SCN5A-1795insD mutation in HEK293A cells and human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). METHODS AND RESULTS To assess only the chronic effects on trafficking, HEK293A cells transfected with wild-type (WT) SCN5A or SCN5A-1795insD were incubated for 48 h with 10 µm mexiletine followed by wash-out, which resulted in an increased peak INa for both SCN5A-WT and SCN5A-1795insD and an increased late INa for SCN5A-1795insD. Acute re-exposure of HEK293A cells to 10 µm mexiletine did not impact on peak INa but significantly decreased SCN5A-1795insD late INa. Chronic incubation of SCN5A-1795insD hiPSC-CMs with mexiletine followed by wash-out increased peak INa, action potential (AP) upstroke velocity, and AP duration. Acute re-exposure did not impact on peak INa or AP upstroke velocity, but significantly decreased AP duration. CONCLUSION These findings demonstrate for the first time the therapeutic benefit of mexiletine in a human cardiomyocyte model of SCN5A overlap syndrome.
Collapse
Affiliation(s)
- Giovanna Nasilli
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, The Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Chiara Palandri
- Department NeuroFarBa, University of Florence, Viale Gaetano Pieraccini 6, 50139, Florence, Italy
| | - Elisabetta Cerbai
- Department NeuroFarBa, University of Florence, Viale Gaetano Pieraccini 6, 50139, Florence, Italy
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Arie O Verkerk
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Simona Casini
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, The Netherlands
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Penttinen K, Prajapati C, Shah D, Rajan DK, Cherian RM, Swan H, Aalto-Setälä K. HiPSC-derived cardiomyocyte to model Brugada syndrome: both asymptomatic and symptomatic mutation carriers reveal increased arrhythmogenicity. BMC Cardiovasc Disord 2023; 23:208. [PMID: 37098502 PMCID: PMC10131315 DOI: 10.1186/s12872-023-03234-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/11/2023] [Indexed: 04/27/2023] Open
Abstract
Brugada syndrome is an inherited cardiac arrhythmia disorder that is mainly associated with mutations of the cardiac voltage-gated sodium channel alpha subunit 5 (SCN5A) gene. The clinical symptoms include ventricular fibrillation and an increased risk of sudden cardiac death. Human-induced pluripotent stem cell (hiPSC) lines were derived from symptomatic and asymptomatic individuals carrying the R1913C mutation in the SCN5A gene. The present work aimed to observe the phenotype-specific differences in hiPSC-derived cardiomyocytes (CMs) obtained from symptomatic and asymptomatic mutation carriers. In this study, CM electrophysiological properties, beating abilities and calcium parameters were measured. Mutant CMs exhibited higher average sodium current densities than healthy CMs, but the differences were not statistically significant. Action potential durations were significantly shorter in CMs from the symptomatic individual, and a spike-and-dome morphology of action potential was exclusively observed in CMs from the symptomatic individual. More arrhythmias occurred in mutant CMs at single cell and cell aggregate levels compared with those observed in wild-type CMs. Moreover, there were no major differences in ionic currents or intracellular calcium dynamics between the CMs of asymptomatic and symptomatic individuals after the administration of adrenaline and flecainide.In conclusion, mutant CMs were more prone to arrhythmia than healthy CMs but did not explain why only one of the mutation carriers was symptomatic.
Collapse
Affiliation(s)
- Kirsi Penttinen
- Faculty of Medicine and Health Technology and BioMediTech Institute, Tampere University, Tampere, 33520, Finland
| | - Chandra Prajapati
- Faculty of Medicine and Health Technology and BioMediTech Institute, Tampere University, Tampere, 33520, Finland.
| | - Disheet Shah
- Faculty of Medicine and Health Technology and BioMediTech Institute, Tampere University, Tampere, 33520, Finland
| | - Dhanesh Kattipparambil Rajan
- Faculty of Medicine and Health Technology and BioMediTech Institute, Tampere University, Tampere, 33520, Finland
| | - Reeja Maria Cherian
- Faculty of Medicine and Health Technology and BioMediTech Institute, Tampere University, Tampere, 33520, Finland
| | - Heikki Swan
- Helsinki University Hospital, Helsinki, 00290, Finland
| | - Katriina Aalto-Setälä
- Faculty of Medicine and Health Technology and BioMediTech Institute, Tampere University, Tampere, 33520, Finland
- Heart Hospital, Tampere University Hospital, Tampere, 33520, Finland
| |
Collapse
|
10
|
Melgari D, Calamaio S, Frosio A, Prevostini R, Anastasia L, Pappone C, Rivolta I. Automated Patch-Clamp and Induced Pluripotent Stem Cell-Derived Cardiomyocytes: A Synergistic Approach in the Study of Brugada Syndrome. Int J Mol Sci 2023; 24:ijms24076687. [PMID: 37047659 PMCID: PMC10095337 DOI: 10.3390/ijms24076687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023] Open
Abstract
The development of high-throughput automated patch-clamp technology is a recent breakthrough in the field of Brugada syndrome research. Brugada syndrome is a heart disorder marked by abnormal electrocardiographic readings and an elevated risk of sudden cardiac death due to arrhythmias. Various experimental models, developed either in animals, cell lines, human tissue or computational simulation, play a crucial role in advancing our understanding of this condition, and developing effective treatments. In the perspective of the pathophysiological role of ion channels and their pharmacology, automated patch-clamp involves a robotic system that enables the simultaneous recording of electrical activity from multiple single cells at once, greatly improving the speed and efficiency of data collection. By combining this approach with the use of patient-derived cardiomyocytes, researchers are gaining a more comprehensive view of the underlying mechanisms of heart disease. This has led to the development of more effective treatments for those affected by cardiovascular conditions.
Collapse
Affiliation(s)
- Dario Melgari
- Institute of Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Serena Calamaio
- Institute of Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Anthony Frosio
- Institute of Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Rachele Prevostini
- Institute of Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Luigi Anastasia
- Institute of Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Carlo Pappone
- Institute of Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Arrhythmology Department, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Ilaria Rivolta
- Institute of Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore, 48, 20900 Monza, Italy
| |
Collapse
|
11
|
Campostrini G, Kosmidis G, Ward-van Oostwaard D, Davis RP, Yiangou L, Ottaviani D, Veerman CC, Mei H, Orlova VV, Wilde AAM, Bezzina CR, Verkerk AO, Mummery CL, Bellin M. Maturation of hiPSC-derived cardiomyocytes promotes adult alternative splicing of SCN5A and reveals changes in sodium current associated with cardiac arrhythmia. Cardiovasc Res 2023; 119:167-182. [PMID: 35394010 PMCID: PMC10022870 DOI: 10.1093/cvr/cvac059] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/07/2022] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS Human-induced pluripotent stem cell-cardiomyocytes (hiPSC-CMs) are widely used to study arrhythmia-associated mutations in ion channels. Among these, the cardiac sodium channel SCN5A undergoes foetal-to-adult isoform switching around birth. Conventional hiPSC-CM cultures, which are phenotypically foetal, have thus far been unable to capture mutations in adult gene isoforms. Here, we investigated whether tri-cellular cross-talk in a three-dimensional (3D) cardiac microtissue (MT) promoted post-natal SCN5A maturation in hiPSC-CMs. METHODS AND RESULTS We derived patient hiPSC-CMs carrying compound mutations in the adult SCN5A exon 6B and exon 4. Electrophysiological properties of patient hiPSC-CMs in monolayer were not altered by the exon 6B mutation compared with isogenic controls since it is not expressed; further, CRISPR/Cas9-mediated excision of the foetal exon 6A did not promote adult SCN5A expression. However, when hiPSC-CMs were matured in 3D cardiac MTs, SCN5A underwent isoform switch and the functional consequences of the mutation located in exon 6B were revealed. Up-regulation of the splicing factor muscleblind-like protein 1 (MBNL1) drove SCN5A post-natal maturation in microtissues since its overexpression in hiPSC-CMs was sufficient to promote exon 6B inclusion, whilst knocking-out MBNL1 failed to foster isoform switch. CONCLUSIONS Our study shows that (i) the tri-cellular cardiac microtissues promote post-natal SCN5A isoform switch in hiPSC-CMs, (ii) adult splicing of SCN5A is driven by MBNL1 in these tissues, and (iii) this model can be used for examining post-natal cardiac arrhythmias due to mutations in the exon 6B. TRANSLATIONAL PERSPECTIVE The cardiac sodium channel is essential for conducting the electrical impulse in the heart. Postnatal alternative splicing regulation causes mutual exclusive inclusion of fetal or adult exons of the corresponding gene, SCN5A. Typically, immature hiPSCCMs fall short in studying the effect of mutations located in the adult exon. We describe here that an innovative tri-cellular three-dimensional cardiac microtissue culture promotes hiPSC-CMs maturation through upregulation of MBNL1, thus revealing the effect of a pathogenic genetic variant located in the SCN5A adult exon. These results help advancing the use of hiPSC-CMs in studying adult heart disease and for developing personalized medicine applications.
Collapse
Affiliation(s)
- Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Georgios Kosmidis
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Dorien Ward-van Oostwaard
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Richard Paul Davis
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Daniele Ottaviani
- Department of Biology, University of Padua, 35121 Padua, Italy
- Veneto Institute of Molecular Medicine, 35129 Padua, Italy
| | - Christiaan Cornelis Veerman
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam University Medical Centre, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, 2333 Leiden, The Netherlands
| | - Valeria Viktorovna Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Arthur Arnold Maria Wilde
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam University Medical Centre, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Connie Rose Bezzina
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam University Medical Centre, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Arie Otto Verkerk
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam University Medical Centre, location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Christine Lindsay Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, 7500 AE, Enschede, The Netherlands
| | | |
Collapse
|
12
|
iPSC-Derived Cardiomyocytes in Inherited Cardiac Arrhythmias: Pathomechanistic Discovery and Drug Development. Biomedicines 2023; 11:biomedicines11020334. [PMID: 36830871 PMCID: PMC9953535 DOI: 10.3390/biomedicines11020334] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/20/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023] Open
Abstract
With the discovery of induced pluripotent stem cell (iPSCs) a wide range of cell types, including iPSC-derived cardiomyocytes (iPSC-CM), can now be generated from an unlimited source of somatic cells. These iPSC-CM are used for different purposes such as disease modelling, drug discovery, cardiotoxicity testing and personalised medicine. The 2D iPSC-CM models have shown promising results, but they are known to be more immature compared to in vivo adult cardiomyocytes. Novel approaches to create 3D models with the possible addition of other (cardiac) cell types are being developed. This will not only improve the maturity of the cells, but also leads to more physiologically relevant models that more closely resemble the human heart. In this review, we focus on the progress in the modelling of inherited cardiac arrhythmias in both 2D and 3D and on the use of these models in therapy development and drug testing.
Collapse
|
13
|
Ko TH, Jeong D, Yu B, Song JE, Le QA, Woo SH, Choi JI. Inhibition of late sodium current via PI3K/Akt signaling prevents cellular remodeling in tachypacing-induced HL-1 atrial myocytes. Pflugers Arch 2023; 475:217-231. [PMID: 36274100 PMCID: PMC9849166 DOI: 10.1007/s00424-022-02754-z] [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: 10/29/2021] [Revised: 07/04/2022] [Accepted: 09/23/2022] [Indexed: 02/01/2023]
Abstract
An aberrant late sodium current (INa,Late) caused by a mutation in the cardiac sodium channel (Nav1.5) has emerged as a contributor to electrical remodeling that causes susceptibility to atrial fibrillation (AF). Although downregulation of phosphoinositide 3-kinase (PI3K)/Akt signaling is associated with AF, the molecular mechanisms underlying the negative regulation of INa,Late in AF remain unclear, and potential therapeutic approaches are needed. In this work, we constructed a tachypacing-induced cellular model of AF by exposing HL-1 myocytes to rapid electrical stimulation (1.5 V/cm, 4 ms, 10 Hz) for 6 h. Then, we gathered data using confocal Ca2+ imaging, immunofluorescence, patch-clamp recordings, and immunoblots. The tachypacing cells displayed irregular Ca2+ release, delayed afterdepolarization, prolonged action potential duration, and reduced PI3K/Akt signaling compared with controls. Those detrimental effects were related to increased INa,Late and were significantly mediated by treatment with the INa,Late blocker ranolazine. Furthermore, decreased PI3K/Akt signaling via PI3K inhibition increased INa,Late and subsequent aberrant myocyte excitability, which were abolished by INa,Late inhibition, suggesting that PI3K/Akt signaling is responsible for regulating pathogenic INa,Late. These results indicate that PI3K/Akt signaling is critical for regulating INa,Late and electrical remodeling, supporting the use of PI3K/Akt-mediated INa,Late as a therapeutic target for AF.
Collapse
Affiliation(s)
- Tae Hee Ko
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Medical Centre, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841 Republic of Korea ,Ion Channel Research Unit, Cardiovascular Research Institute, Korea University, Seoul, Republic of Korea
| | - Daun Jeong
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Medical Centre, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841 Republic of Korea
| | - Byeongil Yu
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Medical Centre, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841 Republic of Korea
| | - Ji Eun Song
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Medical Centre, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841 Republic of Korea
| | - Qui Anh Le
- Laboratory of Pathophysiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134 Republic of Korea
| | - Sun-Hee Woo
- Laboratory of Pathophysiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134 Republic of Korea
| | - Jong-Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Medical Centre, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841 Republic of Korea ,Ion Channel Research Unit, Cardiovascular Research Institute, Korea University, Seoul, Republic of Korea
| |
Collapse
|
14
|
Fujiwara K, Yamamoto R, Kubota T, Tazumi A, Sabuta T, Takahashi MP, Sakurai H. Mature Myotubes Generated From Human-Induced Pluripotent Stem Cells Without Forced Gene Expression. Front Cell Dev Biol 2022; 10:886879. [PMID: 35706901 PMCID: PMC9189389 DOI: 10.3389/fcell.2022.886879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/11/2022] [Indexed: 11/17/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) are a promising tool for disease modeling and drug screening. To apply them to skeletal muscle disorders, it is necessary to establish mature myotubes because the onset of many skeletal muscle disorders is after birth. However, to make mature myotubes, the forced expression of specific genes should be avoided, as otherwise dysregulation of the intracellular networks may occur. Here, we achieved this goal by purifying hiPSC-derived muscle stem cells (iMuSC) by Pax7-fluorescence monitoring and antibody sorting. The resulting myotubes displayed spontaneous self-contraction, aligned sarcomeres, and a triad structure. Notably, the phenotype of sodium channels was changed to the mature type in the course of the differentiation, and a characteristic current pattern was observed. Moreover, the protocol resulted in highly efficient differentiation and high homogeneity and is applicable to drug screening.
Collapse
Affiliation(s)
- Kei Fujiwara
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Risa Yamamoto
- Clinical Neurophysiology, Department of Clinical Laboratory and Biomedical Sciences, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoya Kubota
- Clinical Neurophysiology, Department of Clinical Laboratory and Biomedical Sciences, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsutoshi Tazumi
- Laboratory for Pharmacology, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, Shizuoka, Japan
| | - Tomoka Sabuta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masanori P Takahashi
- Clinical Neurophysiology, Department of Clinical Laboratory and Biomedical Sciences, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
15
|
Generation of an induced pluripotent stem cell line from a healthy Caucasian male. Stem Cell Res 2022; 60:102717. [DOI: 10.1016/j.scr.2022.102717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/10/2022] [Accepted: 02/19/2022] [Indexed: 11/22/2022] Open
|
16
|
Yuan M, Guo Y, Xia H, Xu H, Deng H, Yuan L. Novel SCN5A and GPD1L Variants Identified in Two Unrelated Han-Chinese Patients With Clinically Suspected Brugada Syndrome. Front Cardiovasc Med 2021; 8:758903. [PMID: 34957250 PMCID: PMC8692717 DOI: 10.3389/fcvm.2021.758903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 10/29/2021] [Indexed: 12/25/2022] Open
Abstract
Brugada syndrome (BrS) is a complexly genetically patterned, rare, malignant, life-threatening arrhythmia disorder. It is autosomal dominant in most cases and characterized by identifiable electrocardiographic patterns, recurrent syncope, nocturnal agonal respiration, and other symptoms, including sudden cardiac death. Over the last 2 decades, a great number of variants have been identified in more than 36 pathogenic or susceptibility genes associated with BrS. The present study used the combined method of whole exome sequencing and Sanger sequencing to identify pathogenic variants in two unrelated Han-Chinese patients with clinically suspected BrS. Minigene splicing assay was used to evaluate the effects of the splicing variant. A novel heterozygous splicing variant c.2437-2A>C in the sodium voltage-gated channel alpha subunit 5 gene (SCN5A) and a novel heterozygous missense variant c.161A>T [p.(Asp54Val)] in the glycerol-3-phosphate dehydrogenase 1 like gene (GPD1L) were identified in these two patients with BrS-1 and possible BrS-2, respectively. Minigene splicing assay indicated the deletion of 15 and 141 nucleotides in exon 16, resulting in critical amino acid deletions. These findings expand the variant spectrum of SCN5A and GPD1L, which can be beneficial to genetic counseling and prenatal diagnosis.
Collapse
Affiliation(s)
- Meng Yuan
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yi Guo
- Department of Medical Information, School of Life Sciences, Central South University, Changsha, China
| | - Hong Xia
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hongbo Xu
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hao Deng
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China.,Disease Genome Research Center, Central South University, Changsha, China
| | - Lamei Yuan
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China.,Disease Genome Research Center, Central South University, Changsha, China
| |
Collapse
|
17
|
Iop L, Iliceto S, Civieri G, Tona F. Inherited and Acquired Rhythm Disturbances in Sick Sinus Syndrome, Brugada Syndrome, and Atrial Fibrillation: Lessons from Preclinical Modeling. Cells 2021; 10:3175. [PMID: 34831398 PMCID: PMC8623957 DOI: 10.3390/cells10113175] [Citation(s) in RCA: 3] [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: 09/20/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Rhythm disturbances are life-threatening cardiovascular diseases, accounting for many deaths annually worldwide. Abnormal electrical activity might arise in a structurally normal heart in response to specific triggers or as a consequence of cardiac tissue alterations, in both cases with catastrophic consequences on heart global functioning. Preclinical modeling by recapitulating human pathophysiology of rhythm disturbances is fundamental to increase the comprehension of these diseases and propose effective strategies for their prevention, diagnosis, and clinical management. In silico, in vivo, and in vitro models found variable application to dissect many congenital and acquired rhythm disturbances. In the copious list of rhythm disturbances, diseases of the conduction system, as sick sinus syndrome, Brugada syndrome, and atrial fibrillation, have found extensive preclinical modeling. In addition, the electrical remodeling as a result of other cardiovascular diseases has also been investigated in models of hypertrophic cardiomyopathy, cardiac fibrosis, as well as arrhythmias induced by other non-cardiac pathologies, stress, and drug cardiotoxicity. This review aims to offer a critical overview on the effective ability of in silico bioinformatic tools, in vivo animal studies, in vitro models to provide insights on human heart rhythm pathophysiology in case of sick sinus syndrome, Brugada syndrome, and atrial fibrillation and advance their safe and successful translation into the cardiology arena.
Collapse
Affiliation(s)
- Laura Iop
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padua, Via Giustiniani, 2, I-35124 Padua, Italy; (S.I.); (G.C.)
| | | | | | - Francesco Tona
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padua, Via Giustiniani, 2, I-35124 Padua, Italy; (S.I.); (G.C.)
| |
Collapse
|
18
|
Kamga MVK, Reppel M, Hescheler J, Nguemo F. Modeling genetic cardiac channelopathies using induced pluripotent stem cells - Status quo from an electrophysiological perspective. Biochem Pharmacol 2021; 192:114746. [PMID: 34461117 DOI: 10.1016/j.bcp.2021.114746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022]
Abstract
Long QT syndrome (LQTS), Brugada syndrome (BrS), and catecholaminergic polymorphic ventricular tachycardia (CPVT) are genetic diseases of the heart caused by mutations in specific cardiac ion channels and are characterized by paroxysmal arrhythmias, which can deteriorate into ventricular fibrillation. In LQTS3 and BrS different mutations in the SCN5A gene lead to a gain-or a loss-of-function of the voltage-gated sodium channel Nav1.5, respectively. Although sharing the same gene mutation, these syndromes are characterized by different clinical manifestations and functional perturbations and in some cases even present an overlapping clinical phenotype. Several studies have shown that Na+ current abnormalities in LQTS3 and BrS can also cause Ca2+-signaling aberrancies in cardiomyocytes (CMs). Abnormal Ca2+ homeostasis is also the main feature of CPVT which is mostly caused by heterozygous mutations in the RyR2 gene. Large numbers of disease-causing mutations were identified in RyR2 and SCN5A but it is not clear how different variants in the SCN5A gene produce different clinical syndromes and if in CPVT Ca2+ abnormalities and drug sensitivities vary depending on the mutation site in the RyR2. These questions can now be addressed by using patient-specific in vitro models of these diseases based on induced pluripotent stem cells (iPSCs). In this review, we summarize different insights gained from these models with a focus on electrophysiological perturbations caused by different ion channel mutations and discuss how will this knowledge help develop better stratification and more efficient personalized therapies for these patients.
Collapse
Affiliation(s)
- Michelle Vanessa Kapchoup Kamga
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Michael Reppel
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, 50931 Cologne, Germany; Praxis für Kardiologie und Angiologie, Landsberg am Lech, Germany
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Filomain Nguemo
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
19
|
Zhu Y, Wang L, Cui C, Qin H, Chen H, Chen S, Lin Y, Cheng H, Jiang X, Chen M. Pathogenesis and drug response of iPSC-derived cardiomyocytes from two Brugada syndrome patients with different Na v1.5-subunit mutations. J Biomed Res 2021; 35:395-407. [PMID: 34628405 PMCID: PMC8502687 DOI: 10.7555/jbr.35.20210045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Brugada syndrome (BrS) is a complex genetic cardiac ion channel disease that causes a high predisposition to sudden cardiac death. Considering that its heterogeneity in clinical manifestations may result from genetic background, the application of patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) may help to reveal cell phenotype characteristics underlying different genetic variations. Here, to verify and compare the pathogenicity of mutations (SCN5A c.4213G>A andSCN1B c.590C>T) identified from two BrS patients, we generated two novel BrS iPS cell lines that carried missense mutations inSCN5A or SCN1B, compared their structures and electrophysiology, and evaluated the safety of quinidine in patient-specific iPSC-derived CMs. Compared to the control group, BrS-CMs showed a significant reduction in sodium current, prolonged action potential duration, and varying degrees of decreased Vmax, but no structural difference. After applying different concentrations of quinidine, drug-induced cardiotoxicity was not observed within 3-fold unbound effective therapeutic plasma concentration (ETPC). The data presented proved that iPSC-CMs with variants in SCN5A c.4213G>A orSCN1B c.590C>T are able to recapitulate single-cell phenotype features of BrS and respond appropriately to quinidine without increasing incidence of arrhythmic events.
Collapse
Affiliation(s)
- Yue Zhu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Linlin Wang
- Department of Cardiology, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing Chest Hospital, Nanjing, Jiangsu 210029, China
| | - Chang Cui
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Huiyuan Qin
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Hongwu Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Shaojie Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yongping Lin
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Hongyi Cheng
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiaohong Jiang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Minglong Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
20
|
Behr ER, Ben-Haim Y, Ackerman MJ, Krahn AD, Wilde AAM. Brugada syndrome and reduced right ventricular outflow tract conduction reserve: a final common pathway? Eur Heart J 2021; 42:1073-1081. [PMID: 33421051 DOI: 10.1093/eurheartj/ehaa1051] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/04/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
Brugada syndrome (BrS) was first described as a primary electrical disorder predisposing to the risk of sudden cardiac death and characterized by right precordial lead ST elevation. Early description of right ventricular structural abnormalities and of right ventricular outflow tract (RVOT) conduction delay in BrS patients set the stage for the current controversy over the pathophysiology underlying the syndrome: channelopathy or cardiomyopathy; repolarization or depolarization. This review examines the current understanding of the BrS substrate, its genetic and non-genetic basis, theories of pathophysiology, and the clinical implications thereof. We propose that the final common pathway for BrS could be viewed as a disease of 'reduced RVOT conduction reserve'.
Collapse
Affiliation(s)
- Elijah R Behr
- Cardiovascular Clinical Academic Group, Molecular and Clinical Sciences Research Institute, St. George's University of London, Cranmer Terrace, London SW17 0RE, UK.,St. George's University Hospitals NHS Foundation Trust, Cranmer Terrace, London SW17 0RE, UK.,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart (ERN GUARDHEART http://guardheart.ern-net.eu).,European Cardiac Arrhythmia Genetics Focus Group (ECGen), EHRA
| | - Yael Ben-Haim
- Cardiovascular Clinical Academic Group, Molecular and Clinical Sciences Research Institute, St. George's University of London, Cranmer Terrace, London SW17 0RE, UK.,St. George's University Hospitals NHS Foundation Trust, Cranmer Terrace, London SW17 0RE, UK.,European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart (ERN GUARDHEART http://guardheart.ern-net.eu)
| | - Michael J Ackerman
- Division of Heart Rhythm Services and the Windland Smith Rice Genetic Heart Rhythm Clinic, Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Andrew D Krahn
- Heart Rhythm Services, Division of Cardiology, Department of Medicine, University of British Columbia, 2775 Laurel Street, Vancouver, BC V5Z 1M9, Canada
| | - Arthur A M Wilde
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart: ERN GUARD-Heart (ERN GUARDHEART http://guardheart.ern-net.eu).,European Cardiac Arrhythmia Genetics Focus Group (ECGen), EHRA.,Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| |
Collapse
|
21
|
Giannetti F, Benzoni P, Campostrini G, Milanesi R, Bucchi A, Baruscotti M, Dell'Era P, Rossini A, Barbuti A. A detailed characterization of the hyperpolarization-activated "funny" current (I f) in human-induced pluripotent stem cell (iPSC)-derived cardiomyocytes with pacemaker activity. Pflugers Arch 2021; 473:1009-1021. [PMID: 33934225 PMCID: PMC8245366 DOI: 10.1007/s00424-021-02571-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 04/01/2021] [Accepted: 04/19/2021] [Indexed: 01/09/2023]
Abstract
Properties of the funny current (If) have been studied in several animal and cellular models, but so far little is known concerning its properties in human pacemaker cells. This work provides a detailed characterization of If in human-induced pluripotent stem cell (iPSC)–derived pacemaker cardiomyocytes (pCMs), at different time points. Patch-clamp analysis showed that If density did not change during differentiation; however, after day 30, it activates at more negative potential and with slower time constants. These changes are accompanied by a slowing in beating rate. If displayed the voltage-dependent block by caesium and reversed (Erev) at − 22 mV, compatibly with the 3:1 K+/Na+ permeability ratio. Lowering [Na+]o (30 mM) shifted the Erev to − 39 mV without affecting conductance. Increasing [K+]o (30 mM) shifted the Erev to − 15 mV with a fourfold increase in conductance. pCMs express mainly HCN4 and HCN1 together with the accessory subunits CAV3, KCR1, MiRP1, and SAP97 that contribute to the context-dependence of If. Autonomic agonists modulated the diastolic depolarization, and thus rate, of pCMs. The adrenergic agonist isoproterenol induced rate acceleration and a positive shift of If voltage-dependence (EC50 73.4 nM). The muscarinic agonists had opposite effects (Carbachol EC50, 11,6 nM). Carbachol effect was however small but it could be increased by pre-stimulation with isoproterenol, indicating low cAMP levels in pCMs. In conclusion, we demonstrated that pCMs display an If with the physiological properties expected by pacemaker cells and may thus represent a suitable model for studying human If-related sinus arrhythmias.
Collapse
Affiliation(s)
- Federica Giannetti
- The Cell Physiology MiLab, Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
| | - Patrizia Benzoni
- The Cell Physiology MiLab, Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
| | - Giulia Campostrini
- The Cell Physiology MiLab, Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333ZC, Leiden, The Netherlands
| | - Raffaella Milanesi
- The Cell Physiology MiLab, Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Via dell'Università 6, 26900, Lodi, Italy
| | - Annalisa Bucchi
- The Cell Physiology MiLab, Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
| | - Mirko Baruscotti
- The Cell Physiology MiLab, Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
| | - Patrizia Dell'Era
- Cellular Fate Reprogramming Unit, Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Alessandra Rossini
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Viale Druso 1, 39100, Bolzano, Italy
| | - Andrea Barbuti
- The Cell Physiology MiLab, Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy.
| |
Collapse
|
22
|
Micheu MM, Rosca AM. Patient-specific induced pluripotent stem cells as “disease-in-a-dish” models for inherited cardiomyopathies and channelopathies – 15 years of research. World J Stem Cells 2021; 13:281-303. [PMID: 33959219 PMCID: PMC8080539 DOI: 10.4252/wjsc.v13.i4.281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/11/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Among inherited cardiac conditions, a special place is kept by cardiomyopathies (CMPs) and channelopathies (CNPs), which pose a substantial healthcare burden due to the complexity of the therapeutic management and cause early mortality. Like other inherited cardiac conditions, genetic CMPs and CNPs exhibit incomplete penetrance and variable expressivity even within carriers of the same pathogenic deoxyribonucleic acid variant, challenging our understanding of the underlying pathogenic mechanisms. Until recently, the lack of accurate physiological preclinical models hindered the investigation of fundamental cellular and molecular mechanisms. The advent of induced pluripotent stem cell (iPSC) technology, along with advances in gene editing, offered unprecedented opportunities to explore hereditary CMPs and CNPs. Hallmark features of iPSCs include the ability to differentiate into unlimited numbers of cells from any of the three germ layers, genetic identity with the subject from whom they were derived, and ease of gene editing, all of which were used to generate “disease-in-a-dish” models of monogenic cardiac conditions. Functionally, iPSC-derived cardiomyocytes that faithfully recapitulate the patient-specific phenotype, allowed the study of disease mechanisms in an individual-/allele-specific manner, as well as the customization of therapeutic regimen. This review provides a synopsis of the most important iPSC-based models of CMPs and CNPs and the potential use for modeling disease mechanisms, personalized therapy and deoxyribonucleic acid variant functional annotation.
Collapse
Affiliation(s)
- Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Bucharest 014452, Romania
| | - Ana-Maria Rosca
- Cell and Tissue Engineering Laboratory, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest 050568, Romania
| |
Collapse
|
23
|
iPSC-Cardiomyocyte Models of Brugada Syndrome-Achievements, Challenges and Future Perspectives. Int J Mol Sci 2021; 22:ijms22062825. [PMID: 33802229 PMCID: PMC8001521 DOI: 10.3390/ijms22062825] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/19/2022] Open
Abstract
Brugada syndrome (BrS) is an inherited cardiac arrhythmia that predisposes to ventricular fibrillation and sudden cardiac death. It originates from oligogenic alterations that affect cardiac ion channels or their accessory proteins. The main hurdle for the study of the functional effects of those variants is the need for a specific model that mimics the complex environment of human cardiomyocytes. Traditionally, animal models or transient heterologous expression systems are applied for electrophysiological investigations, each of these models having their limitations. The ability to create induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), providing a source of human patient-specific cells, offers new opportunities in the field of cardiac disease modelling. Contemporary iPSC-CMs constitute the best possible in vitro model to study complex cardiac arrhythmia syndromes such as BrS. To date, thirteen reports on iPSC-CM models for BrS have been published and with this review we provide an overview of the current findings, with a focus on the electrophysiological parameters. We also discuss the methods that are used for cell derivation and data acquisition. In the end, we critically evaluate the knowledge gained by the use of these iPSC-CM models and discuss challenges and future perspectives for iPSC-CMs in the study of BrS and other arrhythmias.
Collapse
|
24
|
Deo M, Akwaboah A, Tsevi B, Treat JA, Cordeiro JM. Role of the rapid delayed rectifier K + current in human induced pluripotent stem cells derived cardiomyocytes. ARCHIVES OF STEM CELL AND THERAPY 2021; 1:14-18. [PMID: 33604593 PMCID: PMC7889062 DOI: 10.46439/stemcell.1.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Makarand Deo
- Department of Engineering, Norfolk State University, Norfolk, Virginia, USA
| | - Akwasi Akwaboah
- Department of Engineering, Norfolk State University, Norfolk, Virginia, USA
| | - Bright Tsevi
- Department of Engineering, Norfolk State University, Norfolk, Virginia, USA
| | - Jacqueline A Treat
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, New York, USA
| | - Jonathan M Cordeiro
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, New York, USA
| |
Collapse
|
25
|
Wang Y, Lei W, Yang J, Ni X, Ye L, Shen Z, Hu S. The updated view on induced pluripotent stem cells for cardiovascular precision medicine. Pflugers Arch 2021; 473:1137-1149. [DOI: 10.1007/s00424-021-02530-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/06/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022]
|
26
|
Pérez-Agustín A, Pinsach-Abuin M, Pagans S. Role of Non-Coding Variants in Brugada Syndrome. Int J Mol Sci 2020; 21:E8556. [PMID: 33202810 PMCID: PMC7698069 DOI: 10.3390/ijms21228556] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022] Open
Abstract
Brugada syndrome (BrS) is an inherited electrical heart disease associated with a high risk of sudden cardiac death (SCD). The genetic characterization of BrS has always been challenging. Although several cardiac ion channel genes have been associated with BrS, SCN5A is the only gene that presents definitive evidence for causality to be used for clinical diagnosis of BrS. However, more than 65% of diagnosed cases cannot be explained by variants in SCN5A or other genes. Therefore, in an important number of BrS cases, the underlying mechanisms are still elusive. Common variants, mostly located in non-coding regions, have emerged as potential modulators of the disease by affecting different regulatory mechanisms, including transcription factors (TFs), three-dimensional organization of the genome, or non-coding RNAs (ncRNAs). These common variants have been hypothesized to modulate the interindividual susceptibility of the disease, which could explain incomplete penetrance of BrS observed within families. Altogether, the study of both common and rare variants in parallel is becoming increasingly important to better understand the genetic basis underlying BrS. In this review, we aim to describe the challenges of studying non-coding variants associated with disease, re-examine the studies that have linked non-coding variants with BrS, and provide further evidence for the relevance of regulatory elements in understanding this cardiac disorder.
Collapse
Affiliation(s)
- Adrian Pérez-Agustín
- Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain;
- Biomedical Research Institute of Girona, 17190 Salt, Spain;
| | | | - Sara Pagans
- Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain;
- Biomedical Research Institute of Girona, 17190 Salt, Spain;
| |
Collapse
|
27
|
Campuzano O, Sarquella-Brugada G, Cesar S, Arbelo E, Brugada J, Brugada R. Update on Genetic Basis of Brugada Syndrome: Monogenic, Polygenic or Oligogenic? Int J Mol Sci 2020; 21:ijms21197155. [PMID: 32998306 PMCID: PMC7582739 DOI: 10.3390/ijms21197155] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Brugada syndrome is a rare inherited arrhythmogenic disease leading to ventricular fibrillation and high risk of sudden death. In 1998, this syndrome was linked with a genetic variant with an autosomal dominant pattern of inheritance. To date, rare variants identified in more than 40 genes have been potentially associated with this disease. Variants in regulatory regions, combinations of common variants and other genetic alterations are also proposed as potential origins of Brugada syndrome, suggesting a polygenic or oligogenic inheritance pattern. However, most of these genetic alterations remain of questionable causality; indeed, rare pathogenic variants in the SCN5A gene are the only established cause of Brugada syndrome. Comprehensive analysis of all reported genetic alterations identified the origin of disease in no more than 40% of diagnosed cases. Therefore, identifying the cause of this rare arrhythmogenic disease in the many families without a genetic diagnosis is a major current challenge in Brugada syndrome. Additional challenges are interpretation/classification of variants and translation of genetic data into clinical practice. Further studies focused on unraveling the pathophysiological mechanisms underlying the disease are needed. Here we provide an update on the genetic basis of Brugada syndrome.
Collapse
Affiliation(s)
- Oscar Campuzano
- Cardiovascular Genetics Centre, University of Girona-IDIBGI, 17190 Girona, Spain
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain;
- Centro Investigación Biomédica en Red: Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (E.A.); (J.B.)
- Correspondence: (O.C.); (R.B.)
| | - Georgia Sarquella-Brugada
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain;
- Arrhythmia Unit, Hospital Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain;
| | - Sergi Cesar
- Arrhythmia Unit, Hospital Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain;
| | - Elena Arbelo
- Centro Investigación Biomédica en Red: Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (E.A.); (J.B.)
- Arrhythmia Unit, Hospital Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain;
| | - Josep Brugada
- Centro Investigación Biomédica en Red: Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (E.A.); (J.B.)
- Arrhythmia Unit, Hospital Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain;
- Arrhythmia Section, Cardiovascular Institute, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| | - Ramon Brugada
- Cardiovascular Genetics Centre, University of Girona-IDIBGI, 17190 Girona, Spain
- Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain;
- Centro Investigación Biomédica en Red: Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (E.A.); (J.B.)
- Familial Cardiomyopathies Unit, Hospital Josep Trueta de Girona, 17007 Girona, Spain
- Correspondence: (O.C.); (R.B.)
| |
Collapse
|
28
|
Blok M, Boukens BJ. Mechanisms of Arrhythmias in the Brugada Syndrome. Int J Mol Sci 2020; 21:ijms21197051. [PMID: 32992720 PMCID: PMC7582368 DOI: 10.3390/ijms21197051] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Arrhythmias in Brugada syndrome patients originate in the right ventricular outflow tract (RVOT). Over the past few decades, the characterization of the unique anatomy and electrophysiology of the RVOT has revealed the arrhythmogenic nature of this region. However, the mechanisms that drive arrhythmias in Brugada syndrome patients remain debated as well as the exact site of their occurrence in the RVOT. Identifying the site of origin and mechanism of Brugada syndrome would greatly benefit the development of mechanism-driven treatment strategies.
Collapse
Affiliation(s)
- Michiel Blok
- Department of Medical Biology, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Bastiaan J. Boukens
- Department of Medical Biology, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-(0)20-566-4659
| |
Collapse
|
29
|
Barajas-Martinez H, Smith M, Hu D, Goodrow RJ, Puleo C, Hasdemir C, Antzelevitch C, Pfeiffer R, Treat JA, Cordeiro JM. Susceptibility to Ventricular Arrhythmias Resulting from Mutations in FKBP1B, PXDNL, and SCN9A Evaluated in hiPSC Cardiomyocytes. Stem Cells Int 2020; 2020:8842398. [PMID: 32952569 PMCID: PMC7481990 DOI: 10.1155/2020/8842398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/29/2020] [Accepted: 08/11/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND We report an inherited cardiac arrhythmia syndrome consisting of Brugada and Early Repolarization Syndrome associated with variants in SCN9A, PXDNL, and FKBP1B. The proband inherited the 3 mutations and exhibited palpitations and arrhythmia-mediated syncope, whereas the parents and sister, who carried one or two of the mutations, were asymptomatic. METHODS AND RESULTS We assessed the functional impact of these mutations in induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) derived from the proband and an unaffected family member. Current and voltage clamp recordings, as well as confocal microscopy analysis of Ca2+ transients, were evaluated in hiPSC-CMs from the proband and compared these results with hiPSC-CMs from undiseased controls. Genetic analysis using next-generation DNA sequencing revealed heterozygous mutations in SCN9A, PXDNL, and FKBP1B in the proband. The proband displayed right bundle branch block and exhibited episodes of syncope. The father carried a mutation in FKBP1B, whereas the mother and sister carried the SCN9A mutation. None of the 3 family members screened developed cardiac events. Action potential recordings from control hiPSC-CM showed spontaneous activity and a low upstroke velocity. In contrast, the hiPSC-CM from the proband showed irregular spontaneous activity. Confocal microscopy of the hiPSC-CM of the proband revealed low fluorescence intensity Ca2+ transients that were episodic in nature. Patch-clamp measurements in hiPSC-CM showed no difference in I Na but reduced I Ca in the proband compared with control. Coexpression of PXDNL-R391Q with SCN5A-WT displayed lower I Na density compared to PXDNL-WT. In addition, coexpression of PXDNL-R391Q with KCND3-WT displayed significantly higher I to density compared to PXDNL-WT. CONCLUSION SCN9A, PXDNL, and FKBP1B variants appeared to alter spontaneous activity in hiPSC-CM. Only the proband carrying all 3 mutations displayed the ERS/BrS phenotype, whereas one nor two mutations alone did not produce the clinical phenotype. Our results suggest a polygenic cause of the BrS/ERS arrhythmic phenotype due to mutations in these three gene variants caused a very significant loss of function of I Na and I Ca and gain of function of I to.
Collapse
Affiliation(s)
- Hector Barajas-Martinez
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
- Department of Cardiovascular Research, Lakenau Institute for Medical Research, Wynnewood, PA, USA
| | - Maya Smith
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Dan Hu
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
- Department of Cardiology & Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, China
| | - Robert J. Goodrow
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Colleen Puleo
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Can Hasdemir
- Department of Cardiology, Ege University School of Medicine, Izmir, Turkey
| | - Charles Antzelevitch
- Department of Cardiovascular Research, Lakenau Institute for Medical Research, Wynnewood, PA, USA
- Kimmel College of Medicine of Thomas Jefferson University, Philadelphia, PA, USA
| | - Ryan Pfeiffer
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Jacqueline A. Treat
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| | - Jonathan M. Cordeiro
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, NY, USA
| |
Collapse
|
30
|
Glazer AM, Wada Y, Li B, Muhammad A, Kalash OR, O'Neill MJ, Shields T, Hall L, Short L, Blair MA, Kroncke BM, Capra JA, Roden DM. High-Throughput Reclassification of SCN5A Variants. Am J Hum Genet 2020; 107:111-123. [PMID: 32533946 PMCID: PMC7332654 DOI: 10.1016/j.ajhg.2020.05.015] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 05/19/2020] [Indexed: 12/19/2022] Open
Abstract
Partial or complete loss-of-function variants in SCN5A are the most common genetic cause of the arrhythmia disorder Brugada syndrome (BrS1). However, the pathogenicity of SCN5A variants is often unknown or disputed; 80% of the 1,390 SCN5A missense variants observed in at least one individual to date are variants of uncertain significance (VUSs). The designation of VUS is a barrier to the use of sequence data in clinical care. We selected 83 variants: 10 previously studied control variants, 10 suspected benign variants, and 63 suspected Brugada syndrome-associated variants, selected on the basis of their frequency in the general population and in individuals with Brugada syndrome. We used high-throughput automated patch clamping to study the function of the 83 variants, with the goal of reclassifying variants with functional data. The ten previously studied controls had functional properties concordant with published manual patch clamp data. All 10 suspected benign variants had wild-type-like function. 22 suspected BrS variants had loss of channel function (<10% normalized peak current) and 22 variants had partial loss of function (10%-50% normalized peak current). The previously unstudied variants were initially classified as likely benign (n = 2), likely pathogenic (n = 10), or VUSs (n = 61). After the patch clamp studies, 16 variants were benign/likely benign, 45 were pathogenic/likely pathogenic, and only 12 were still VUSs. Structural modeling identified likely mechanisms for loss of function including altered thermostability and disruptions to alpha helices, disulfide bonds, or the permeation pore. High-throughput patch clamping enabled reclassification of the majority of tested VUSs in SCN5A.
Collapse
Affiliation(s)
- Andrew M Glazer
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Yuko Wada
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Bian Li
- Department of Biological Sciences, Center for Structural Biology, and Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Ayesha Muhammad
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Olivia R Kalash
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Matthew J O'Neill
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Tiffany Shields
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lynn Hall
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Laura Short
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Marcia A Blair
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Brett M Kroncke
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John A Capra
- Department of Biological Sciences, Center for Structural Biology, and Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Dan M Roden
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
31
|
Nakao S, Ihara D, Hasegawa K, Kawamura T. Applications for Induced Pluripotent Stem Cells in Disease Modelling and Drug Development for Heart Diseases. Eur Cardiol 2020; 15:1-10. [PMID: 32180835 PMCID: PMC7066852 DOI: 10.15420/ecr.2019.03] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/09/2019] [Indexed: 12/22/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are derived from reprogrammed somatic cells by the introduction of defined transcription factors. They are characterised by a capacity for self-renewal and pluripotency. Human (h)iPSCs are expected to be used extensively for disease modelling, drug screening and regenerative medicine. Obtaining cardiac tissue from patients with mutations for genetic studies and functional analyses is a highly invasive procedure. In contrast, disease-specific hiPSCs are derived from the somatic cells of patients with specific genetic mutations responsible for disease phenotypes. These disease-specific hiPSCs are a better tool for studies of the pathophysiology and cellular responses to therapeutic agents. This article focuses on the current understanding, limitations and future direction of disease-specific hiPSC-derived cardiomyocytes for further applications.
Collapse
Affiliation(s)
- Shu Nakao
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan.,Global Innovation Research Organization, Ritsumeikan University, Kusatsu, Japan.,Division of Translational Research, Kyoto Medical Center, National Hospital Organization, Kyoto, Japan
| | - Dai Ihara
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan.,Global Innovation Research Organization, Ritsumeikan University, Kusatsu, Japan
| | - Koji Hasegawa
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization, Kyoto, Japan
| | - Teruhisa Kawamura
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan.,Global Innovation Research Organization, Ritsumeikan University, Kusatsu, Japan.,Division of Translational Research, Kyoto Medical Center, National Hospital Organization, Kyoto, Japan
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
de la Roche J, Angsutararux P, Kempf H, Janan M, Bolesani E, Thiemann S, Wojciechowski D, Coffee M, Franke A, Schwanke K, Leffler A, Luanpitpong S, Issaragrisil S, Fischer M, Zweigerdt R. Comparing human iPSC-cardiomyocytes versus HEK293T cells unveils disease-causing effects of Brugada mutation A735V of Na V1.5 sodium channels. Sci Rep 2019; 9:11173. [PMID: 31371804 PMCID: PMC6673693 DOI: 10.1038/s41598-019-47632-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 07/22/2019] [Indexed: 11/23/2022] Open
Abstract
Loss-of-function mutations of the SCN5A gene encoding for the sodium channel α-subunit NaV1.5 result in the autosomal dominant hereditary disease Brugada Syndrome (BrS) with a high risk of sudden cardiac death in the adult. We here engineered human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) carrying the CRISPR/Cas9 introduced BrS-mutation p.A735V-NaV1.5 (g.2204C > T in exon 14 of SCN5A) as a novel model independent of patient´s genetic background. Recent studies raised concern regarding the use of hiPSC-CMs for studying adult-onset hereditary diseases due to cells' immature phenotype. To tackle this concern, long-term cultivation of hiPSC-CMs on a stiff matrix (27-42 days) was applied to promote maturation. Patch clamp recordings of A735V mutated hiPSC-CMs revealed a substantially reduced upstroke velocity and sodium current density, a prominent rightward shift of the steady state activation curve and decelerated recovery from inactivation as compared to isogenic hiPSC-CMs controls. These observations were substantiated by a comparative study on mutant A735V-NaV1.5 channels heterologously expressed in HEK293T cells. In contrast to mutated hiPSC-CMs, a leftward shift of sodium channel inactivation was not observed in HEK293T, emphasizing the importance of investigating mechanisms of BrS in independent systems. Overall, our approach supports hiPSC-CMs' relevance for investigating channelopathies in a dish.
Collapse
Affiliation(s)
- Jeanne de la Roche
- Institute for Neurophysiology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany.
| | - Paweorn Angsutararux
- Siriraj Center of Excellence for Stem Cell Research (SiSCR), Faculty of Medicine, Siriraj Hospital, 2, Bangkoknoi, Bangkok, 10700, Thailand
| | - Henning Kempf
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) and Hans Borst Zentrum (HBZ), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
- Department of Stem Cell Discovery, Novo Nordisk A/S, 2760, Maaloev, Denmark
| | - Montira Janan
- Siriraj Center of Excellence for Stem Cell Research (SiSCR), Faculty of Medicine, Siriraj Hospital, 2, Bangkoknoi, Bangkok, 10700, Thailand
| | - Emiliano Bolesani
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) and Hans Borst Zentrum (HBZ), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Stefan Thiemann
- Institute for Neurophysiology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Daniel Wojciechowski
- Institute for Neurophysiology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Michelle Coffee
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) and Hans Borst Zentrum (HBZ), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Annika Franke
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) and Hans Borst Zentrum (HBZ), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Kristin Schwanke
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) and Hans Borst Zentrum (HBZ), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Andreas Leffler
- Department of Anaesthesiology and Intensive Care, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research (SiSCR), Faculty of Medicine, Siriraj Hospital, 2, Bangkoknoi, Bangkok, 10700, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research (SiSCR), Faculty of Medicine, Siriraj Hospital, 2, Bangkoknoi, Bangkok, 10700, Thailand.
| | - Martin Fischer
- Institute for Neurophysiology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Robert Zweigerdt
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) and Hans Borst Zentrum (HBZ), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany.
| |
Collapse
|
35
|
Cuyàs E, Gumuzio J, Lozano-Sánchez J, Carreras D, Verdura S, Llorach-Parés L, Sanchez-Martinez M, Selga E, Pérez GJ, Scornik FS, Brugada R, Bosch-Barrera J, Segura-Carretero A, Martin ÁG, Encinar JA, Menendez JA. Extra Virgin Olive Oil Contains a Phenolic Inhibitor of the Histone Demethylase LSD1/KDM1A. Nutrients 2019; 11:nu11071656. [PMID: 31331073 PMCID: PMC6683035 DOI: 10.3390/nu11071656] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
The lysine-specific histone demethylase 1A (LSD1) also known as lysine (K)-specific demethylase 1A (KDM1A) is a central epigenetic regulator of metabolic reprogramming in obesity-associated diseases, neurological disorders, and cancer. Here, we evaluated the ability of oleacein, a biophenol secoiridoid naturally present in extra virgin olive oil (EVOO), to target LSD1. Molecular docking and dynamic simulation approaches revealed that oleacein could target the binding site of the LSD1 cofactor flavin adenosine dinucleotide with high affinity and at low concentrations. At higher concentrations, oleacein was predicted to target the interaction of LSD1 with histone H3 and the LSD1 co-repressor (RCOR1/CoREST), likely disturbing the anchorage of LSD1 to chromatin. AlphaScreen-based in vitro assays confirmed the ability of oleacein to act as a direct inhibitor of recombinant LSD1, with an IC50 as low as 2.5 μmol/L. Further, oleacein fully suppressed the expression of the transcription factor SOX2 (SEX determining Region Y-box 2) in cancer stem-like and induced pluripotent stem (iPS) cells, which specifically occurs under the control of an LSD1-targeted distal enhancer. Conversely, oleacein failed to modify ectopic SOX2 overexpression driven by a constitutive promoter. Overall, our findings provide the first evidence that EVOO contains a naturally occurring phenolic inhibitor of LSD1, and support the use of oleacein as a template to design new secoiridoid-based LSD1 inhibitors.
Collapse
Affiliation(s)
- Elisabet Cuyàs
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism & Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
| | | | - Jesús Lozano-Sánchez
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, 18071 Granada, Spain
- Research and Development Functional Food Centre (CIDAF), PTS Granada, 18100 Granada, Spain
| | - David Carreras
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- Cardiovascular Genetics Centre, Department of Medical Sciences, University of Girona, 17071 Girona, Spain
| | - Sara Verdura
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism & Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
| | | | | | - Elisabet Selga
- Cardiovascular Genetics Centre, Department of Medical Sciences, University of Girona, 17071 Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
| | - Guillermo J Pérez
- Cardiovascular Genetics Centre, Department of Medical Sciences, University of Girona, 17071 Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Fabiana S Scornik
- Cardiovascular Genetics Centre, Department of Medical Sciences, University of Girona, 17071 Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Ramon Brugada
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- Cardiovascular Genetics Centre, Department of Medical Sciences, University of Girona, 17071 Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Dr. Josep Trueta Hospital of Girona, 17007 Girona, Spain
| | - Joaquim Bosch-Barrera
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- Medical Oncology, Catalan Institute of Oncology (ICO), 17007 Girona, Spain
- Department of Medical Sciences, Medical School University of Girona, 17071 Girona, Spain
| | - Antonio Segura-Carretero
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, 18071 Granada, Spain
- Research and Development Functional Food Centre (CIDAF), PTS Granada, 18100 Granada, Spain
| | | | - José Antonio Encinar
- Institute of Research, Development and Innovation in Biotechnology of Elche (IDiBE) and Molecular and Cell Biology Institute (IBMC), Miguel Hernández University (UMH), 03202 Elche, Spain
| | - Javier A Menendez
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism & Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain.
| |
Collapse
|
36
|
Abstract
Brugada syndrome is an inherited, rare cardiac arrhythmogenic disease, associated with sudden cardiac death. It accounts for up to 20% of sudden deaths in patients without structural cardiac abnormalities. The majority of mutations involve the cardiac sodium channel gene SCN5A and give rise to classical abnormal electrocardiogram with ST segment elevation in the right precordial leads V1 to V3 and a predisposition to ventricular fibrillation. The pathophysiological mechanisms of Brugada syndrome have been investigated using model systems including transgenic mice, canine heart preparations, and expression systems to study different SCN5A mutations. These models have a number of limitations. The recent development of pluripotent stem cell technology creates an opportunity to study cardiomyocytes derived from patients and healthy individuals. To date, only a few studies have been done using Brugada syndrome patient-specific iPS-CM, which have provided novel insights into the mechanisms and pathophysiology of Brugada syndrome. This review provides an evaluation of the strengths and limitations of each of these model systems and summarizes the key mechanisms that have been identified to date.
Collapse
|
37
|
Liang W, Gasparyan L, AlQarawi W, Davis DR. Disease modeling of cardiac arrhythmias using human induced pluripotent stem cells. Expert Opin Biol Ther 2019; 19:313-333. [PMID: 30682895 DOI: 10.1080/14712598.2019.1575359] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Inherited arrhythmias are an uncommon, but malignant family of cardiac diseases that result from genetic abnormalities in the ion channels and/or structural proteins within cardiomyocytes. Given the inherent differences between species and the limited reproducibility of in vitro heterologous cell models, progress in understanding the mechanisms underlying these malignant diseases has always languished far behind the clinical science and need. The ability to study human induced pluripotent stem cells (iPSCs) derived cardiomyocytes promises to change this paradigm as patient cells have the potential to become testing platforms for disease phenotyping or therapeutic discovery. AREAS COVERED This review will outline methods developed to genetically reprogram adult cells into iPSCs, differentiate iPSCs into ex vivo models of adult cardiac tissue and iPSCs-based progress in exploring the mechanisms underlying pro-arrhythmic disease phenotypes. EXPERT OPINION Despite being discovered less than 15 years ago, several studies have successfully leveraged iPSCs-derived cardiomyocytes to study malignant arrhythmogenic diseases. These models promise to increase our understanding of the pathophysiology underlying these complex diseases and may identify personalized approaches to treatment.
Collapse
Affiliation(s)
- Wenbin Liang
- a Division of Cardiology, Department of Medicine , University of Ottawa Heart Institute , Ottawa , Canada.,b Department of Cellular and Molecular Medicine , University of Ottawa , Ottawa , Canada
| | - Lilit Gasparyan
- a Division of Cardiology, Department of Medicine , University of Ottawa Heart Institute , Ottawa , Canada
| | - Wael AlQarawi
- a Division of Cardiology, Department of Medicine , University of Ottawa Heart Institute , Ottawa , Canada
| | - Darryl R Davis
- a Division of Cardiology, Department of Medicine , University of Ottawa Heart Institute , Ottawa , Canada.,b Department of Cellular and Molecular Medicine , University of Ottawa , Ottawa , Canada
| |
Collapse
|
38
|
Monasky MM, Pappone C, Piccoli M, Ghiroldi A, Micaglio E, Anastasia L. Calcium in Brugada Syndrome: Questions for Future Research. Front Physiol 2018; 9:1088. [PMID: 30147658 PMCID: PMC6095984 DOI: 10.3389/fphys.2018.01088] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/23/2018] [Indexed: 12/11/2022] Open
Abstract
The Brugada syndrome (BrS) is characterized by coved-type ST-segment elevation in the right precordial leads on the electrocardiogram (ECG) and increased risk of sudden cardiac death (SCD). While it is an inheritable disease, determining the true prevalence is a challenge, since patients may report no known family history of the syndrome, present with a normal spontaneous ECG pattern at the time of examination, and test negative for all known BrS-causative genes. In fact, SCD is often the first indication that a person is affected by the syndrome. Men are more likely to be symptomatic than women. Abnormal, low-voltage, fractionated electrograms have been found in the epicardium of the right ventricular outflow tract (RVOT). Ablation of this area abolishes the abnormal electrograms and helps to prevent arrhythmic recurrences. BrS patients are more likely to experience ventricular tachycardia/fibrillation (VT/VF) during fever or during an increase in vagal tone. Isoproterenol helps to reverse the ECG BrS phenotype. In this review, we discuss roles of calcium in various conditions that are relevant to BrS, such as changes in temperature, heart rate, and vagal tone, and the effects of gender and isoproterenol on calcium handling. Studies are warranted to further investigate these mechanisms in models of BrS.
Collapse
Affiliation(s)
| | - Carlo Pappone
- Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy
| | - Marco Piccoli
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, Milan, Italy
| | - Andrea Ghiroldi
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, Milan, Italy
| | - Emanuele Micaglio
- Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy
| | - Luigi Anastasia
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, Milan, Italy.,Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|
39
|
Bruyneel AA, McKeithan WL, Feyen DA, Mercola M. Will iPSC-cardiomyocytes revolutionize the discovery of drugs for heart disease? Curr Opin Pharmacol 2018; 42:55-61. [PMID: 30081259 DOI: 10.1016/j.coph.2018.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/01/2018] [Indexed: 12/30/2022]
Abstract
Cardiovascular disease remains the largest single cause of mortality in the Western world, despite significant advances in clinical management over the years. Unfortunately, the development of new cardiovascular medicines is stagnating and can in part be attributed to the difficulty of screening for novel therapeutic strategies due to a lack of suitable models. The advent of human induced pluripotent stem cells and the ability to make limitless numbers of cardiomyocytes could revolutionize heart disease modeling and drug discovery. This review summarizes the state of the art in the field, describes the strengths and weaknesses of the technology, and applications where the model system would be most appropriate.
Collapse
Affiliation(s)
- Arne An Bruyneel
- The Cardiovascular Institute and Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Wesley L McKeithan
- The Cardiovascular Institute and Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Dries Am Feyen
- The Cardiovascular Institute and Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark Mercola
- The Cardiovascular Institute and Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
40
|
Han D, Tan H, Sun C, Li G. Dysfunctional Nav1.5 channels due to SCN5A mutations. Exp Biol Med (Maywood) 2018; 243:852-863. [PMID: 29806494 DOI: 10.1177/1535370218777972] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The voltage-gated sodium channel 1.5 (Nav1.5), encoded by the SCN5A gene, is responsible for the rising phase of the action potential of cardiomyocytes. The sodium current mediated by Nav1.5 consists of peak and late components (INa-P and INa-L). Mutant Nav1.5 causes alterations in the peak and late sodium current and is associated with an increasingly wide range of congenital arrhythmias. More than 400 mutations have been identified in the SCN5A gene. Although the mechanisms of SCN5A mutations leading to a variety of arrhythmias can be classified according to the alteration of INa-P and INa-L as gain-of-function, loss-of-function and both, few researchers have summarized the mechanisms in this way before. In this review article, we aim to review the mechanisms underlying dysfunctional Nav1.5 due to SCN5A mutations and to provide some new insights into further approaches in the treatment of arrhythmias. Impact statement The field of ion channelopathy caused by dysfunctional Nav1.5 due to SCN5A mutations is rapidly evolving as novel technologies of electrophysiology are introduced and our understanding of the mechanisms of various arrhythmias develops. In this review, we focus on the dysfunctional Nav1.5 related to arrhythmias and the underlying mechanisms. We update SCN5A mutations in a precise way since 2013 and presents novel classifications of SCN5A mutations responsible for the dysfunction of the peak (INa-P) and late (INa-L) sodium channels based on their phenotypes, including loss-, gain-, and coexistence of gain- and loss-of function mutations in INa-P, INa-L, respectively. We hope this review will provide a new comprehensive way to better understand the electrophysiological mechanisms underlying arrhythmias from cell to bedside, promoting the management of various arrhythmias in practice.
Collapse
Affiliation(s)
- Dan Han
- 1 Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Hui Tan
- 2 Department of Respiratory Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Chaofeng Sun
- 1 Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Guoliang Li
- 1 Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| |
Collapse
|