Human iPSC-Derived Cardiomyocytes for Investigation of Disease Mechanisms and Therapeutic Strategies in Inherited Arrhythmia Syndromes: Strengths and Limitations

During the last two decades, significant progress has been made in the identification of genetic defects underlying inherited arrhythmia syndromes, which has provided some clinical benefit through elucidation of gene-specific arrhythmia triggers and treatment. However, for most arrhythmia syndromes, clinical management is hindered by insufficient knowledge of the functional consequences of the mutation in question, the pro-arrhythmic mechanisms involved, and hence the most optimal treatment strategy. Moreover, disease expressivity and sensitivity to therapeutic interventions often varies between mutations and/or patients, underlining the need for more individualized strategies. The development of the induced pluripotent stem cell (iPSC) technology now provides the opportunity for generating iPSC-derived cardiomyocytes (CMs) from human material (hiPSC-CMs), enabling patient- and/or mutation-specific investigations. These hiPSC-CMs may furthermore be employed for identification and assessment of novel therapeutic strategies for arrhythmia syndromes. However, due to their relative immaturity, hiPSC-CMs also display a number of essential differences as compared to adult human CMs, and hence there are certain limitations in their use. We here review the electrophysiological characteristics of hiPSC-CMs, their use for investigating inherited arrhythmia syndromes, and their applicability for identification and assessment of (novel) anti-arrhythmic treatment strategies.

[1]  A. McCulloch,et al.  Lentiviral Vectors and Protocols for Creation of Stable hESC Lines for Fluorescent Tracking and Drug Resistance Selection of Cardiomyocytes , 2009, PloS one.

[2]  J. Jalife,et al.  Ion channel macromolecular complexes in cardiomyocytes: roles in sudden cardiac death. , 2015, Circulation research.

[3]  Aarti S. Dalal,et al.  A human pluripotent stem cell model of catecholaminergic polymorphic ventricular tachycardia recapitulates patient-specific drug responses , 2016, Disease Models & Mechanisms.

[4]  Michael Glikson,et al.  Modeling of catecholaminergic polymorphic ventricular tachycardia with patient-specific human-induced pluripotent stem cells. , 2012, Journal of the American College of Cardiology.

[5]  P. Benzoni,et al.  Human derived cardiomyocytes: A decade of knowledge after the discovery of induced pluripotent stem cells , 2016, Developmental dynamics : an official publication of the American Association of Anatomists.

[6]  E. Schulze-Bahr,et al.  Human iPS cell model of type 3 long QT syndrome recapitulates drug-based phenotype correction , 2016, Basic Research in Cardiology.

[7]  Lior Gepstein,et al.  Monitoring Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes with Genetically Encoded Calcium and Voltage Fluorescent Reporters , 2015, Stem cell reports.

[8]  U. Ravens,et al.  Transient outward current in human ventricular myocytes of subepicardial and subendocardial origin. , 1994, Circulation research.

[9]  K. Morgan,et al.  Allele-specific RNA interference rescues the long-QT syndrome phenotype in human-induced pluripotency stem cell cardiomyocytes , 2013, European heart journal.

[10]  Ki-Suk Kim,et al.  Evaluation of nefazodone-induced cardiotoxicity in human induced pluripotent stem cell-derived cardiomyocytes. , 2016, Toxicology and applied pharmacology.

[11]  Divya Rajamohan,et al.  Drug evaluation in cardiomyocytes derived from human induced pluripotent stem cells carrying a long QT syndrome type 2 mutation , 2011, European Heart Journal.

[12]  A. Mehta,et al.  Re-trafficking of hERG reverses long QT syndrome 2 phenotype in human iPS-derived cardiomyocytes. , 2014, Cardiovascular research.

[13]  A. Wilde,et al.  Late Sodium Current Inhibition in Acquired and Inherited Ventricular (dys)function and Arrhythmias , 2013, Cardiovascular Drugs and Therapy.

[14]  A Varró,et al.  The slow component of the delayed rectifier potassium current in undiseased human ventricular myocytes. , 2001, Cardiovascular research.

[15]  Qinlian Zhou,et al.  Electronic "expression" of the inward rectifier in cardiocytes derived from human-induced pluripotent stem cells. , 2013, Heart rhythm.

[16]  Azra Fatima,et al.  The Disease-Specific Phenotype in Cardiomyocytes Derived from Induced Pluripotent Stem Cells of Two Long QT Syndrome Type 3 Patients , 2013, PloS one.

[17]  Ronald Wilders,et al.  Dynamic clamp: a powerful tool in cardiac electrophysiology , 2006, The Journal of physiology.

[18]  S. Priori,et al.  Adeno-associated virus-mediated CASQ2 delivery rescues phenotypic alterations in a patient-specific model of recessive catecholaminergic polymorphic ventricular tachycardia , 2016, Cell Death and Disease.

[19]  Shinsuke Yuasa,et al.  Disease characterization using LQTS-specific induced pluripotent stem cells. , 2012, Cardiovascular research.

[20]  James A Thomson,et al.  High purity human-induced pluripotent stem cell-derived cardiomyocytes: electrophysiological properties of action potentials and ionic currents. , 2011, American journal of physiology. Heart and circulatory physiology.

[21]  遠山 周吾 Distinct metabolic flow enables large-scale purification of mouse and human pluripotent stem cell-derived cardiomyocytes , 2013 .

[22]  Jean A Boutin,et al.  High-throughput drug profiling with voltage- and calcium-sensitive fluorescent probes in human iPSC-derived cardiomyocytes. , 2016, American journal of physiology. Heart and circulatory physiology.

[23]  L. Maier,et al.  Novel aspects of excitation–contraction coupling in heart failure , 2013, Basic Research in Cardiology.

[24]  Charles C Hong,et al.  Comparable calcium handling of human iPSC-derived cardiomyocytes generated by multiple laboratories. , 2015, Journal of molecular and cellular cardiology.

[25]  Michael Xavier Doss,et al.  Identification and characterization of a transient outward K+ current in human induced pluripotent stem cell-derived cardiomyocytes. , 2013, Journal of molecular and cellular cardiology.

[26]  Michael Xavier Doss,et al.  Maximum Diastolic Potential of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes Depends Critically on IKr , 2012, PloS one.

[27]  C. Siu,et al.  Overexpression of myocardin induces partial transdifferentiation of human‐induced pluripotent stem cell‐derived mesenchymal stem cells into cardiomyocytes , 2014, Physiological reports.

[28]  R. Passier,et al.  Interpretation of field potentials measured on a multi electrode array in pharmacological toxicity screening on primary and human pluripotent stem cell-derived cardiomyocytes , 2017, Biochemical and biophysical research communications.

[29]  D. Bers,et al.  Cardiac myocytes Ca2+ and Na+ regulation in normal and failing hearts. , 2006, Journal of pharmacological sciences.

[30]  Ard Teisman,et al.  Blockade of the I(Ks) potassium channel: an overlooked cardiovascular liability in drug safety screening? , 2009, Journal of pharmacological and toxicological methods.

[31]  D. Bers,et al.  Na⁺ transport in the normal and failing heart - remember the balance. , 2013, Journal of molecular and cellular cardiology.

[32]  C. Bezzina,et al.  Anti-arrhythmic potential of the late sodium current inhibitor GS-458967 in murine Scn5a-1798insD+/− and human SCN5A-1795insD+/− iPSC-derived cardiomyocytes , 2017, Cardiovascular research.

[33]  Katriina Aalto-Setälä,et al.  Model for long QT syndrome type 2 using human iPS cells demonstrates arrhythmogenic characteristics in cell culture , 2011, Disease Models & Mechanisms.

[34]  P. Lyu,et al.  Trypsin-induced proteome alteration during cell subculture in mammalian cells , 2010, Journal of Biomedical Science.

[35]  J. Magyar,et al.  Effects of endothelin-1 on calcium and potassium currents in undiseased human ventricular myocytes , 2000, Pflügers Archiv.

[36]  Karl-Ludwig Laugwitz,et al.  Patient-specific induced pluripotent stem-cell models for long-QT syndrome. , 2010, The New England journal of medicine.

[37]  Milena Bellin,et al.  Recessive cardiac phenotypes in induced pluripotent stem cell models of Jervell and Lange-Nielsen syndrome: Disease mechanisms and pharmacological rescue , 2014, Proceedings of the National Academy of Sciences.

[38]  Jürgen Hescheler,et al.  Human Pluripotent Stem Cell-Derived Cardiomyocytes: Response to TTX and Lidocain Reveals Strong Cell to Cell Variability , 2012, PloS one.

[39]  Balázs Horváth,et al.  Contribution of IKr and IK1 to ventricular repolarization in canine and human myocytes: is there any influence of action potential duration? , 2008, Basic Research in Cardiology.

[40]  Ruben Coronel,et al.  Pacemaker current (I(f)) in the human sinoatrial node. , 2007, European heart journal.

[41]  Xuetao Sun,et al.  Biowire platform for maturation of human pluripotent stem cell-derived cardiomyocytes. , 2016, Methods.

[42]  Simona Casini,et al.  Isogenic human pluripotent stem cell pairs reveal the role of a KCNH2 mutation in long-QT syndrome , 2013, The EMBO journal.

[43]  Praveen Shukla,et al.  Chemically defined generation of human cardiomyocytes , 2014, Nature Methods.

[44]  Gordon Keller,et al.  Production of de novo cardiomyocytes: human pluripotent stem cell differentiation and direct reprogramming. , 2012, Cell stem cell.

[45]  Kirsi Penttinen,et al.  Antiarrhythmic Effects of Dantrolene in Patients with Catecholaminergic Polymorphic Ventricular Tachycardia and Replication of the Responses Using iPSC Models , 2015, PloS one.

[46]  Jonathan A. Bernstein,et al.  Using iPS cells to investigate cardiac phenotypes in patients with Timothy Syndrome , 2011, Nature.

[47]  Robert Passier,et al.  Atrial-like cardiomyocytes from human pluripotent stem cells are a robust preclinical model for assessing atrial-selective pharmacology , 2015, EMBO molecular medicine.

[48]  M. Morad,et al.  Ca2+ signaling in human induced pluripotent stem cell-derived cardiomyocytes (iPS-CM) from normal and catecholaminergic polymorphic ventricular tachycardia (CPVT)-afflicted subjects. , 2013, Cell calcium.

[49]  N. Hellen,et al.  The Fallacy of Assigning Chamber Specificity to iPSC Cardiac Myocytes from Action Potential Morphology. , 2016, Biophysical journal.

[50]  C. January,et al.  IK1-enhanced human-induced pluripotent stem cell-derived cardiomyocytes: an improved cardiomyocyte model to investigate inherited arrhythmia syndromes. , 2016, American journal of physiology. Heart and circulatory physiology.

[51]  J. Nerbonne,et al.  Molecular physiology of cardiac repolarization. , 2005, Physiological reviews.

[52]  A. Leenhardt,et al.  Catecholaminergic Polymorphic Ventricular Tachycardia , 2012, Circulation. Arrhythmia and electrophysiology.

[53]  Kevin E. Healy,et al.  Calcium Transients Closely Reflect Prolonged Action Potentials in iPSC Models of Inherited Cardiac Arrhythmia , 2014, Stem cell reports.

[54]  Huei-Sheng Vincent Chen,et al.  Non-cardiomyocytes influence the electrophysiological maturation of human embryonic stem cell-derived cardiomyocytes during differentiation. , 2010, Stem cells and development.

[55]  Wendy K. Chung,et al.  Dual Optical Recordings for Action Potentials and Calcium Handling in Induced Pluripotent Stem Cell Models of Cardiac Arrhythmias Using Genetically Encoded Fluorescent Indicators , 2015, Stem cells translational medicine.

[56]  Brian J. Stevenson,et al.  TECRL, a new life‐threatening inherited arrhythmia gene associated with overlapping clinical features of both LQTS and CPVT , 2016, EMBO molecular medicine.

[57]  A. V. van Ginneken,et al.  Ion channelopathies in human induced pluripotent stem cell derived cardiomyocytes: a dynamic clamp study with virtual IK1 , 2015, Front. Physiol..

[58]  Antonis A Armoundas,et al.  Phenotypic differences in transient outward K+ current of human and canine ventricular myocytes: insights into molecular composition of ventricular Ito. , 2004, American journal of physiology. Heart and circulatory physiology.

[59]  Teng Hong Tan,et al.  Modeling type 3 long QT syndrome with cardiomyocytes derived from patient-specific induced pluripotent stem cells. , 2013, International journal of cardiology.

[60]  B. Attali,et al.  SK4 K+ channels are therapeutic targets for the treatment of cardiac arrhythmias , 2017, EMBO molecular medicine.

[61]  T. Ichisaka,et al.  Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors , 2007, Cell.

[62]  Deok‐Ho Kim,et al.  Human iPSC-derived cardiomyocytes and tissue engineering strategies for disease modeling and drug screening. , 2017, Biotechnology advances.

[63]  Gordon Keller,et al.  Biophysical properties of slow potassium channels in human embryonic stem cell derived cardiomyocytes implicate subunit stoichiometry , 2011, The Journal of physiology.

[64]  G. Bett,et al.  Action Potential Shape Is a Crucial Measure of Cell Type of Stem Cell-Derived Cardiocytes. , 2016, Biophysical journal.

[65]  Simona Casini,et al.  Immaturity of human stem-cell-derived cardiomyocytes in culture: fatal flaw or soluble problem? , 2015, Stem cells and development.

[66]  G. Steinbeck,et al.  Regional differences in current density and rate-dependent properties of the transient outward current in subepicardial and subendocardial myocytes of human left ventricle. , 1996, Circulation.

[67]  Laura Iop,et al.  Dantrolene rescues arrhythmogenic RYR2 defect in a patient-specific stem cell model of catecholaminergic polymorphic ventricular tachycardia , 2012, EMBO molecular medicine.

[68]  S. Priori,et al.  Genetics of sudden cardiac death. , 2015, Circulation research.

[69]  Adriaan P IJzerman,et al.  A new hERG allosteric modulator rescues genetic and drug‐induced long‐QT syndrome phenotypes in cardiomyocytes from isogenic pairs of patient induced pluripotent stem cells , 2016, EMBO molecular medicine.

[70]  Deborah K. Lieu,et al.  Mechanism-Based Facilitated Maturation of Human Pluripotent Stem Cell–Derived Cardiomyocytes , 2013, Circulation. Arrhythmia and electrophysiology.

[71]  Kevin E Healy,et al.  In vitro cardiac tissue models: Current status and future prospects. , 2016, Advanced drug delivery reviews.

[72]  Marc A. Vos,et al.  Probing the Contribution of IKs to Canine Ventricular Repolarization: Key Role for &bgr;-Adrenergic Receptor Stimulation , 2003, Circulation.

[73]  Niels Fertig,et al.  HTS techniques for patch clamp-based ion channel screening – advances and economy , 2012, Expert opinion on drug discovery.

[74]  C. Mummery,et al.  Induced pluripotent stem cell derived cardiomyocytes as models for cardiac arrhythmias , 2012, Front. Physio..

[75]  Wataru Shimizu,et al.  HRS/EHRA/APHRS Expert Consensus Statement on the Diagnosis and Management of Patients with Inherited Primary Arrhythmia Syndromes , 2013 .

[76]  U Ravens,et al.  L-type calcium currents of human myocytes from ventricle of non-failing and failing hearts and from atrium. , 1994, Journal of molecular and cellular cardiology.

[77]  C. Mummery,et al.  Cardiomyocytes Derived From Pluripotent Stem Cells Recapitulate Electrophysiological Characteristics of an Overlap Syndrome of Cardiac Sodium Channel Disease , 2012, Circulation.

[78]  C. Valdivia,et al.  Increased late sodium current in myocytes from a canine heart failure model and from failing human heart. , 2005, Journal of molecular and cellular cardiology.

[79]  L. Schild,et al.  Molecular characterization of two founder mutations causing long QT syndrome and identification of compound heterozygous patients , 2006, Annals of medicine.

[80]  Lior Gepstein,et al.  Modelling the long QT syndrome with induced pluripotent stem cells , 2011, Nature.

[81]  Li Zhiyuan,et al.  A Comparison of the Performance and Application Differences Between Manual and Automated Patch-Clamp Techniques , 2012, Current chemical genomics.

[82]  T. Ichisaka,et al.  Induction of Pluripotent Stem Cells From Adult Human Fibroblasts by Defined Factors , 2008 .

[83]  Kevin D. Costa,et al.  Advancing functional engineered cardiac tissues toward a preclinical model of human myocardium , 2014, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[84]  A. Baartscheer,et al.  Sodium ion transporters as new therapeutic targets in heart failure. , 2008, Cardiovascular & hematological agents in medicinal chemistry.

[85]  Samira M. Azarin,et al.  Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling , 2012, Proceedings of the National Academy of Sciences.

[86]  Peter Kohl,et al.  Simultaneous Voltage and Calcium Mapping of Genetically Purified Human Induced Pluripotent Stem Cell–Derived Cardiac Myocyte Monolayers , 2012, Circulation research.

[87]  Divya Rajamohan,et al.  Evaluating the utility of cardiomyocytes from human pluripotent stem cells for drug screening. , 2010, Biochemical Society transactions.

[88]  C. Mummery,et al.  Readthrough-Promoting Drugs Gentamicin and PTC124 Fail to Rescue Nav1.5 Function of Human-Induced Pluripotent Stem Cell–Derived Cardiomyocytes Carrying Nonsense Mutations in the Sodium Channel Gene SCN5A , 2016, Circulation. Arrhythmia and electrophysiology.

[89]  Stuart A Cook,et al.  Characterization of a novel KCNQ1 mutation for type 1 long QT syndrome and assessment of the therapeutic potential of a novel IKs activator using patient-specific induced pluripotent stem cell-derived cardiomyocytes , 2015, Stem Cell Research & Therapy.

[90]  José Jalife,et al.  The inward rectifier current (IK1) controls cardiac excitability and is involved in arrhythmogenesis. , 2005, Heart rhythm.

[91]  Denis Noble,et al.  Rigorous Phenotyping of Cardiac iPSC Preparations Requires Knowledge of Their Resting Potential(s). , 2016, Biophysical journal.

[92]  A. Wilde,et al.  Implantable cardioverter-defibrillator harm in young patients with inherited arrhythmia syndromes: A systematic review and meta-analysis of inappropriate shocks and complications. , 2016, Heart rhythm.

[93]  C. Remme Cardiac sodium channelopathy associated with SCN5A mutations: electrophysiological, molecular and genetic aspects , 2013, The Journal of physiology.

[94]  S. Yamanaka,et al.  Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors , 2006, Cell.

[95]  Robert Passier,et al.  Prediction of drug-induced cardiotoxicity using human embryonic stem cell-derived cardiomyocytes. , 2010, Stem cell research.

[96]  László Virág,et al.  Restricting Excessive Cardiac Action Potential and QT Prolongation: A Vital Role for IKs in Human Ventricular Muscle , 2005, Circulation.

[97]  M Miragoli,et al.  CaMKII inhibition rectifies arrhythmic phenotype in a patient-specific model of catecholaminergic polymorphic ventricular tachycardia , 2013, Cell Death and Disease.

[98]  Xu Xiaoping,et al.  Human-induced pluripotent stem cell-derived cardiomyocytes exhibit temporal changes in phenotype. , 2013, American journal of physiology. Heart and circulatory physiology.

[99]  M. Morad,et al.  Regionally diverse mitochondrial calcium signaling regulates spontaneous pacing in developing cardiomyocytes. , 2015, Cell calcium.

[100]  H. Wichmann,et al.  Sodium channel β1 subunit mutations associated with Brugada syndrome and cardiac conduction disease in humans. , 2008, The Journal of clinical investigation.

[101]  Xuebin B. Yang,et al.  Automated Electrophysiological and Pharmacological Evaluation of Human Pluripotent Stem Cell-Derived Cardiomyocytes , 2016, Stem cells and development.

[102]  Arie O. Verkerk,et al.  hiPSC-derived cardiomyocytes from Brugada Syndrome patients without identified mutations do not exhibit clear cellular electrophysiological abnormalities , 2016, Scientific Reports.

[103]  Jackie Schiller,et al.  Calcium Handling in Human Induced Pluripotent Stem Cell Derived Cardiomyocytes , 2011, PloS one.

[104]  Sean P. Palecek,et al.  Functional Cardiomyocytes Derived From Human Induced Pluripotent Stem Cells , 2009, Circulation research.

[105]  Gordon Keller,et al.  Induced pluripotent stem cells used to reveal drug actions in a long QT syndrome family with complex genetics , 2013, The Journal of general physiology.

[106]  Lei Yang,et al.  Mechanism of automaticity in cardiomyocytes derived from human induced pluripotent stem cells. , 2015, Journal of molecular and cellular cardiology.

[107]  R. Wilders,et al.  Atrio-Sinus Interaction Demonstrated by Blockade of the Rapid Delayed Rectifier Current , 2002, Circulation.

[108]  C. Jayle,et al.  The Heart Rate‐Lowering Agent Ivabradine Inhibits the Pacemaker Current If in Human Atrial Myocytes , 2007, Journal of cardiovascular electrophysiology.

[109]  Lauri Toivonen,et al.  The Jervell and Lange-Nielsen Syndrome: Natural History, Molecular Basis, and Clinical Outcome , 2006, Archives des maladies du coeur et des vaisseaux.

[110]  Michael George,et al.  Planar patch clamp: advances in electrophysiology. , 2008, Methods in molecular biology.

[111]  A Varró,et al.  Delayed rectifier potassium current in undiseased human ventricular myocytes. , 1998, Cardiovascular research.

[112]  Divya Rajamohan,et al.  Cardiomyocytes from human pluripotent stem cells: From laboratory curiosity to industrial biomedical platform☆ , 2016, Biochimica et biophysica acta.

[113]  D. Roden,et al.  Striking In Vivo Phenotype of a Disease-Associated Human SCN5A Mutation Producing Minimal Changes in Vitro , 2011, Circulation.

[114]  Donald M Bers,et al.  Drug Screening Using a Library of Human Induced Pluripotent Stem Cell–Derived Cardiomyocytes Reveals Disease-Specific Patterns of Cardiotoxicity , 2013, Circulation.

[115]  C. Bezzina,et al.  Sodium channel (dys)function and cardiac arrhythmias. , 2010, Cardiovascular therapeutics.

[116]  Kyoichi Ono,et al.  Cardiac T-type Ca(2+) channels in the heart. , 2010, Journal of molecular and cellular cardiology.

[117]  D. Beuckelmann,et al.  Hyperpolarization-activated inward current in ventricular myocytes from normal and failing human hearts. , 1998, Circulation.

[118]  Ronald A. Li,et al.  Modeling susceptibility to drug-induced long QT with a panel of subject-specific induced pluripotent stem cells , 2017, eLife.

[119]  S. Nattel,et al.  Molecular basis of funny current (If) in normal and failing human heart. , 2008, Journal of molecular and cellular cardiology.

[120]  Ofer Binah,et al.  Cardiomyocytes generated from CPVTD307H patients are arrhythmogenic in response to β-adrenergic stimulation , 2012, Journal of cellular and molecular medicine.

[121]  B. Kornreich,et al.  The patch clamp technique: principles and technical considerations. , 2007, Journal of veterinary cardiology : the official journal of the European Society of Veterinary Cardiology.

[122]  S. Yamanaka,et al.  Patient-Specific Human Induced Pluripotent Stem Cell Model Assessed with Electrical Pacing Validates S107 as a Potential Therapeutic Agent for Catecholaminergic Polymorphic Ventricular Tachycardia , 2016, PloS one.

[123]  Udi Nussinovitch,et al.  Sinoatrial node cardiomyocytes derived from human pluripotent cells function as a biological pacemaker , 2016, Nature Biotechnology.