Review Article

Applications for Induced Pluripotent Stem Cells in Disease Modelling and Drug Development for Heart Diseases

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Information image
Average (ratings)
No ratings
Your rating

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.

Disclosure:This work was supported by grants from JSPS KAKENHI (to TK and SN) and an Inamori Foundation Research Grant for Natural Sciences (to SN). TK is the group leader of the research project supported by Ritsumeikan Global Innovation Research Organization (R-GIRO). KH is on the European Cardiology Review editorial board; this did not influence peer review. DI has no conflicts of interest to declare.

Received:

Accepted:

Published online:

Acknowledgements:The authors thank Steven H DeVries (Ophthalmology & Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, US) for helpful suggestions during writing of the manuscript. The authors also thank all members of the Laboratory of Stem Cell and Regenerative Medicine for helpful discussions.

Correspondence Details:Teruhisa Kawamura, 1–1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan. E: kawater@fc.ritsumei.ac.jp

Open Access:

This work is open access under the CC-BY-NC 4.0 License which allows users to copy, redistribute and make derivative works for non-commercial purposes, provided the original work is cited correctly.

Induced Pluripotent Stem Cells and Their Potential Applications

Induced pluripotent stem cells (iPSCs) are generated from somatic cells, such as skin fibroblasts, by ectopic expression of defined reprogramming factors. Within a few years of the first report of the generation of mouse iPSCs, several laboratories reportedly reproduced these cells using other cell types and species using similar approaches.1–4 This early attention on reproducible methods for the production of iPSCs from mammalian cells accelerated research into iPSC technology for clinical applications. iPSCs show unlimited proliferation capacity and pluripotency, as observed in embryonic stem cells (ESCs), and thus have significant advantages as a cell source for producing sufficient numbers of any cell type. In contrast with ESCs, human (h) iPSCs can be established from differentiated cells without destroying human embryos, thereby overcoming related ethical issues. Thus, iPSCs have been extensively investigated worldwide for applications in disease modelling, drug screening and regenerative medicine (Figure 1).2,5

When hiPSCs are derived from patients with a genetic disease caused by a mutation, such patient-derived iPSCs are called disease-specific hiPSCs. As disease-specific hiPSCs contain the same genetic information as the patient, including mutations corresponding to the altered gene function,6,7 disease-specific hiPSCs could potentially be a powerful tool for modelling human disease. Particularly in cardiovascular research, obtaining a sufficient number of cardiomyocytes (CMs) from patients is challenging due to the highly invasive procedures required to extract them. Further, the low proliferation capacity of CMs limits researchers’ ability to maintain these cells in culture. Being able to generate iPSC-derived CMs (hiPSC-CMs) from a specific patient overcomes this problem, and enables identification of typical cellular responses to pathological stress and therapeutic agents because these cells potentially reflect the biological responses of an individual patient’s own CMs (Figure 1).

Recent genetic research has led to the identification of gene mutations responsible for hereditary heart diseases. Investigations into the pathophysiology of those inherited diseases often use animal models that partially mirror the disease conditions. However, animal studies are low throughput, time consuming and relatively expensive. Moreover, there are interspecies differences between humans and the experimental animals in terms of molecular and physiological properties (e.g. ion channel expression profile, heart rate), as well as in the cellular responses to pathological stress. Therefore, experimental results obtained from animal models do not perfectly recapitulate the conditions occurring in humans, and are less reliable for the purpose of extrapolation. In contrast, disease-specific hiPSCs could be a valuable tool in research on inherited diseases and for testing therapeutic agents. hiPSCs are created from somatic cells, which can be easily collected from accessible patient tissues, such as skin and blood. Owing to their self-renewal property, hiPSCs could be used to produce a sufficient number of specific cell types following appropriate differentiation methods for further experiments in vitro.

Human Induced Pluripotent Stem Cell Applications in Cardiovascular Medicine

Open in new tab
Open ppt

Human Induced Pluripotent Stem Cells for Modelling Inherited Arrhythmias

Advances in cardiovascular research have increased our understanding of the molecular mechanisms underlying various genetic diseases. Comprehensive genetic studies have identified causal mutations responsible for phenotypes of inherited cardiovascular diseases such as long QT syndrome (LQTS), Brugada syndrome and cardiomyopathies.

LQTS is characterised by a significantly prolonged QT interval attributable to delayed repolarisation in the ventricular myocardium. Some types of LQTS cause life-threatening arrhythmias in response to stimuli such as swimming and sudden loud noise. Genetic studies have found a number of gene loci responsible for LQTS in families with a high incidence of the disease. Despite an absence of clinical symptoms under sedentary conditions in patients with LQTS, once ventricular tachyarrhythmias are triggered by specific stimuli, patients with LQTS are prone to exhibit syncope. Sustained arrhythmias ultimately lead to VF, resulting in sudden cardiac death. Several studies on patients with LQTS have identified a number of mutations in genes encoding cardiac ion channels, which are membrane proteins regulating the generation and propagation of action potential.8–10 However, these mutations are not always responsible for the observed symptoms, even when the patients are exposed to the stimuli that trigger electrophysiological changes.

Effects of the stimuli or therapeutic agents, as well as the incidence of cardiac events, vary considerably among individual patients. Therefore, to address issues related to proarrhythmic mechanisms in individuals with inherited LQTS, patient-derived hiPSC-CMs with the corresponding mutation(s) could serve as powerful tools for in vitro experiments. Previous studies characterising mutations of the alpha-subunit of the potassium voltage-gated channel subfamily Q member 1 (KCNQ1; also known as KVLQT1 and KV7.1) using patient-derived iPSC-CMs revealed that impaired membrane trafficking of Ks channels and reduced delayed rectifier potassium channel current (IKr) cause LQT1.9,11,12 Itzhaki et al. introduced reprogramming factors into dermal fibroblasts obtained from patients with a mutation in the alpha-subunit of potassium voltage-gated channel subfamily H member 2 (KCNH2; responsible for IKr) causing LQT2.13 Spontaneously beating hiPSC-CMs carrying this mutation were used for functional analysis and exhibited a prolonged QT interval similar to that in LQTS patients.

Similar studies using hiPSC-CMs derived from a patient with a missense mutation in KCNH2 also exhibited action potential prolongation, smaller IKr, early afterdepolarisations and arrhythmias. These changes were recovered or exaggerated by pharmacological agents or selective RNA interference in disease-specific hiPSC-CMs.13–16

Disease-specific hiPSC-CMs from patients and families with Timothy syndrome (LQT8) that have a mutation located in calcium voltage-gated channel subunit alpha1 C (CACNA1C; responsible for the L-type calcium current, ICa,L) have been established and assessed for mutation-associated phenotypes in vitro.10,17,18 An LQT8 model using patient-specific hiPSC-CMs reflected cellular electrical abnormalities, including prolonged action potential duration, delayed afterdepolarisations and altered Ca2+ transients. In contrast, roscovitine, an inhibitor of cyclin-dependent kinase 5, a key mediator involved in the regulation of CaV1.2 channels, enhanced ICa,L inactivation, shortened action potential duration, restored the irregular Ca2+ transient and decreased the frequency of abnormal depolarisations in LQT8 hiPSC-CMs.10,17,18

Furthermore, other inherited arrhythmias have been investigated using disease-specific hiPSC-CMs, including various types of LQTS – mutations in sodium voltage-gated channel alpha subunit 5 (SCN5A), potassium inwardly rectifying channel subfamily J member 2 (KCNJ2), calmodulin 1 (CALM1) or calmodulin 2 (CALM2), short QT syndrome (KCNH2 mutation), Brugada syndrome type 1 (SCN5A mutation) and catecholaminergic polymorphic ventricular tachycardia (mutations in ryanodine receptor 2 (RYR2) or calsequestrin 2 [CASQ2]).19–40 These cells recapitulated cellular electrophysiological changes in the heart of patients. Table 1 summarises the different studies that have used hiPSC-CMs as models to investigate inherited arrhythmias.

Human Induced Pluripotent Stem Cells for Modelling of Inherited Cardiomyopathies

In addition to inherited arrhythmias, there are some incidences of cardiomyopathies in families carrying specific genetic variant(s) that are responsible for causing the disease. Dilated cardiomyopathy (DCM) is a major type of cardiomyopathy that is characterised by systolic dysfunction and dilated cardiac chambers comprised of thin myocardial walls.41 Most cases of DCM without any identifiable cause (e.g. coronary artery disease, systemic hypertension, viral infection) are diagnosed as ‘idiopathic’ DCM.

Based on family history and clinical findings, including sudden cardiac death, heart failure and abnormal echocardiography, previous clinical studies have proposed that familial transmission of idiopathic DCM is observed in 20–50% of patients.42–44 When idiopathic DCM is identified in two or more family members, it is defined as familial DCM (FDC). FDC is largely caused by autosomal dominant mutations in key cardiac genes encoding sarcomere-related proteins, cytoskeletal proteins, mitochondrial proteins, nuclear membrane proteins and calcium regulators.43,45,46 These loss-of-function mutations lead to the abnormal morphology and function of the heart that is seen in idiopathic DCM. Moreover, recently developed high-throughput gene analyses have revealed that inherited DCM is associated with mutations in more than 100 gene loci.47

Human Induced Pluripotent Stem Cell

Open in new tab
Open ppt

Human Induced Pluripotent Stem Cell

Open in new tab
Open ppt

Although the pathophysiology of FDC is heterogeneous, the effect of each individual mutation has been unclear in the context of FDC. To address this, human CMs are ideal for in vitro functional analysis of mutations associated with FDC, but, as mentioned earlier, it is difficult to acquire a renewable source of cardiac cells. Compared with animal models and non-CMs expressing DCM mutant proteins, hiPSC-CMs are expected to exhibit responses similar to those observed in native human myocardium. For example, individual families carry a mutation that causes an arginine-to-tryptophan substitution at amino acid position 173 in the cardiac troponin T (cTnT) protein.48 Patient-specific hiPSCs were produced using minimally invasive procedures from skin fibroblasts of family members, and hiPSC-CMs were generated and tested to investigate the mechanisms underlying FDC. The FDC hiPSC-CMs exhibited reduced Ca2+ influx and contractility, despite normal electrophysiological properties. These cells also showed the characteristic patchy structure of myofilaments, which was enhanced upon noradrenaline stimulation and stretching, leading to systolic dysfunction.48

This is consistent with the fact that the tendency towards DCM is enhanced by increases in inotropic effects and hypertension. These findings explain the involvement of cTnT dysfunction in the development of DCM. Thus, FDC hiPSC-CMs recreate, at least in part, the pathophysiology of FDC in human patients. Other causal gene mutations responsible for inherited cardiomyopathies, including DCM, hypertrophic cardiomyopathy and arrhythmogenic right ventricular cardiomyopathy/dysplasia, have been reported.49–57 Table 2 lists studies that have used hiPSC-CMs as models for investigating inherited cardiomyopathies.

Although numerous studies have summarised the characteristic features of familial heart diseases using patient-specific hiPSC-CMs, as described above, it is still challenging to fully recapitulate the disease phenotype using iPSC-CM-based disease modelling, primarily because hiPSC-CMs exhibit immature functions and morphology. For example, an incomplete ion channel profile (e.g. lack of IK1, corresponding to slower action potential kinetics and a relatively positive diastolic potential) and subcellular structure (e.g. the absence of or underdeveloped T-tubule and sarcomere formation) are commonly observed in hiPSC-CMs.58–60 The gene expression profile of hiPSC-CMs also resembles that of foetal CMs and is distinct from that of adult CMs.60,61 The immaturity of hiPSC-CMs in terms of function and gene expression profile may result in controversial findings, particularly in the investigation of late-onset cardiac diseases that largely require adult CM-like cells for disease modelling.

In an in vitro study using hiPSC-CMs to investigate the pathophysiology of late-onset Pompe disease, which is characterised by slow progression of muscle weakness, although patient-specific hiPSC-CMs exhibited typical features associated with the disease, such as intracellular glycogen accumulation and mitochondrial dysfunction, they did not fully exhibit the autophagic abnormalities that are observed in vivo.62,63 This may be overcome by using fully differentiated hiPSC-CMs assembled along with a complete subcellular system for muscle contraction, Ca2+ cycling, metabolism and protein recycling. Recent studies have contributed to the development of protocols for the maturation of hiPSC-CMs using electrical and/or mechanical stimulation, a 3D culture system with scaffold materials, coculture with fibroblasts or CMs in vitro and in vivo and a combination of these techniques, leading to improvement in contractility, Ca2+ handling and electrophysiological properties.64–68

Lack of chamber-specific characteristics is another major concern regarding the use of hiPSC-CMs for disease modelling. As the structure, haemodynamic stress, developmental origin and protein expression profile are quite distinctive among the cardiac chambers,59,69,70 the molecular features of individual CMs in each chamber would also differ. Some inherited arrhythmias and cardiomyopathies have chamber-specific characteristics. Clinical phenotypes of Brugada syndrome and ARVC/D likely originate from the right ventricular outflow tract. However, disease models based on hiPSC-CMs may not fully recapitulate the characteristic features of any specific region of the heart.

A differentiated hiPSC-CM cluster usually consists of electrophysiologically heterogeneous subtypes including ventricular-, atrial- and nodal-like myocytes. The ventricular-like hiPSC-CMs exhibit properties analogous to those of human ventricular myocytes (e.g. steep upstroke (Phase 0) and plateau phase (Phase 4) of action potentials), whereas the nodal-type hiPSC-CMs exhibit slower action potential kinetics and depolarising diastolic potential.71 This mixed subtype of hiPSC-CMs leads to a wide range of results rather than being representative of a specific subtype of CMs. The development of protocols for subtype-specific and/or chamber-specific differentiation of hiPSC-CMs will accelerate research to identify the chamber-specific phenotypes associated with heart diseases. Although some genetic heart diseases are rare, many of them lead to life-threatening conditions. Therefore, further intensive research using disease-specific hiPSC-CMs should be promoted to gain insights into the underlying mechanisms and to identify potential therapeutic targets of these genetic diseases in order to develop novel therapeutic approaches for individual patients.

Human Induced Pluripotent Stem Cells as a Tool for Drug Screening

Currently, the development of new drugs requires multiple processes, including screening of numerous putative drug compounds based on chemical structure and in vitro assays of pharmacological activity, followed by analyses of pharmacokinetics and safety in vitro and in vivo and, finally, clinical trials in humans. In most cases, these processes take many years until the candidate compounds are tested in humans.72 Even though the effectiveness of compounds may be promising in cell culture and animal experiments, problems identified in clinical trials assessing the effects of these compounds on the QT interval (known as a thorough QT/QTc study) following pharmacokinetics examination in humans may halt the further development of these compounds. However, if human cardiac cells were widely available, drug testing in human CMs might provide effective and safe drug candidates rapidly and economically, because the response to compounds tested using in vitro experiments with human CMs could resemble that of the human body.

Human Induced Pluripotent Stem Cell-Derived

Open in new tab
Open ppt

Disease-specific hiPSC-derived CMs potentially exhibit similar physiological characteristics as diseased cells in patients, and may be a useful tool to predict the benefits and side-effects of drug candidates in patients. Drug screening using hiPSC-CMs to detect side effects such as drug-induced QT prolongation and ventricular tachyarrhythmias could contribute to the early withdrawal of therapeutic compounds with undesirable cardiac effects before the initiation of in vivo experiments and clinical trials.72,73 Other than the development of new drugs, the cardiac side effects of some already marketed drugs, including anti-arrhythmic drugs and non-cardiac drugs such as antihistamines, antipsychotics and anti-infective drugs, have been widely recognised. These drugs have the potential to cause torsade de pointes, in combination with other endogenous and environmental factors.73 Drug testing using hiPSC-CMs may also be applicable in this context.

Although hiPSC-CMs share some characteristics with adult human ventricular myocytes, hiPSC-CMs are commonly known to exhibit the features of foetal ‘immature’ CMs in terms of their gene expression profile, structure and electrophysiology, as noted above. hiPSC-CMs express cardiac-specific genes (e.g. those encoding cTnT, alpha-myosin heavy chain) and exhibit ion channel activity (e.g. similar INa, IKr and ICa,L current density to that in adult ventricular CMs);12–14,16,71,74–83 however, morphologically they are more rounded or multiangular in shape and smaller in size, with disorganised myofibrils and a lack of t-tubules, which contribute to the slower kinetics of the Ca2+ transient.38,76,83–87 These important differences should be considered when using hiPSC-CMs in drug screening. Further investigations are needed to develop optimal methods for more efficient differentiation into functional CMs that exhibit the typical properties of adult CMs.

Gene Editing to Create Disease-Specific Human Induced Pluripotent Stem Cells

Comprehensive genetic studies have identified causal mutations responsible for genetic heart diseases. hiPSC-CMs have emerged as a highly effective tool for modelling such diseases. Although it is technically possible to induce disease-specific hiPSC-CMs, patient-derived somatic cells may not be readily available, especially in the case of rare diseases. In addition, interclonal variation is seen among hiPSC clones, resulting from different genetic backgrounds associated with individual cells.

Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein (Cas) 9 is a gene-editing technology that can solve the challenges associated with the genetic variability.88,89 CRISPR is a DNA sequence found in bacterial genomes; it is thought to be derived from viruses, is known to protect bacteria from repeated viral infections and acts as a basic adaptive immune system for prokaryotes. Cas9 is a DNA-cutting enzyme that recognises CRISPR sequences and causes site-specific DNA double-strand breaks (Figure 2). Recent advances in CRISPR/Cas9-based gene editing have markedly improved the efficiency and specificity of the method and expanded its applications, including knockout, repression and activation of genes of interest.90

In phenotypic analysis of monogenic inherited diseases, this technology is also applicable to either disease-associated mutagenesis in wild-type hiPSCs or to the correction of pathogenic gene mutations in disease-specific hiPSCs (Figure 3).89 Analysis of disease-specific hiPSCs versus wild-type hiPSCs established from healthy donor cells as a control may result in unreliable outcomes due to the different genetic backgrounds of the disease-specific hiPSCs and control cells. However, CRISPR/Cas9-based gene editing enables the preparation of an isogenic control by normalising a disease-relevant mutation in disease-specific hiPSCs or by inducing the mutation in wild-type hiPSCs so that diseased and control cells with the same genetic background are obtained. In addition, CRISPR/Cas9-based gene editing could allow the production of isogenic cells with intact and/or corrected variant alleles in non-coding regions including enhancers that may reveal the role of mutations in the transcriptional regulation of genes responsible for a disease phenotype. This method shows promise for the proper evaluation of the involvement of mutated genes in disease phenotype following in vitro differentiation (Figure 3).

Principle of Clustered Regularly Interspaced Short Palindromic Repeat

Open in new tab
Open ppt

Gene Editing in Human Induced Pluripotent Stem Cell

Open in new tab
Open ppt

Polygenic diseases, which differ from monogenic inherited diseases in that more than one gene is involved in their dysfunction, impose another limitation on the use of hiPSCs. Polygenic diseases are thought to be caused by a combination of multiple mutations, each of which has a small effect, with or without extrinsic factors. Although gene editing has been used to edit multiple regions of the genome, a major challenge towards using hiPSCs to investigate polygenic diseases is identification of the corresponding mutations and understanding how each mutation contributes to the pathogenesis of these multifactorial diseases. Moreover, in some cases, environmental factors may strongly affect disease phenotypes, making experimental conditions and further analysis more complicated. Comprehensive reviews are available for detailed information regarding the use of gene editing in iPSC research.89,91

Consideration of Human Induced Pluripotent Stem Cells for Application in Disease Modelling and Clinical Use

Despite extensive benefits, there are still many unsolved issues regarding the use of hiPSCs in further applications. One of the major issues is that the quality of individual hiPSC lines is variable, even when an hiPSC line is derived from one individual. Classical iPSC reprogramming methods using retroviral or lentiviral vectors may cause random insertional mutations in the host genome, resulting in alteration of subsequent cell phenotypes.92

Recent advances in reprogramming strategies using non-integrating, virus-free and vector-free methods are overcoming this issue.93,94 However, it is still technically difficult to eliminate the risk of gene mutations during the reprogramming process because forced expression of reprogramming factors can induce DNA damage.95 In fact, protein-coding point mutations acquired during or after reprogramming were identified in multiple hiPSC lines, some of which exhibit unpredictable phenotypes.96 Thus, accumulating evidence regarding the mechanism underlying the reprogramming of iPSCs is expected to provide insights into how the quality of hiPSC lines may be stabilised and standardised for use as a cell source for further experiments and clinical application.

Precise investigations into the pathophysiology of inherited diseases using patient-derived iPSCs require improved protocols that allow highly efficient differentiation of hiPSCs into a specific cell type, because the differentiation efficiency in current experiments remains significantly lower than what is desired. The characteristic variability of cells differentiated from disease-specific hiPSCs is a considerable hurdle that research into pathophysiology must overcome. Epigenetic modifications are presumably one of the causes of phenotype variability. Optimised sorting methods to collect only a desired cell type from the heterogeneous cell population need to be developed. Current research efforts are advancing cardiac differentiation protocols to generate spontaneously beating CM-like cell clusters, but the clusters of differentiated cells that are heterogeneous also contain other mesodermal derivatives, such as smooth muscle cells and endothelial cells, as well as undifferentiated cells, which may increase the risk of tumourigenesis.

Pathophysiological studies using disease-specific hiPSCs allow us to determine the cellular characteristics of a disease, but do not recreate the function of the whole organ within the body. Although complex bioengineering approaches, such as organoid formation and 3D culture systems, are available,97,98 it is difficult to use these methods in the heart because CMs in the heart are predominantly situated in a highly organised structure comprising vessels, nerves, mesenchymal cells, extracellular matrix and myocytes. In addition, CMs are continuously exposed to dynamically changing neuroendocrine factors and mechanical stresses. Therefore, it should be considered that studies using disease-specific hiPSC-CMs fundamentally provide simplified information regarding the pathophysiology in patients with a familial disease. Nevertheless, the experimental data from these cells may reveal responses that mirror actual phenomena in human patients, and are thus valuable for gaining an understanding of the inherited disease.

Conclusion

Disease-specific hiPSC-CMs, which carry the same genomic information as patients with inherited diseases, can undoubtedly be of use in research to address the pathophysiology of monogenic inherited diseases, the drug responsiveness of patients for personalised medicine and drug development by providing a cell source for screening compounds and drug safety testing. A combination of disease-specific hiPSC-CMs and gene-editing technologies may further advance our understanding of genetic diseases and drug development in cardiovascular medicine.

References

  1. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007;131:861–72.
    Crossref | PubMed
  2. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126:663–76.
    Crossref | PubMed
  3. Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007;318:1917–20.
    Crossref | PubMed
  4. Park IH, Zhao R, West JA, et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature 2008;451:141–6.
    Crossref | PubMed
  5. Takahashi K, Yamanaka S. A decade of transcription factor-mediated reprogramming to pluripotency. Nat Rev Mol Cell Biol 2016;17:183–93.
    Crossref | PubMed
  6. Muller R, Lengerke C. Patient-specific pluripotent stem cells: promises and challenges. Nat Rev Endocrinol 2009;5:195–203.
    Crossref | PubMed
  7. Siller R, Greenhough S, Park IH, Sullivan GJ. Modelling human disease with pluripotent stem cells. Curr Gene Ther 2013;13:99–110.
    Crossref | PubMed
  8. Matsa E, Rajamohan D, Dick E, et al. Drug evaluation in cardiomyocytes derived from human induced pluripotent stem cells carrying a long QT syndrome type 2 mutation. Eur Heart J 2011;32:952–62.
    Crossref | PubMed
  9. Moretti A, Bellin M, Welling A, et al. Patient-specific induced pluripotent stem-cell models for long-QT syndrome. N Engl J Med 2010;363:1397–409.
    Crossref | PubMed
  10. Yazawa M, Hsueh B, Jia X, et al. Using induced pluripotent stem cells to investigate cardiac phenotypes in Timothy syndrome. Nature 2011;471:230–4.
    Crossref | PubMed
  11. Egashira T, Yuasa S, Suzuki T, et al. Disease characterization using LQTS-specific induced pluripotent stem cells. Cardiovasc Res 2012;95:419–29.
    Crossref | PubMed
  12. Ma D, Wei H, Lu J, 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. Stem Cell Res Ther 2015;6:39.
    Crossref | PubMed
  13. Itzhaki I, Maizels L, Huber I, et al. Modelling the long QT syndrome with induced pluripotent stem cells. Nature 2011;471:225–9.
    Crossref | PubMed
  14. Lahti AL, Kujala VJ, Chapman H, et al. Model for long QT syndrome type 2 using human iPS cells demonstrates arrhythmogenic characteristics in cell culture. Dis Model Mech 2012;5:220–30.
    Crossref | PubMed
  15. Matsa E, Dixon JE, Medway C, et al. Allele-specific RNA interference rescues the long-QT syndrome phenotype in human-induced pluripotency stem cell cardiomyocytes. Eur Heart J 2014;35:1078–87.
    Crossref | PubMed
  16. Sala L, Yu Z, Ward-van Oostwaard D, 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. EMBO Mol Med 2016;8:1065–81.
    Crossref | PubMed
  17. Song L, Park SE, Isseroff Y, et al. Inhibition of CDK5 alleviates the cardiac phenotypes in Timothy syndrome. Stem Cell Rep 2017;9:50–7.
    Crossref | PubMed
  18. Yazawa M, Dolmetsch RE. Modeling Timothy syndrome with iPS cells. J Cardiovasc Transl Res 2013;6:1–9.
    Crossref | PubMed
  19. Davis RP, Casini S, van den Berg CW, et al. Cardiomyocytes derived from pluripotent stem cells recapitulate electrophysiological characteristics of an overlap syndrome of cardiac sodium channel disease. Circulation 2012;125:3079–91.
    Crossref | PubMed
  20. Fatima A, Kaifeng S, Dittmann S, et al. The disease-specific phenotype in cardiomyocytes derived from induced pluripotent stem cells of two long QT syndrome type 3 patients. PLoS One 2013;8:e83005.
    Crossref | PubMed
  21. Kuroda Y, Yuasa S, Watanabe Y, et al. Flecainide ameliorates arrhythmogenicity through NCX flux in Andersen–Tawil syndrome-iPS cell-derived cardiomyocytes. Biochem Biophys Rep 2017;9:245–56.
    Crossref | PubMed
  22. Limpitikul WB, Dick IE, Tester DJ, et al. A precision medicine approach to the rescue of function on malignant calmodulinopathic long-QT syndrome. Circ Res 2017;120:39–48.
    Crossref | PubMed
  23. Ma D, Wei H, Zhao Y, et al. Modeling type 3 long QT syndrome with cardiomyocytes derived from patient-specific induced pluripotent stem cells. Int J Cardiol 2013;168:5277–86.
    Crossref | PubMed
  24. Malan D, Zhang M, Stallmeyer B, et al. Human iPS cell model of type 3 long QT syndrome recapitulates drug-based phenotype correction. Basic Res Cardiol 2016;111:14.
    PubMed
  25. Rocchetti M, Sala L, Dreizehnter L, et al. Elucidating arrhythmogenic mechanisms of long-QT syndrome CALM1-F142L mutation in patient-specific induced pluripotent stem cell-derived cardiomyocytes. Cardiovasc Res 2017;113:531–41.
    Crossref | PubMed
  26. Terrenoire C, Wang K, Tung KW, et al. Induced pluripotent stem cells used to reveal drug actions in a long QT syndrome family with complex genetics. J Gen Physiol 2012;141:61–72.
    Crossref | PubMed
  27. Yamamoto Y, Makiyama T, Harita T, et al. Allele-specific ablation rescues electrophysiological abnormalities in a human iPS cell model of long-QT syndrome with a CALM2 mutation. Hum Mol Genet 2017;26:1670–7.
    Crossref | PubMed
  28. El-Battrawy I, Lan H, Cyganek L, et al. Modeling short QT syndrome using human-induced pluripotent stem cell-derived cardiomyocytes. J Am Heart Assoc 2018;7:e007394.
    Crossref | PubMed
  29. Cerrone M, Lin X, Zhang M, et al. Missense mutations in plakophilin-2 cause sodium current deficit and associate with a Brugada syndrome phenotype. Circulation 2014;129:1092–103.
    Crossref | PubMed
  30. Liang P, Sallam K, Wu H, et al. Patient-specific and genome-edited induced pluripotent stem cell-derived cardiomyocytes elucidate single-cell phenotype of Brugada syndrome. J Am Coll Cardiol 2016;68:2086–96.
    Crossref | PubMed
  31. Selga E, Sendfeld F, Martinez-Moreno R, et al. Sodium channel current loss of function in induced pluripotent stem cell-derived cardiomyocytes from a Brugada syndrome patient. J Mol Cell Cardiol 2018;114:10–9.
    Crossref | PubMed
  32. Fatima A, Xu G, Shao K, et al. In vitro modeling of ryanodine receptor 2 dysfunction using human induced pluripotent stem cells. Cell Physiol Biochem 2011;28:579–92.
    Crossref | PubMed
  33. Itzhaki I, Maizels L, Huber I, et al. Modeling of catecholaminergic polymorphic ventricular tachycardia with patient-specific human-induced pluripotent stem cells. J Am Coll Cardiol 2012;60:990–1000.
    Crossref | PubMed
  34. Jung CB, Moretti A, Mederos y Schnitzler M, et al. Dantrolene rescues arrhythmogenic RYR2 defect in a patient-specific stem cell model of catecholaminergic polymorphic ventricular tachycardia. EMBO Mol Med 2012;4:180–91.
    Crossref | PubMed
  35. Kujala K, Paavola J, Lahti A, et al. Cell model of catecholaminergic polymorphic ventricular tachycardia reveals early and delayed afterdepolarizations. PLoS One 2012;7:e44660.
    Crossref | PubMed
  36. Maizels L, Huber I, Arbel G, et al. Patient-specific drug screening using a human induced pluripotent stem cell model of catecholaminergic polymorphic ventricular tachycardia type 2. Circ Arrhythm Electrophysiol 2017;10:pii:e004725.
    Crossref | PubMed
  37. Novak A, Barad L, Lorber A, et al. Functional abnormalities in iPSC-derived cardiomyocytes generated from CPVT1 and CPVT2 patients carrying ryanodine or calsequestrin mutations. J Cell Mol Med 2015;19:2006–18.
    Crossref | PubMed
  38. Novak A, Barad L, Zeevi-Levin N, et al. Cardiomyocytes generated from CPVTD307H patients are arrhythmogenic in response to beta-adrenergic stimulation. J Cell Mol Med 2012;16:468–82.
    Crossref | PubMed
  39. Preininger MK, Jha R, Maxwell JT, et al. A human pluripotent stem cell model of catecholaminergic polymorphic ventricular tachycardia recapitulates patient-specific drug responses. Dis Model Mech 2016;9:927–39.
    Crossref | PubMed
  40. Sasaki K, Makiyama T, Yoshida Y, 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. PLoS One 2016;11:e0164795.
    Crossref | PubMed
  41. Maron BJ, Towbin JA, Thiene G, et al. Contemporary definitions and classification of the cardiomyopathies. Circulation 2006;113:1807–16.
    Crossref | PubMed
  42. Baig MK, Goldman JH, Caforio ALP, et al. Familial dilated cardiomyopathy: cardiac abnormalities are common in asymptomatic relatives and may represent early disease. J Am Coll Cardiol 1998;31:195–201.
    Crossref | PubMed
  43. Burkett EL, Hershberger RE. Clinical and genetic issues in familial dilated cardiomyopathy. J Am Coll Cardiol 2005;45:969–81.
    Crossref | PubMed
  44. Grunig E, Tasman JA, Kucherer H, et al. Frequency and phenotypes of familial dilated cardiomyopathy. J Am Coll Cardiol 1998;31:186–94.
    Crossref | PubMed
  45. Morita H, Seidman J, Seidman CE. Genetic causes of human heart failure. J Clin Invest 2005;115:518–26.
    Crossref | PubMed
  46. Mestroni L, Rocco C, Gregori D, et al. Familial dilated cardiomyopathy: evidence for genetic and phenotypic heterogeneity. Heart Muscle Disease Study Group. J Am Coll Cardiol 1999;34:181–90.
    Crossref | PubMed
  47. Dellefave L, McNally EM. The genetics of dilated cardiomyopathy. Curr Opin Cardiol 2010;25:198–204.
    Crossref | PubMed
  48. Sun N, Yazawa M, Liu JW, et al. Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy. Sci Transl Med 2012;4:130ra47.
    Crossref | PubMed
  49. Tse HF, Ho JC, Choi SW, et al. Patient-specific induced-pluripotent stem cells-derived cardiomyocytes recapitulate the pathogenic phenotypes of dilated cardiomyopathy due to a novel DES mutation identified by whole exome sequencing. Hum Mol Genet 2013;22:1395–403.
    Crossref | PubMed
  50. Carvajal-Vergara X, Sevilla A, D’Souza SL, et al. Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome. Nature 2010;465:808–12.
    Crossref | PubMed
  51. Huang HP, Chen PH, Hwu WL, et al. Human Pompe disease-induced pluripotent stem cells for pathogenesis modeling, drug testing and disease marker identification. Hum Mol Genet 2011;20:4851–64.
    Crossref | PubMed
  52. Lan F, Lee AS, Liang P, et al. Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell 2013;12:101–13.
    Crossref | PubMed
  53. Lee YK, Lau YM, Cai ZJ, et al. Modeling treatment response for lamin A/C related dilated cardiomyopathy in human induced pluripotent stem cells. J Am Heart Assoc 2017;6:pii:e005677.
    Crossref | PubMed
  54. Siu CW, Lee YK, Ho JC, et al. Modeling of lamin A/C mutation premature cardiac aging using patient-specific induced pluripotent stem cells. Aging (Albany NY) 2012;4:803–22.
    Crossref | PubMed
  55. Caspi O, Huber I, Gepstein A, et al. Modeling of arrhythmogenic right ventricular cardiomyopathy with human induced pluripotent stem cells. Circ Cardiovasc Genet 2013;6:557–68.
    Crossref | PubMed
  56. Kim C, Wong J, Wen JY, et al. Studying arrhythmogenic right ventricular dysplasia with patient-specific iPSCs. Nature 2013;494:105–10.
    Crossref | PubMed
  57. Ma D, Wei H, Lu J, et al. Generation of patient-specific induced pluripotent stem cell-derived cardiomyocytes as a cellular model of arrhythmogenic right ventricular cardiomyopathy. Eur Heart J 2013;34:1122–33.
    Crossref | PubMed
  58. Cordeiro JM, Nesterenko VV, Sicouri S, et al. Identification and characterization of a transient outward K+ current in human induced pluripotent stem cell-derived cardiomyocytes. J Mol Cell Cardiol 2013;60:36–46.
    Crossref | PubMed
  59. Doll S, Dressen M, Geyer PE, et al. Region and cell-type resolved quantitative proteomic map of the human heart. Nat Commun 2017;8:1469.
    Crossref | PubMed
  60. Karakikes I, Ameen M, Termglinchan V, Wu JC. Human induced pluripotent stem cell-derived cardiomyocytes: insights into molecular, cellular, and functional phenotypes. Circ Res 2015;117:80–8.
    Crossref | PubMed
  61. Yang X, Pabon L, Murry CE. Engineering adolescence: maturation of human pluripotent stem cell-derived cardiomyocytes. Circ Res 2014;114:511–23.
    Crossref | PubMed
  62. Fukuda T, Ahearn M, Roberts A, et al. Autophagy and mistargeting of therapeutic enzyme in skeletal muscle in Pompe disease. Mol Ther 2006;14:831–9.
    Crossref | PubMed
  63. Raben N, Ralston E, Chien YH, et al. Differences in the predominance of lysosomal and autophagic pathologies between infants and adults with Pompe disease: implications for therapy. Mol Genet Metab 2010;101:324–31.
    Crossref | PubMed
  64. Veerman CC, Kosmidis G, Mummery CL, et al. Immaturity of human stem-cell-derived cardiomyocytes in culture: fatal flaw or soluble problem? Stem Cells Dev 2015;24:1035–52.
    Crossref | PubMed
  65. Denning C, Borgdorff V, Crutchley J, et al. Cardiomyocytes from human pluripotent stem cells: from laboratory curiosity to industrial biomedical platform. Biochim Biophys Acta 2016;1863:1728–48.
    Crossref | PubMed
  66. Mathur A, Ma Z, Loskill P, et al. In vitro cardiac tissue models: Current status and future prospects. Adv Drug Deliv Rev 2016;96:203–13.
    Crossref | PubMed
  67. Cho GS, Lee DI, Tampakakis E, et al. Neonatal transplantation confers maturation of PSC-derived cardiomyocytes conducive to modeling cardiomyopathy. Cell Rep 2017;18:571–82.
    Crossref | PubMed
  68. Nunes SS, Miklas JW, Liu J, et al. Biowire: a platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nat Methods 2013;10:781–7.
    Crossref | PubMed
  69. Rana MS, Christoffels VM, Moorman AF. A molecular and genetic outline of cardiac morphogenesis. Acta Physiol (Oxf) 2013;207:588–615.
    Crossref | PubMed
  70. Srivastava D. Making or breaking the heart: from lineage determination to morphogenesis. Cell 2006;126:1037–48.
    Crossref | PubMed
  71. Ma J, Guo L, Fiene SJ, et al. High purity human-induced pluripotent stem cell-derived cardiomyocytes: electrophysiological properties of action potentials and ionic currents. Am J Physiol Heart Circ Physiol 2011;301:H2006–17.
    Crossref | PubMed
  72. Gromo G, Mann J, Fitzgerald JD. Cardiovascular drug discovery: a perspective from a research-based pharmaceutical company. Cold Spring Harb Perspect Med 2014;4:a014092.
    Crossref | PubMed
  73. Ritter JM. Cardiac safety, drug-induced QT prolongation and torsade de pointes (TdP). Br J Clin Pharmacol 2012;73:331–4.
    Crossref | PubMed
  74. Bellin M, Casini S, Davis RP, et al. Isogenic human pluripotent stem cell pairs reveal the role of a KCNH2 mutation in long-QT syndrome. EMBO J 2013;32:3161–75.
    Crossref | PubMed
  75. Doss MX, Di Diego JM, Goodrow RJ, et al. Maximum diastolic potential of human induced pluripotent stem cell-derived cardiomyocytes depends critically on I(Kr). PLoS One 2012;7:e40288.
    Crossref | PubMed
  76. Hwang HS, Kryshtal DO, Feaster TK, et al. Comparable calcium handling of human iPSC-derived cardiomyocytes generated by multiple laboratories. J Mol Cell Cardiol 2015;85:79–88.
    Crossref | PubMed
  77. Iost N, Virag L, Opinicariu M, et al. Delayed rectifier potassium current in undiseased human ventricular myocytes. Cardiovasc Res 1998;40:508–15.
    Crossref | PubMed
  78. Ivashchenko CY, Pipes GC, Lozinskaya IM, et al. Human-induced pluripotent stem cell-derived cardiomyocytes exhibit temporal changes in phenotype. Am J Physiol Heart Circ Physiol 2013;305:H913–22.
    Crossref | PubMed
  79. Jost N, Acsai K, Horvath B, et al. Contribution of I Kr and I K1 to ventricular repolarization in canine and human myocytes: is there any influence of action potential duration? Basic Res Cardiol 2009;104:33–41.
    Crossref | PubMed
  80. Lee S, Lee HA, Choi SW, et al. Evaluation of nefazodone-induced cardiotoxicity in human induced pluripotent stem cell-derived cardiomyocytes. Toxicol Appl Pharmacol 2016;296:42–53.
    Crossref | PubMed
  81. Zhang J, Wilson GF, Soerens AG, et al. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ Res 2009;104:e30–41.
    Crossref | PubMed
  82. Zhang M, D’Aniello C, Verkerk AO, et al. Recessive cardiac phenotypes in induced pluripotent stem cell models of Jervell and Lange-Nielsen syndrome: disease mechanisms and pharmacological rescue. Proc Natl Acad Sci USA 2014;111:e5383–92.
    Crossref | PubMed
  83. Zhang XH, Haviland S, Wei H, et al. Ca2+ signaling in human induced pluripotent stem cell-derived cardiomyocytes (iPS-CM) from normal and catecholaminergic polymorphic ventricular tachycardia (CPVT)-afflicted subjects. Cell Calcium 2013;54:57–70.
    Crossref | PubMed
  84. Dick E, Rajamohan D, Ronksley J, Denning C. Evaluating the utility of cardiomyocytes from human pluripotent stem cells for drug screening. Biochem Soc Trans 2010;38:1037–45.
    Crossref | PubMed
  85. Itzhaki I, Rapoport S, Huber I, et al. Calcium handling in human induced pluripotent stem cell derived cardiomyocytes. PLoS One 2011;6:e18037.
    Crossref | PubMed
  86. Spencer CI, Baba S, Nakamura K, et al. Calcium transients closely reflect prolonged action potentials in iPSC models of inherited cardiac arrhythmia. Stem Cell Rep 2014;3:269–81.
    Crossref | PubMed
  87. Devalla HD, Gelinas R, Aburawi EH, et al. TECRL, a new life-threatening inherited arrhythmia gene associated with overlapping clinical features of both LQTS and CPVT. EMBO Mol Med 2016;8:1390–408.
    Crossref | PubMed
  88. Jinek M, Chylinski K, Fonfara I, et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012;337:816–21.
    Crossref | PubMed
  89. Hockemeyer D, Jaenisch R. Induced pluripotent stem cells meet genome editing. Cell Stem Cell 2016;18:573–86.
    Crossref | PubMed
  90. Wang F, Qi LS. Applications of CRISPR genome engineering in cell biology. Trends Cell Biol 2016;26:875–88.
    Crossref | PubMed
  91. Soldner F, Jaenisch R. Stem cells, genome editing, and the path to translational medicine. Cell 2018;175:615–32.
    Crossref | PubMed
  92. Saha K, Jaenisch R. Technical challenges in using human induced pluripotent stem cells to model disease. Cell Stem Cell 2009;5:584–95.
    Crossref | PubMed
  93. de Almeida PE, Ransohoff JD, Nahid A, Wu JC. Immunogenicity of pluripotent stem cells and their derivatives. Circ Res 2013;112:549–61.
    Crossref | PubMed
  94. Okano H, Nakamura M, Yoshida K, et al. Steps toward safe cell therapy using induced pluripotent stem cells. Circ Res 2013;112:523–33.
    Crossref | PubMed
  95. Kawamura T, Suzuki J, Wang YV, et al. Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature 2009;460:1140–4.
    Crossref | PubMed
  96. Gore A, Li Z, Fung HL, et al. Somatic coding mutations in human induced pluripotent stem cells. Nature 2011;471:63–7.
    Crossref | PubMed
  97. Masumoto H, Ikuno T, Takeda M, et al. Human iPS cell-engineered cardiac tissue sheets with cardiomyocytes and vascular cells for cardiac regeneration. Sci Rep 2014;4:6716.
    Crossref | PubMed
  98. Mills RJ, Parker BL, Quaife-Ryan GA, et al. Drug screening in human PSC-cardiac organoids identifies pro-proliferative compounds acting via the mevalonate pathway. Cell Stem Cell 2019;24:895–907.e6.
    Crossref | PubMed